1
|
Teichman EM, Hu J, Lin HY, Fisher-Foye RL, Blando A, Hu X, Kaniskan HÜ, Montgomery SE, Cai M, Parise LF, Wang J, Russo SJ, Han MH, Jin J, Morel C. Design and validation of novel brain-penetrant HCN channel inhibitors to ameliorate social stress-induced susceptible phenotype. Mol Psychiatry 2025:10.1038/s41380-025-02972-8. [PMID: 40199995 DOI: 10.1038/s41380-025-02972-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 12/16/2024] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Major Depressive Disorder (MDD) is a devastating, multifactorial disease with limited pharmacological treatment options. Patients with MDD exhibit alterations in their dopamine (DA) signaling pathways through the midbrain ventral tegmental area (VTA). A similar observation is also detected in preclinical models of stress - mice exhibit behavioral and physiological impairments following chronic social defeat stress (CSDS). Prior studies demonstrate that CSDS-susceptible mice have increased VTA DA neuronal excitability, in part driven by an upregulation in hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels. Inhibiting HCN channels with known inhibitors such as Cilobradine alleviates the negative behavioral effects of CSDS. Here, we aimed to identify Cilobradine analogs with improved neural tropism and inhibitory efficacy. Two compounds, MS7710 and MS7712, differing by their left-hand side moieties, have a similar, potent inhibitory effect on VTA DA Ih currents as compared to Cilobradine, and a greater inhibitory effect than Cilobradine on VTA DA firing rate. We demonstrate that MS7710 and MS7712 have superior brain/plasma concentration ratios as compared to Cilobradine. They were efficacious at inhibiting VTA DA neuron firing rate and bursting activity in CSDS-susceptible male mice at lower doses than Cilobradine, which was recapitulated in female CSDS-susceptible mice with MS7710. Finally, we define that a single intraperitoneal injection of MS7710 ameliorates CSDS-induced social interaction deficits and reward-associated cognitive inflexibility for at least two weeks in male and female mice. These findings yield a novel HCN channel inhibitor with improved neural tropism and stress-alleviating effects that could provide a basis for future antidepressant drug discovery.
Collapse
Affiliation(s)
- Emily M Teichman
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jianping Hu
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Hsiao-Yun Lin
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rachel L Fisher-Foye
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anthony Blando
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Xiaoping Hu
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - H Ümit Kaniskan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sarah E Montgomery
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Modendo Inc., 3415 Colorado Ave, Boulder, Colorado, 80303, USA
| | - Min Cai
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lyonna F Parise
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jun Wang
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, New York, NY, USA
| | - Scott J Russo
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Brain-Body Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ming-Hu Han
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology; Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.
| | - Jian Jin
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Carole Morel
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
2
|
Morris LS, Beltrán JM, Murrough JW, Morel C. Cross-species dissection of the modular role of the ventral tegmental area in depressive disorders. Neuroscience 2025; 569:248-266. [PMID: 39914519 PMCID: PMC11885014 DOI: 10.1016/j.neuroscience.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/17/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Depressive disorders, including major depressive disorder (MDD), represent one of the most prevalent set of disorders worldwide. MDD is characterized by a range of cognitive, behavioral, and neurobiological changes that contribute to the vast array of symptom profiles that make this disorder particularly difficult to treat. A multitude of established evidence suggests a role for the dopamine system, stemming in part from the ventral tegmental area (VTA), in mediating symptoms and behavioral changes that underlie depression. Developments in cutting-edge technologies in pre-clinical models of depressive phenotypes, such as retrograde tracing, electrophysiological recordings, immunohistochemistry, and molecular profiling, have allowed a deeper characterization of singular VTA neuron molecular, physiological, and projection properties. These developments have highlighted that the VTA is not a homogenous cell population but instead comprises vast cellular diversity that underscores its modular role across various functions related to reward processing, aversion, salience processing, learning and motivation. In this review, we begin by introducing the various cell types and brain regions that comprise the VTA circuitry. Then, we introduce the role of the VTA in reward processing as it compares to aversion processing. Next, we characterize distinct neural pathways within the VTA circuitry to understand the effects of chronic social and non-social stress and tie together how these neurobiological changes manifest into specific behavioral phenotypes. Finally, we relate these preclinical findings to clinical findings to parse the heterogeneity of depressive phenotypes and explain the efficacy of recent novel pharmacological interventions that may target the VTA in MDD.
Collapse
Affiliation(s)
- L S Morris
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai New York NY United States; Nuffield Department of Clinical Neurosciences, University of Oxford, UK; Department of Experimental Psychology, University of Oxford, UK.
| | - J M Beltrán
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai New York NY United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai New York NY United States
| | - J W Murrough
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai New York NY United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai New York NY United States; VISN 2 Mental Illness Research, Education, and Clinical Center (MIRECC), James J. Peters VA Medical Center Bronx NY United States
| | - C Morel
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai New York NY United States.
| |
Collapse
|
3
|
Yabe T, Mitsui Y, Ohnishi M, Tanigawa R, Tanizaki M, Sugiyama R, Kiriyama N, Otsuka A, Munekazu K. Social-defeat stress exposure during pregnancy induces abnormalities in spontaneous activity, sociality, and resilience to stress in offspring of mice. Behav Brain Res 2025; 480:115367. [PMID: 39631504 DOI: 10.1016/j.bbr.2024.115367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/25/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Environmental stress during prenatal periods can lead to neurodevelopmental disorders. Psychosocial stress can be studied using the social-defeat stress (SDS) animal model. However, the effects of prenatal exposure to SDS on the behavior of mature offspring mice have not been clarified. The present study assessed the spontaneous activity and social interaction of pups born to mothers exposed to SDS during gestation, as well as their post-maturity responses to environmental stimuli, focusing on changes in anxiety-like behavior following restraint stress exposure. METHODS Pregnant C57BL/6 J mice were subjected to SDS for 4 days, from E12.5-E15.5, using aggressive male ICR mice. We assessed the mature offspring (after 10 weeks of age) born to these mothers for spontaneous activity, anxiety-like behavior, and social interactions, and evaluated their activity levels post-maturity following restraint stress exposure. RESULTS The open field test (OF) indicated reduced travel distance and duration in the SDS group versus controls, whereas home-cage monitoring showed increased area traveled. In a novel environment, the SDS group showed a decrease in interest in stranger mice. In a multiple-animal rearing environment, the SDS group showed an increase in the frequency and number of contact with other individuals. Movement duration in the OF following restraint stress reduced significantly from 30 min to 4 h in the control versus SDS group. CONCLUSIONS Prenatal exposure to SDS can result in behavior resembling developmental disorders, impacting spontaneous activity and social interactions. Altered responses to stress suggest potential brain function abnormalities in offspring after maturation due to maternal SDS exposure.
Collapse
Affiliation(s)
- Tamaki Yabe
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Yuko Mitsui
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Momoka Ohnishi
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Rena Tanigawa
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Mizuki Tanizaki
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Rei Sugiyama
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Niina Kiriyama
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Airi Otsuka
- Laboratory of Nutrition and Health Science, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Komada Munekazu
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan.
| |
Collapse
|
4
|
Tofani GSS, Clarke G, Cryan JF. I "Gut" Rhythm: the microbiota as a modulator of the stress response and circadian rhythms. FEBS J 2025; 292:1454-1479. [PMID: 39841560 PMCID: PMC11927059 DOI: 10.1111/febs.17400] [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: 08/02/2024] [Revised: 11/20/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025]
Abstract
Modern habits are becoming more and more disruptive to health. As our days are often filled with circadian disruption and stress exposures, we need to understand how our responses to these external stimuli are shaped and how their mediators can be targeted to promote health. A growing body of research demonstrates the role of the gut microbiota in influencing brain function and behavior. The stress response and circadian rhythms, which are essential to maintaining appropriate responses to the environment, are known to be impacted by the gut microbiota. Gut microbes have been shown to alter the host's response to stress and modulate circadian rhythmicity. Although studies demonstrated strong links between the gut microbiota, circadian rhythms and the stress response, such studies were conducted in an independent manner not conducive to understanding the interface between these factors. Due to the interconnected nature of the stress response and circadian rhythms, in this review we explore how the gut microbiota may play a role in regulating the integration of stress and circadian signals in mammals and the consequences for brain health and disease.
Collapse
Affiliation(s)
- Gabriel S. S. Tofani
- APC MicrobiomeUniversity College CorkIreland
- Department of Anatomy & NeuroscienceUniversity College CorkIreland
| | - Gerard Clarke
- APC MicrobiomeUniversity College CorkIreland
- Department of Psychiatry & Neurobehavioural ScienceUniversity College CorkIreland
| | - John F. Cryan
- APC MicrobiomeUniversity College CorkIreland
- Department of Anatomy & NeuroscienceUniversity College CorkIreland
| |
Collapse
|
5
|
Morel C, Parise LF, Van der Zee YY, Issler O, Cai M, Browne CJ, Blando A, LeClair KB, Aubry AV, Haynes S, Williams RW, Mulligan MK, Russo SJ, Nestler EJ, Han MH. Male and female behavioral variability and morphine response in C57BL/6J, DBA/2J, and their BXD progeny following chronic stress exposure. Sci Rep 2024; 14:30785. [PMID: 39730457 PMCID: PMC11680947 DOI: 10.1038/s41598-024-80767-7] [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: 02/23/2024] [Accepted: 11/21/2024] [Indexed: 12/29/2024] Open
Abstract
Drug addiction is a multifactorial syndrome in which genetic predispositions and exposure to environmental stressors constitute major risk factors for the early onset, escalation, and relapse of addictive behaviors. While it is well known that stress plays a key role in drug addiction, the genetic factors that make certain individuals particularly sensitive to stress and, thereby, more vulnerable to becoming addicted are unknown. In an effort to test a complex set of gene x environment interactions-specifically gene x chronic stress-here we leveraged a systems genetics resource: BXD recombinant inbred mice (BXD5, BXD8, BXD14, BXD22, BXD29, and BXD32) and their parental mouse lines, C57BL/6J and DBA/2J. Utilizing the chronic social defeat stress (CSDS) and chronic variable stress (CVS) paradigms, we first showed sexual dimorphism in social and exploratory behaviors between the mouse strains. Further, we observed an interaction between genetic background and vulnerability to prolonged exposure to non-social stressors. Finally, we found that DBA/2J and C57BL/6J mice pre-exposed to stress displayed differences in morphine sensitivity. Our results support the hypothesis that genetic variation influences chronic stress-induced behavioral outcomes such as social and approach-avoidance behaviors, reward responses, as well as morphine sensitivity, and is likely to modulate the development of drug addiction.
Collapse
Affiliation(s)
- Carole Morel
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, and Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lyonna F Parise
- Friedman Brain Institute, and Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yentl Y Van der Zee
- Friedman Brain Institute, and Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Orna Issler
- Friedman Brain Institute, and Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Min Cai
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Caleb J Browne
- Friedman Brain Institute, and Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anthony Blando
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine B LeClair
- Friedman Brain Institute, and Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Antonio V Aubry
- Friedman Brain Institute, and Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sherod Haynes
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Megan K Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Scott J Russo
- Friedman Brain Institute, and Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Eric J Nestler
- Friedman Brain Institute, and Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Ming-Hu Han
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, and Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology (SIAT), and Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology (SUAT), Chinese Academy of Sciences, Shenzhen, Guangdong, People's Republic of China.
| |
Collapse
|
6
|
Fayad SL, Reynolds LM, Torquet N, Tolu S, Mondoloni S, Nguyen C, Siriphanh A, Justo R, Didienne S, Debray N, Viollet C, Raynaud L, Layadi Y, Fouquet C, Hannesse B, Capaz AM, Topilko T, Renier N, Mourot A, Marti F, Faure P. Individualistic reward-seeking strategies that predict response to nicotine emerge among isogenic male mice living in a micro-society. PLoS Biol 2024; 22:e3002850. [PMID: 39446878 PMCID: PMC11501037 DOI: 10.1371/journal.pbio.3002850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/18/2024] [Indexed: 10/26/2024] Open
Abstract
Individual animals differ in their traits and preferences, which shape their social interactions, survival, and susceptibility to disease, including addiction. Nicotine use is highly heterogenous and has been linked to the expression of personality traits. Although these relationships are well documented, we have limited understanding of the neurophysiological mechanisms that give rise to distinct behavioral profiles and their connection to nicotine susceptibility. To address this question, we conducted a study using a semi-natural and social environment called "Souris-City" to observe the long-term behavior of individual male mice. Souris-City provided both a communal living area and a separate test area where mice engaged in a reward-seeking task isolated from their peers. Mice developed individualistic reward-seeking strategies when choosing between water and sucrose in the test compartment, which, in turn, predicted how they adapted to the introduction of nicotine as a reinforcer. Moreover, the profiles mice developed while isolated in the test area correlated with their behavior within the social environment, linking decision-making strategies to the expression of behavioral traits. Neurophysiological markers of adaptability within the dopamine system were apparent upon nicotine challenge and were associated with specific profiles. Our findings suggest that environmental adaptations influence behavioral traits and sensitivity to nicotine by acting on dopaminergic reactivity in the face of nicotine exposure, potentially contributing to addiction susceptibility. These results further emphasize the importance of understanding interindividual variability in behavior to gain insight into the mechanisms of decision-making and addiction.
Collapse
Affiliation(s)
- Sophie L. Fayad
- Sorbonne University, INSERM, CNRS UMR8246, Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
- ESPCI Paris, PSL Research University, Brain Plasticity laboratory, CNRS UMR8249, Paris, France
| | - Lauren M. Reynolds
- Sorbonne University, INSERM, CNRS UMR8246, Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
- ESPCI Paris, PSL Research University, Brain Plasticity laboratory, CNRS UMR8249, Paris, France
| | - Nicolas Torquet
- Sorbonne University, INSERM, CNRS UMR8246, Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
| | - Stefania Tolu
- Sorbonne University, INSERM, CNRS UMR8246, Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
| | - Sarah Mondoloni
- Sorbonne University, INSERM, CNRS UMR8246, Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
| | - Claire Nguyen
- Sorbonne University, INSERM, CNRS UMR8246, Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
| | - Amy Siriphanh
- Sorbonne University, INSERM, CNRS UMR8246, Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
| | - Robin Justo
- Sorbonne University, INSERM, CNRS UMR8246, Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
- ESPCI Paris, PSL Research University, Brain Plasticity laboratory, CNRS UMR8249, Paris, France
| | - Steve Didienne
- Sorbonne University, INSERM, CNRS UMR8246, Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
- ESPCI Paris, PSL Research University, Brain Plasticity laboratory, CNRS UMR8249, Paris, France
| | - Nicolas Debray
- ESPCI Paris, PSL Research University, Brain Plasticity laboratory, CNRS UMR8249, Paris, France
| | - Cécile Viollet
- ESPCI Paris, PSL Research University, Brain Plasticity laboratory, CNRS UMR8249, Paris, France
| | - Louis Raynaud
- ESPCI Paris, PSL Research University, Brain Plasticity laboratory, CNRS UMR8249, Paris, France
| | - Yasmine Layadi
- ESPCI Paris, PSL Research University, Brain Plasticity laboratory, CNRS UMR8249, Paris, France
| | - Coralie Fouquet
- Sorbonne University, INSERM, CNRS UMR8246, Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
| | - Bernadette Hannesse
- Sorbonne University, INSERM, CNRS UMR8246, Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
| | - Ana-Marta Capaz
- Sorbonne University, ICM Institut du Cerveau et de la Moelle Epinière, Laboratoire de Plasticité Structurale INSERM U1127, CNRS UMR7225, Paris, France
| | - Thomas Topilko
- Sorbonne University, ICM Institut du Cerveau et de la Moelle Epinière, Laboratoire de Plasticité Structurale INSERM U1127, CNRS UMR7225, Paris, France
| | - Nicolas Renier
- Sorbonne University, ICM Institut du Cerveau et de la Moelle Epinière, Laboratoire de Plasticité Structurale INSERM U1127, CNRS UMR7225, Paris, France
| | - Alexandre Mourot
- Sorbonne University, INSERM, CNRS UMR8246, Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
- ESPCI Paris, PSL Research University, Brain Plasticity laboratory, CNRS UMR8249, Paris, France
| | - Fabio Marti
- Sorbonne University, INSERM, CNRS UMR8246, Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
- ESPCI Paris, PSL Research University, Brain Plasticity laboratory, CNRS UMR8249, Paris, France
| | - Philippe Faure
- Sorbonne University, INSERM, CNRS UMR8246, Neuroscience Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), Paris, France
- ESPCI Paris, PSL Research University, Brain Plasticity laboratory, CNRS UMR8249, Paris, France
| |
Collapse
|
7
|
Sales ISL, de Souza AG, Chaves Filho AJM, Sampaio TL, da Silva DMA, Valentim JT, Chaves RDC, Soares MVR, Costa Júnior DC, Barbosa Filho JM, Macêdo DS, de Sousa FCF. Antidepressant-like effect of riparin I and riparin II against CUMS-induced neuroinflammation via astrocytes and microglia modulation in mice. Behav Pharmacol 2024; 35:314-326. [PMID: 39094014 DOI: 10.1097/fbp.0000000000000788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Depression is a common mood disorder and many patients do not respond to conventional pharmacotherapy or experience a variety of adverse effects. This work proposed that riparin I (RIP I) and riparin II (RIP II) present neuroprotective effects through modulation of astrocytes and microglia, resulting in the reversal of depressive-like behaviors. To verify our hypothesis and clarify the pathways underlying the effect of RIP I and RIP II on neuroinflammation, we used the chronic unpredictable mild stress (CUMS) depression model in mice. Male Swiss mice were exposed to stressors for 28 days. From 15 th to the 22 nd day, the animals received RIP I or RIP II (50 mg/kg) or fluoxetine (FLU, 10 mg/kg) or vehicle, by gavage. On the 29 th day, behavioral tests were performed. Expressions of microglia (ionized calcium-binding adaptor molecule-1 - Iba-1) and astrocyte (glial fibrillary acidic protein - GFAP) markers and levels of cytokines tumor necrosis factor alfa (TNF-α) and interleukin 1 beta (IL-1β) were measured in the hippocampus. CUMS induced depressive-like behaviors and cognitive impairment, high TNF-α and IL-1β levels, decreased GFAP, and increased Iba-1 expressions. RIP I and RIP II reversed these alterations. These results contribute to the understanding the mechanisms underlying the antidepressant effect of RIP I and RIP II, which may be related to neuroinflammatory suppression.
Collapse
Affiliation(s)
- Iardja S L Sales
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza
| | - Alana G de Souza
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza
- Brazilian Hospital Services Company (EBSERH) - University Hospital, Federal University of Goias, Goiania
| | - Adriano J M Chaves Filho
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza
| | - Tiago L Sampaio
- Department of Clinical and Toxicological Analysis, Federal University of Ceara, Fortaleza, Ceara
| | - Daniel M A da Silva
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza
| | - José T Valentim
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza
| | - Raquell de C Chaves
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza
| | - Michelle V R Soares
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza
| | - Dilailson C Costa Júnior
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza
| | - José M Barbosa Filho
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutical Science, Federal University of Paraiba, João Pessoa, Brazil
| | - Danielle S Macêdo
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza
| | - Francisca Cléa Florenço de Sousa
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza
| |
Collapse
|
8
|
Smith A, Rodrigues T, Wallace C, Mezher K, MacAulay B, Prowse R, Hyland L, Abizaid A. Growth Hormone Secretagogue Receptor (GHSR) Signaling in the Ventral Tegmental Area (VTA) Mediates Feeding Produced by Chronic Social Defeat Stress in Male Mice. Neuroscience 2024; 547:17-27. [PMID: 38583506 DOI: 10.1016/j.neuroscience.2024.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/04/2024] [Accepted: 03/18/2024] [Indexed: 04/09/2024]
Abstract
Ghrelin, a hormone secreted by the stomach, binds to the growth hormone secretagogue receptor (GHSR) in various brain regions to produce a number of behavioral effects that include increased feeding motivation. During social defeat stress, ghrelin levels rise in correlation with increased feeding and potentially play a role in attenuating the anxiogenic effects of social defeat. One region implicated in the feeding effects of ghrelin is the ventral tegmental area (VTA), a region implicated in reward seeking behaviors, and linked to social defeat in mice. Here we examined the role of GHSR signaling in the VTA in feeding behavior in mice exposed to social defeat stress. Male C57BL/J6 mice that were socially defeated once daily for 3 weeks ate more, had higher plasma ghrelin level and increased GHSR expression in the VTA compared to non-stressed mice. Socially defeated GHSR KO mice failed to increase their caloric intake in response to this stressor but rescue of GHSR expression in the VTA restored feeding responses. Finally, we pharmacologically blocked VTA GHSR signalling with JMV2959 infused via an indwelling VTA cannula connected to a minipump. Vehicle-treated mice increased their caloric intake during social defeat, but JMV2959-infusions attenuated feeding responses and increased anxiety-like behaviors. The data suggest that GHSR signalling in the VTA is critical for the increases in appetite observed during chronic social defeat stress. Furthermore, these data support the idea that GHSR signaling in the VTA may also have anxiolytic effects, and blocking GHSR in this region may result in an anxiety-like phenotype.
Collapse
Affiliation(s)
- Andrea Smith
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Trevor Rodrigues
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Caroline Wallace
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Karen Mezher
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Brenna MacAulay
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Rebecca Prowse
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Lindsay Hyland
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Alfonso Abizaid
- Carleton University, Neuroscience Department, Ottawa, ON, Canada.
| |
Collapse
|
9
|
Cuttoli RDD, Sweis BM. Ketamine reverses stress-induced hypersensitivity to sunk costs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.12.593597. [PMID: 38798536 PMCID: PMC11118454 DOI: 10.1101/2024.05.12.593597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
How mood interacts with information processing in the brain is thought to mediate the maladaptive behaviors observed in depressed individuals. However, the neural mechanisms underlying impairments in emotion-cognition interactions are poorly understood. This includes influencing the balance between how past-sensitive vs. future-looking one is during decision-making. Recent insights from the field of neuroeconomics offer novel approaches to study changes in such valuation processes in a manner that is biologically tractable and readily translatable across species. We recently discovered that rodents are sensitive to "sunk costs" - a feature of higher cognition previously thought to be unique to humans. The sunk costs bias describes the phenomenon in which an individual overvalues and escalates commitment to continuing an ongoing endeavor, even if suboptimal, as a function of irrecoverable past (sunk) losses - information that, according to classic economic theory, should be ignored. In the present study, mice were exposed to chronic social defeat stress paradigm, a well-established animal model used for the study of depression. Mice were then tested on our longitudinal neuroeconomic foraging task, Restaurant Row. We found mice exposed to this severe stressor displayed an increased sensitivity to sunk costs, without altering overall willingness to wait. Mice were then randomly assigned to receive a single intraperitoneal injection of either saline or ketamine (20 mg/kg). We discovered that stress-induced hypersensitivity to sunk costs was renormalized following a single dose of ketamine. Interestingly, in non-defeated mice, ketamine treatment completely abolished sunk cost sensitivity, causing mice to no longer value irrecoverable losses during re-evaluation decisions who instead based choices solely on the future investment required to obtain a goal. These findings suggest that the antidepressant effects of ketamine may be mediated in part through changes in the processing of past-sensitive information during on-going decision-making, reducing its weight as a potential source of cognitive dissonance that could modulate behavior and instead promoting more future-thinking behavior.
Collapse
|
10
|
Kukucka T, Ferencova N, Visnovcova Z, Ondrejka I, Hrtanek I, Kovacova V, Macejova A, Mlyncekova Z, Tonhajzerova I. Mechanisms Involved in the Link between Depression, Antidepressant Treatment, and Associated Weight Change. Int J Mol Sci 2024; 25:4511. [PMID: 38674096 PMCID: PMC11050075 DOI: 10.3390/ijms25084511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Major depressive disorder is a severe mood disorder associated with a marked decrease in quality of life and social functioning, accompanied by a risk of suicidal behavior. Therefore, seeking out and adhering to effective treatment is of great personal and society-wide importance. Weight changes associated with antidepressant therapy are often cited as the reason for treatment withdrawal and thus are an important topic of interest. There indeed exists a significant mechanistic overlap between depression, antidepressant treatment, and the regulation of appetite and body weight. The suggested pathomechanisms include the abnormal functioning of the homeostatic (mostly humoral) and hedonic (mostly dopaminergic) circuits of appetite regulation, as well as causing neuromorphological and neurophysiological changes underlying the development of depressive disorder. However, this issue is still extensively discussed. This review aims to summarize mechanisms linked to depression and antidepressant therapy in the context of weight change.
Collapse
Affiliation(s)
- Tomas Kukucka
- Clinic of Psychiatry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, 03659 Martin, Slovakia; (T.K.); (I.O.); (I.H.); (V.K.); (A.M.); (Z.M.)
| | - Nikola Ferencova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (N.F.); (Z.V.)
| | - Zuzana Visnovcova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (N.F.); (Z.V.)
| | - Igor Ondrejka
- Clinic of Psychiatry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, 03659 Martin, Slovakia; (T.K.); (I.O.); (I.H.); (V.K.); (A.M.); (Z.M.)
| | - Igor Hrtanek
- Clinic of Psychiatry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, 03659 Martin, Slovakia; (T.K.); (I.O.); (I.H.); (V.K.); (A.M.); (Z.M.)
| | - Veronika Kovacova
- Clinic of Psychiatry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, 03659 Martin, Slovakia; (T.K.); (I.O.); (I.H.); (V.K.); (A.M.); (Z.M.)
| | - Andrea Macejova
- Clinic of Psychiatry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, 03659 Martin, Slovakia; (T.K.); (I.O.); (I.H.); (V.K.); (A.M.); (Z.M.)
| | - Zuzana Mlyncekova
- Clinic of Psychiatry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, 03659 Martin, Slovakia; (T.K.); (I.O.); (I.H.); (V.K.); (A.M.); (Z.M.)
| | - Ingrid Tonhajzerova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| |
Collapse
|
11
|
López-Otín C, Kroemer G. The missing hallmark of health: psychosocial adaptation. Cell Stress 2024; 8:21-50. [PMID: 38476764 PMCID: PMC10928495 DOI: 10.15698/cst2024.03.294] [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: 01/11/2024] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
The eight biological hallmarks of health that we initially postulated (Cell. 2021 Jan 7;184(1):33-63) include features of spatial compartmentalization (integrity of barriers, containment of local perturbations), maintenance of homeostasis over time (recycling & turnover, integration of circuitries, rhythmic oscillations) and an array of adequate responses to stress (homeostatic resilience, hormetic regulation, repair & regeneration). These hallmarks affect all eight somatic strata of the human body (molecules, organelles, cells, supracellular units, organs, organ systems, systemic circuitries and meta-organism). Here we postulate that mental and socioeconomic factors must be added to this 8×8 matrix as an additional hallmark of health ("psychosocial adaptation") and as an additional stratum ("psychosocial interactions"), hence building a 9×9 matrix. Potentially, perturbation of each of the somatic hallmarks and strata affects psychosocial factors and vice versa. Finally, we discuss the (patho)physiological bases of these interactions and their implications for mental health improvement.
Collapse
Affiliation(s)
- Carlos López-Otín
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Facultad de Ciencias de la Vida y la Naturaleza, Universidad Nebrija, Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
12
|
Morel C, Parise LF, Van der Zee Y, Issler O, Cai M, Browne C, Blando A, Leclair K, Haynes S, Williams RW, Mulligan MK, Russo SJ, Nestler EJ, Han MH. Male and female variability in response to chronic stress and morphine in C57BL/6J, DBA/2J, and their BXD progeny. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.23.581795. [PMID: 38464110 PMCID: PMC10925176 DOI: 10.1101/2024.02.23.581795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Drug addiction is a multifactorial syndrome in which genetic predispositions and exposure to environmental stressors constitute major risk factors for the early onset, escalation, and relapse of addictive behaviors. While it is well known that stress plays a key role in drug addiction, the genetic factors that make certain individuals particularly sensitive to stress and thereby more vulnerable to becoming addicted are unknown. In an effort to test a complex set of gene x environment interactions-specifically gene x chronic stress -here we leveraged a systems genetics resource: BXD recombinant inbred mice (BXD5, BXD8, BXD14, BXD22, BXD29, and BXD32) and their parental mouse lines, C57BL/6J and DBA/2J. Utilizing the chronic social defeat stress (CSDS) and chronic variable stress (CVS) paradigms, we first showed sexual dimorphism in the behavioral stress response between the mouse strains. Further, we observed an interaction between genetic background and vulnerability to prolonged exposure to non-social stressors. Finally, we found that DBA/2J and C57BL/6J mice pre-exposed to stress displayed differences in morphine sensitivity. Our results support the hypothesis that genetic variation in predisposition to stress responses influences morphine sensitivity and is likely to modulate the development of drug addiction.
Collapse
|
13
|
de Kloet ER, Joëls M. The cortisol switch between vulnerability and resilience. Mol Psychiatry 2024; 29:20-34. [PMID: 36599967 DOI: 10.1038/s41380-022-01934-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023]
Abstract
In concert with neuropeptides and transmitters, the end products of the hypothalamus-pituitary-adrenal (HPA) axis, the glucocorticoid hormones cortisol and corticosterone (CORT), promote resilience: i.e., the ability to cope with threats, adversity, and trauma. To exert this protective action, CORT activates mineralocorticoid receptors (MR) and glucocorticoid receptors (GR) that operate in a complementary manner -as an on/off switch- to coordinate circadian events, stress-coping, and adaptation. The evolutionary older limbic MR facilitates contextual memory retrieval and supports an on-switch in the selection of stress-coping styles at a low cost. The rise in circulating CORT concentration after stress subsequently activates a GR-mediated off-switch underlying recovery of homeostasis by providing the energy for restraining the primary stress reactions and promoting cognitive control over emotional reactivity. GR activation facilitates contextual memory storage of the experience to enable future stress-coping. Such complementary MR-GR-mediated actions involve rapid non-genomic and slower gene-mediated mechanisms; they are time-dependent, conditional, and sexually dimorphic, and depend on genetic background and prior experience. If coping fails, GR activation impairs cognitive control and promotes emotional arousal which eventually may compromise resilience. Such breakdown of resilience involves a transition to a chronic stress construct, where information processing is crashed; it leads to an imbalanced MR-GR switch and hence increased vulnerability. Novel MR-GR modulators are becoming available that may reset a dysregulated stress response system to reinstate the cognitive flexibility required for resilience.
Collapse
Affiliation(s)
- E Ronald de Kloet
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden University, Leiden, The Netherlands.
- Leiden/Amsterdam Center of Drug Research, Leiden University, Leiden, The Netherlands.
| | - Marian Joëls
- Dept. Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
14
|
Tringali G, Lavanco G, Castelli V, Pizzolanti G, Kuchar M, Currò D, Cannizzaro C, Brancato A. Cannabidiol tempers alcohol intake and neuroendocrine and behavioural correlates in alcohol binge drinking adolescent rats. Focus on calcitonin gene-related peptide's brain levels. Phytother Res 2023; 37:4870-4884. [PMID: 37525534 DOI: 10.1002/ptr.7972] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/13/2023] [Accepted: 07/15/2023] [Indexed: 08/02/2023]
Abstract
Alcohol binge drinking is common among adolescents and may challenge the signalling systems that process affective stimuli, including calcitonin gene-related peptide (CGRP) signalling. Here, we employed a rat model of adolescent binge drinking to evaluate reward-, social- and aversion-related behaviour, glucocorticoid output and CGRP levels in affect-related brain regions. As a potential rescue, the effect of the phytocannabinoid cannabidiol was explored. Adolescent male rats underwent the intermittent 20% alcohol two-bottle choice paradigm; at the binge day (BD) and the 24 h withdrawal day (WD), we assessed CGRP expression in medial prefrontal cortex (mPFC), nucleus accumbens (NAc), amygdala, hypothalamus and brainstem; in addition, we evaluated sucrose preference, social motivation and drive, nociceptive response, and serum corticosterone levels. Cannabidiol (40 mg/kg, i.p.) was administered before each drinking session, and its effect was measured on the above-mentioned readouts. At BD and WD, rats displayed decreased CGRP expression in mPFC, NAc and amygdala; increased CGRP levels in the brainstem; increased response to rewarding- and nociceptive stimuli and decreased social drive; reduced serum corticosterone levels. Cannabidiol reduced alcohol consumption and preference; normalised the abnormal corticolimbic CGRP expression, and the reward and aversion-related hyper-responsivity, as well as glucocorticoid levels in alcohol binge-like drinking rats. Overall, CGRP can represent both a mediator and a target of alcohol binge-like drinking and provides a further piece in the intricate puzzle of alcohol-induced behavioural and neuroendocrine sequelae. CBD shows promising effects in limiting adolescent alcohol binge drinking and rebalancing the bio-behavioural abnormalities.
Collapse
Affiliation(s)
- Giuseppe Tringali
- Pharmacology Section, Department of Health Care Surveillance and Bioethics, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome, Italy
| | - Gianluca Lavanco
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties of Excellence "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Valentina Castelli
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giuseppe Pizzolanti
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties of Excellence "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Martin Kuchar
- Forensic Laboratory of Biologically Active Compounds, Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Prague, Czechia
- Psychedelics Research Centre, National Institute of Mental Health, Prague, Czechia
| | - Diego Currò
- Pharmacology Section, Department of Health Care Surveillance and Bioethics, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome, Italy
| | - Carla Cannizzaro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Anna Brancato
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties of Excellence "G. D'Alessandro", University of Palermo, Palermo, Italy
| |
Collapse
|
15
|
Battivelli D, Vernochet C, Conabady E, Nguyen C, Zayed A, Lebel A, Meirsman AC, Messaoudene S, Fieggen A, Dreux G, Rigoni D, Le Borgne T, Marti F, Contesse T, Barik J, Tassin JP, Faure P, Parnaudeau S, Tronche F. Dopamine Neuron Activity and Stress Signaling as Links Between Social Hierarchy and Psychopathology Vulnerability. Biol Psychiatry 2023:S0006-3223(23)01600-1. [PMID: 37804900 DOI: 10.1016/j.biopsych.2023.08.029] [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: 09/13/2022] [Revised: 07/28/2023] [Accepted: 08/19/2023] [Indexed: 10/09/2023]
Abstract
BACKGROUND Social status in humans, generally reflected by socioeconomic status, has been associated, when constrained, with heightened vulnerability to pathologies including psychiatric diseases. Social hierarchy in mice translates into individual and interdependent behavioral strategies of animals within a group. The rules leading to the emergence of a social organization are elusive, and detangling the contribution of social status from other factors, whether environmental or genetic, to normal and pathological behaviors remains challenging. METHODS We investigated the mechanisms shaping the emergence of a social hierarchy in isogenic C57BL/6 mice raised in groups of 4 using conditional mutant mouse models and chemogenetic manipulation of dopamine midbrain neuronal activity. We further studied the evolution of behavioral traits and the vulnerability to psychopathological-like phenotypes according to the social status of the animals. RESULTS Higher sociability predetermined higher social hierarchy in the colony. Upon hierarchy establishment, higher-ranked mice showed increased anxiety and better cognitive abilities in a working memory task. Strikingly, the higher-ranked mice displayed a reduced activity of dopaminergic neurons within the ventral tegmental area, paired with a decreased behavioral response to cocaine and a decreased vulnerability to depressive-like behaviors following repeated social defeats. The pharmacogenetic inhibition of this neuronal population and the genetic inactivation of glucocorticoid receptor signaling in dopamine-sensing brain areas that resulted in decreased dopaminergic activity promoted accession to higher social ranks. CONCLUSIONS Dopamine activity and its modulation by the stress response shapes social organization in mice, potentially linking interindividual and social status differences in vulnerability to psychopathologies.
Collapse
Affiliation(s)
- Dorian Battivelli
- Gene Regulation and Adaptive Behaviors group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Institut de Biologie Paris Seine, Paris, France; Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France
| | - Cécile Vernochet
- Gene Regulation and Adaptive Behaviors group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Institut de Biologie Paris Seine, Paris, France; Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France
| | - Estelle Conabady
- Gene Regulation and Adaptive Behaviors group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Institut de Biologie Paris Seine, Paris, France; Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France
| | - Claire Nguyen
- Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France; Neurophysiology and Behavior group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Paris, France
| | - Abdallah Zayed
- Gene Regulation and Adaptive Behaviors group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Institut de Biologie Paris Seine, Paris, France; Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France
| | - Ashley Lebel
- Gene Regulation and Adaptive Behaviors group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Institut de Biologie Paris Seine, Paris, France; Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France
| | - Aura Carole Meirsman
- Gene Regulation and Adaptive Behaviors group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Institut de Biologie Paris Seine, Paris, France; Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France
| | - Sarah Messaoudene
- Gene Regulation and Adaptive Behaviors group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Institut de Biologie Paris Seine, Paris, France; Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France
| | - Alexandre Fieggen
- Gene Regulation and Adaptive Behaviors group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Institut de Biologie Paris Seine, Paris, France; Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France
| | - Gautier Dreux
- Gene Regulation and Adaptive Behaviors group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Institut de Biologie Paris Seine, Paris, France; Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France
| | - Daiana Rigoni
- Université Côte d'Azur, Nice, France; Institut de Pharmacologie Moléculaire and Cellulaire, Centre National de la Recherche Scientifique Unité Mixte de Recherche 7275, Valbonne, France
| | - Tinaïg Le Borgne
- Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France; Neurophysiology and Behavior group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Paris, France
| | - Fabio Marti
- Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France; Neurophysiology and Behavior group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Paris, France
| | - Thomas Contesse
- Université Côte d'Azur, Nice, France; Institut de Pharmacologie Moléculaire and Cellulaire, Centre National de la Recherche Scientifique Unité Mixte de Recherche 7275, Valbonne, France
| | - Jacques Barik
- Université Côte d'Azur, Nice, France; Institut de Pharmacologie Moléculaire and Cellulaire, Centre National de la Recherche Scientifique Unité Mixte de Recherche 7275, Valbonne, France
| | - Jean-Pol Tassin
- Gene Regulation and Adaptive Behaviors group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Institut de Biologie Paris Seine, Paris, France; Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France
| | - Philippe Faure
- Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France; Neurophysiology and Behavior group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Paris, France
| | - Sébastien Parnaudeau
- Gene Regulation and Adaptive Behaviors group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Institut de Biologie Paris Seine, Paris, France; Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France.
| | - François Tronche
- Gene Regulation and Adaptive Behaviors group, Department of Neuroscience Paris Seine, Unité Mixte de Recherche 8246, Centre National de la Recherche Scientifique, Institut de Biologie Paris Seine, Paris, France; Institut National de la Santé et de la Recherche Médicale U1130, Paris, France; Sorbonne Université, Paris, France.
| |
Collapse
|
16
|
Jeanneteau F. Stress and the risk of Alzheimer dementia: Can deconstructed engrams be rebuilt? J Neuroendocrinol 2023; 35:e13235. [PMID: 36775895 DOI: 10.1111/jne.13235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023]
Abstract
The exact neuropathological mechanism by which the dementia process unfolds is under intense scrutiny. The disease affects about 38 million people worldwide, 70% of which are clinically diagnosed with Alzheimer's disease (AD). If the destruction of synapses essential for learning, planning and decision-making is part of the problem, must the restoration of previously lost synapses be part of the solution? It is plausible that neuronal capacity to restitute information corresponds with the adaptive capacity of its connectivity reserve. A challenge will be to promote the functional connectivity that can compensate for the lost one. This will require better clarification of the remodeling of functional connectivity during the progression of AD dementia and its reversal upon experimental treatment. A major difficulty is to promote the neural pathways that are atrophied in AD dementia while suppressing others that are bolstered. Therapeutic strategies should aim at scaling functional connectivity to a just balance between the atrophic and hypertrophic systems. However, the exact factors that can help reach this objective are still unclear. Similarities between the effects of chronic stress and some neuropathological mechanisms underlying AD dementia support the idea that common components deserve prime attention as therapeutic targets.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de génomique fonctionnelle, Université de Montpellier, INSERM, CNRS, Montpellier, France
| |
Collapse
|
17
|
Zhao F, Behnisch T. The Enigmatic CA2: Exploring the Understudied Region of the Hippocampus and Its Involvement in Parkinson's Disease. Biomedicines 2023; 11:1996. [PMID: 37509636 PMCID: PMC10377725 DOI: 10.3390/biomedicines11071996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease that affects both motor and non-motor functions. Although motor impairment is a prominent clinical sign of PD, additional neurological symptoms may also occur, particularly in the preclinical and prodromal stages. Among these symptoms, social cognitive impairment is common and detrimental. This article aims to review non-motor symptoms in PD patients, focusing on social cognitive deficits. It also examines the specific characteristics of the CA2 region and its involvement in social behavior, highlighting recent advances and perspectives. Additionally, this review provides critical insights into and analysis of research conducted in rodents and humans, which may help improve the understanding of the current status of putative therapeutic strategies for social cognitive dysfunction in PD and potential avenues related to the function of the hippocampal CA2 region.
Collapse
Affiliation(s)
- Fang Zhao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Thomas Behnisch
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
18
|
Alhaddad A, Radwan A, Mohamed NA, Mehanna ET, Mostafa YM, El-Sayed NM, Fattah SA. Rosiglitazone Mitigates Dexamethasone-Induced Depression in Mice via Modulating Brain Glucose Metabolism and AMPK/mTOR Signaling Pathway. Biomedicines 2023; 11:biomedicines11030860. [PMID: 36979839 PMCID: PMC10046017 DOI: 10.3390/biomedicines11030860] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Major depressive disorder (MDD) is a common, complex disease with poorly understood pathogenesis. Disruption of glucose metabolism is implicated in the pathogenesis of depression. AMP-activated protein kinase (AMPK) has been shown to regulate the activity of several kinases, including pAKT, p38MAPK, and mTOR, which are important signaling pathways in the treatment of depression. This study tested the hypothesis that rosiglitazone (RGZ) has an antidepressant impact on dexamethasone (DEXA)-induced depression by analyzing the function of the pAKT/p38MAPK/mTOR pathway and NGF through regulation of AMPK. MDD-like pathology was induced by subcutaneous administration of DEXA (20 mg/kg) for 21 days in all groups except in the normal control group, which received saline. To investigate the possible mechanism of RGZ, the protein expression of pAMPK, pAKT, p38MAPK, and 4EBP1 as well as the levels of hexokinase, pyruvate kinase, and NGF were assessed in prefrontal cortex and hippocampal samples. The activities of pAMPK and NGF increased after treatment with RGZ. The administration of RGZ also decreased the activity of mTOR as well as downregulating the downstream signaling pathways pAKT, p38MAPK, and 4EBP1. Here, we show that RGZ exerts a potent inhibitory effect on the pAKT/p38MAPK/mTOR/4EBP1 pathway and causes activation of NGF in brain cells. This study has provided sufficient evidence of the potential for RGZ to ameliorate DEXA-induced depression. A new insight has been introduced into the critical role of NGF activation in brain cells in depression. These results suggest that RGZ is a promising antidepressant for the treatment of MDD.
Collapse
Affiliation(s)
- Aisha Alhaddad
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al-Madinah Al-Munawwarah 30078, Saudi Arabia
| | - Asmaa Radwan
- Department of Pharmacology &Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Noha A. Mohamed
- Department of Forgery & Counterfeiting, Forensic Medicine, Ministry of Justice, Ismailia 41522, Egypt
| | - Eman T. Mehanna
- Department of Biochemistry & Molecular Biology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
- Correspondence: (E.T.M.); (N.M.E.-S.)
| | - Yasser M. Mostafa
- Department of Pharmacology &Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University in Cairo, Badr 11829, Egypt
| | - Norhan M. El-Sayed
- Department of Pharmacology &Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
- Correspondence: (E.T.M.); (N.M.E.-S.)
| | - Shaimaa A. Fattah
- Department of Biochemistry & Molecular Biology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
19
|
Lei H, Shu H, Xiong R, He T, Lv J, Liu J, Pi G, Ke D, Wang Q, Yang X, Wang JZ, Yang Y. Poststress social isolation exerts anxiolytic effects by activating the ventral dentate gyrus. Neurobiol Stress 2023; 24:100537. [PMID: 37081927 PMCID: PMC10112178 DOI: 10.1016/j.ynstr.2023.100537] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/21/2023] [Accepted: 03/25/2023] [Indexed: 03/30/2023] Open
Abstract
After aversive stress, people either choose to return to their previously familiar social environment or tend to adopt temporary social withdrawal to buffer negative emotions. However, which behavior intervention is more appropriate and when remain elusive. Here, we unexpectedly found that stressed mice experiencing social isolation exhibited less anxiety than those experiencing social contact. Within the first 24 h after returning to their previous social environment, mice experienced acute restraint stress (ARS) displayed low social interest but simultaneously received excessive social disturbance from their cage mates, indicating a critical time window for social isolation to balance the conflict. To screen brain regions that were differentially activated between the poststress social isolation and poststress social contact groups, we performed ΔFosB immunostaining and found that ΔFosB + signals were remarkably increased in the vDG of poststress social isolation group compared with poststress social contact group. There were no significant differences between the two groups in the other anxiety- and social-related brain regions, such as prelimbic cortex, infralimbic cortex, nucleus accumbens, etc. These data indicate that vDG is closely related to the differential phenotypes between the poststress social isolation and poststress social contact groups. Electrophysiological recording, further, revealed a higher activity of vDG in the poststress social isolation group than the poststress social contact group. Chemogenetically inhibiting vDG excitatory neurons within the first 24 h after ARS completely abolished the anxiolytic effects of poststress social isolation, while stimulating vDG excitatory neurons remarkably reduced anxiety-like behaviors in the poststress social contact group. Together, these data suggest that the activity of vDG excitatory neurons is essential and sufficient to govern the anxiolytic effect of poststress social isolation. To the best of our knowledge, this is the first report to uncover a beneficial role of temporal social isolation in acute stress-induced anxiety. In addition to the critical 24-h time window, activation of vDG is crucial for ameliorating anxiety through poststress social isolation.
Collapse
Affiliation(s)
- Huiyang Lei
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huaqing Shu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Corresponding author.
| | - Rui Xiong
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ting He
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingru Lv
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiale Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guilin Pi
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, 8 Longyuan Road, Nanshan District, Shenzhen, 518055, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China
- Corresponding author. Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ying Yang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Corresponding author.
| |
Collapse
|
20
|
Glucocorticoid-Responsive Tissue Plasminogen Activator (tPA) and Its Inhibitor Plasminogen Activator Inhibitor-1 (PAI-1): Relevance in Stress-Related Psychiatric Disorders. Int J Mol Sci 2023; 24:ijms24054496. [PMID: 36901924 PMCID: PMC10003592 DOI: 10.3390/ijms24054496] [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: 01/30/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Stressful events trigger a set of complex biological responses which follow a bell-shaped pattern. Low-stress conditions have been shown to elicit beneficial effects, notably on synaptic plasticity together with an increase in cognitive processes. In contrast, overly intense stress can have deleterious behavioral effects leading to several stress-related pathologies such as anxiety, depression, substance use, obsessive-compulsive and stressor- and trauma-related disorders (e.g., post-traumatic stress disorder or PTSD in the case of traumatic events). Over a number of years, we have demonstrated that in response to stress, glucocorticoid hormones (GCs) in the hippocampus mediate a molecular shift in the balance between the expression of the tissue plasminogen activator (tPA) and its own inhibitor plasminogen activator inhibitor-1 (PAI-1) proteins. Interestingly, a shift in favor of PAI-1 was responsible for PTSD-like memory induction. In this review, after describing the biological system involving GCs, we highlight the key role of tPA/PAI-1 imbalance observed in preclinical and clinical studies associated with the emergence of stress-related pathological conditions. Thus, tPA/PAI-1 protein levels could be predictive biomarkers of the subsequent onset of stress-related disorders, and pharmacological modulation of their activity could be a potential new therapeutic approach for these debilitating conditions.
Collapse
|
21
|
CHIBA S, NUMAKAWA T, MURATA T, KAWAMINAMI M, HIMI T. Enhanced social reward response and anxiety-like behavior with downregulation of nucleus accumbens glucocorticoid receptor in BALB/c mice. J Vet Med Sci 2023; 85:30-39. [PMID: 36403974 PMCID: PMC9887208 DOI: 10.1292/jvms.22-0103] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 11/06/2022] [Indexed: 11/19/2022] Open
Abstract
Social anhedonia is a psychological state with difficulty in experiencing pleasure from social interactions and is observed in various diseases, such as depressive disorders. Although the relationships between social reward responses and anxiety- and depression-like behaviors have remained unclear, a social reward conditioned place preference (SCPP) test can be used to analyze the rewarding nature of social interactions. To elucidate these relationships, we used 5-week-old male mice of AKR, BALB/c, and C57BL/6J strains and conducted behavioral tests in the following order: elevated plus-maze test (EPM), open field test (OFT), SCPP, saccharin preference test (SPT), and passive avoidance test. The nucleus accumbens of these mice were collected 24 hr after these behavioral tests and were used for western blotting to determine the levels of receptors for brain-derived neurotrophic factors and glucocorticoids. BALB/c mice displayed the highest levels of anxiety-like behavior in EPM and OFT as well as physical anhedonia-like behaviors in SPT. They also showed increased responses to social rewards and huddling behaviors in SCPP, with downregulated glucocorticoid receptor (GR). Regression analysis results revealed positive influences of anxiety- and physical anhedonia-like behaviors and expressions of GR on social reward responses. Collectively, temperament associated with anxiety and physical anhedonia may affect social reward responses, which possibly is influenced by the expression of GR that can modify these psychological traits.
Collapse
Affiliation(s)
- Shuichi CHIBA
- Faculty of Veterinary Medicine, Okayama University of Science, Ehime, Japan
| | - Tadahiro NUMAKAWA
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takuya MURATA
- Faculty of Veterinary Medicine, Okayama University of Science, Ehime, Japan
| | | | - Toshiyuki HIMI
- Faculty of Pharmacy and Research Institute of Pharmaceutical Science, Musashino University, Tokyo, Japan
| |
Collapse
|
22
|
Teal LB, Ingram SM, Bubser M, McClure E, Jones CK. The Evolving Role of Animal Models in the Discovery and Development of Novel Treatments for Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2023; 30:37-99. [PMID: 36928846 DOI: 10.1007/978-3-031-21054-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Historically, animal models have been routinely used in the characterization of novel chemical entities (NCEs) for various psychiatric disorders. Animal models have been essential in the in vivo validation of novel drug targets, establishment of lead compound pharmacokinetic to pharmacodynamic relationships, optimization of lead compounds through preclinical candidate selection, and development of translational measures of target occupancy and functional target engagement. Yet, with decades of multiple NCE failures in Phase II and III efficacy trials for different psychiatric disorders, the utility and value of animal models in the drug discovery process have come under intense scrutiny along with the widespread withdrawal of the pharmaceutical industry from psychiatric drug discovery. More recently, the development and utilization of animal models for the discovery of psychiatric NCEs has undergone a dynamic evolution with the application of the Research Domain Criteria (RDoC) framework for better design of preclinical to clinical translational studies combined with innovative genetic, neural circuitry-based, and automated testing technologies. In this chapter, the authors will discuss this evolving role of animal models for improving the different stages of the discovery and development in the identification of next generation treatments for psychiatric disorders.
Collapse
Affiliation(s)
- Laura B Teal
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Shalonda M Ingram
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Michael Bubser
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Elliott McClure
- College of Pharmacy and Health Sciences, Lipscomb University, Nashville, TN, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
23
|
Jiang A, Zhou C, Samsom J, Yan S, Yu DZ, Jia ZP, Wong AHC, Liu F. The GR-FKBP51 interaction modulates fear memory but not spatial or recognition memory. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110604. [PMID: 35839967 DOI: 10.1016/j.pnpbp.2022.110604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 10/17/2022]
Abstract
The glucocorticoid receptor (GR) forms a protein complex with FKBP51 that is increased in post-traumatic stress disorder (PTSD) and by fear conditioned learning. Disrupting the GR-FKBP51 complex with a synthetic peptide can block the storage or retrieval of fear conditioned memories, which could be a novel approach to the alleviate fear associated memory in PTSD. However, a potential unacceptable side effect could be the impairment of other types of memory. Thus, we investigated the effect of disrupting the GR-FKBP51 complex on recognition memory using the novel object and displaced object recognition tasks, spatial memory in the Morris water maze, and on social interaction in Crawley's three-chamber social interaction test. We did not observe adverse effects on these other types of memory and conclude that the GR-FKBP51 interaction remains a promising target for treating psychiatric disorders characterized by unwanted aversive memories such as in PTSD.
Collapse
Affiliation(s)
- Anlong Jiang
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Chanjuan Zhou
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - James Samsom
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Shuxin Yan
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Dian Zheng Yu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Zheng-Ping Jia
- Neurosciences & Mental Health, The Hospital for Sick Children, 555 University Ave., M5G 1X8, Toronto, Ontario, Canada
| | - Albert H C Wong
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; Department of Pharmacology, University of Toronto, Toronto, ON M5S 1A8, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
24
|
Glucocorticoid Receptor-Dependent Astrocytes Mediate Stress Vulnerability. Biol Psychiatry 2022; 92:204-215. [PMID: 35151464 DOI: 10.1016/j.biopsych.2021.11.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/04/2021] [Accepted: 11/28/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Major depressive disorder is a devastating psychiatric illness that affects approximately 17% of the population worldwide. Astrocyte dysfunction has been implicated in its pathophysiology. Traumatic experiences and stress contribute to the onset of major depressive disorder, but how astrocytes respond to stress is poorly understood. METHODS Using Western blotting analysis, we identified that stress vulnerability was associated with reduced astrocytic glucocorticoid receptor (GR) expression in mouse models of depression. We further investigated the functions of astrocytic GRs in regulating depression and the underlying mechanisms by using a combination of behavioral studies, fiber photometry, biochemical experiments, and RNA sequencing methods. RESULTS GRs in astrocytes were more sensitive to stress than those in neurons. GR absence in astrocytes induced depressive-like behaviors, whereas restoring astrocytic GR expression in the medial prefrontal cortex prevented the depressive-like phenotype. Furthermore, we found that GRs in the medial prefrontal cortex affected astrocytic Ca2+ activity and dynamic ATP (adenosine 5'-triphosphate) release in response to stress. RNA sequencing of astrocytes isolated from GR deletion mice identified the PI3K-Akt (phosphoinositide 3-kinase-Akt) signaling pathway, which was required for astrocytic GR-mediated ATP release. CONCLUSIONS These findings reveal that astrocytic GRs play an important role in stress response and that reduced astrocytic GR expression in the stressed subject decreases ATP release to mediate stress vulnerability.
Collapse
|
25
|
Lamontagne SJ, Wash SIJ, Irwin SH, Zucconi KE, Olmstead MC. Effects of dopamine modulation on chronic stress-induced deficits in reward learning. COGNITIVE, AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2022; 22:736-753. [PMID: 35396630 DOI: 10.3758/s13415-022-01001-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/21/2022] [Indexed: 06/14/2023]
Abstract
Anhedonia is characteristically preceded by chronic stress, likely involving downstream effects of glucocorticoid alterations on dopamine (DA) function. To elucidate the neural underpinnings of this interaction, we examined whether acute pharmacological modulation of DA alters reward learning after chronic mild stress (CMS). Forty-eight male Wistar rats were exposed to a 21-day CMS regime (n = 48 no stress controls) before completing the probabilistic reward task (PRT), a well-validated cross-species test of reward learning. We first examined whether stress-induced reward dysfunction could be restored by systemic injections of low-dose amisulpride (AMI), which increases DA transmission via D2-like autoreceptor blockade. Then, we investigated region-specific effects through bilateral infusions of quinpirole (QUIN), a D2-like receptor agonist, into either the nucleus accumbens core (NAcc) or medial prefrontal cortex (mPFC). Blunted reward learning in CMS animals was reversed by acute AMI administration, but this treatment did not alter reward learning in the no stress group. Elevated adrenal gland weight, a proxy for stress reactivity, predicted lower reward learning in the untreated CMS group. This effect was extinguished following AMI treatment. These findings might be attributed to significantly higher D2 receptor density in the NAcc of high stress reactive animals. To this end, NAcc QUIN infusions potentiated reward learning relative to mPFC QUIN infusions in CMS rats, but there was no effect in no stress control rats. Collectively, these findings suggest that DA modulation reverses stress-induced reward dysfunction, even among the most stress-reactive animals. The effect might depend on D2-like receptor activation in the mesolimbic system.
Collapse
Affiliation(s)
- Steven J Lamontagne
- Department of Psychology, Queen's University, 62 Arch Street, Kingston, ON, K7L 3N6, USA.
| | - Sarah I J Wash
- Department of Psychology, Queen's University, 62 Arch Street, Kingston, ON, K7L 3N6, USA
| | - Samantha H Irwin
- Department of Psychology, Queen's University, 62 Arch Street, Kingston, ON, K7L 3N6, USA
| | - Kate E Zucconi
- Department of Psychology, Queen's University, 62 Arch Street, Kingston, ON, K7L 3N6, USA
| | - Mary C Olmstead
- Department of Psychology, Queen's University, 62 Arch Street, Kingston, ON, K7L 3N6, USA
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| |
Collapse
|
26
|
Nicotinic receptors promote susceptibility to social stress in female mice linked with neuroadaptations within VTA dopamine neurons. Neuropsychopharmacology 2022; 47:1587-1596. [PMID: 35459925 PMCID: PMC9283477 DOI: 10.1038/s41386-022-01314-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/14/2022]
Abstract
There are about twice as many women as men who experience depression during their lifetime. Although life circumstances and especially exposure to stressful situations constitute a major risk factor to develop depression, the underlying mechanisms have yet to be unraveled. We employed the chronic social defeat procedure to elicit depressive-like symptoms in females and ketamine to validate the model. We performed ex-vivo patch clamp recordings to assess cellular adaptations and used pharmacological agents to dissect these deregulations. Chronic social defeat exposure triggers a hyperactivity of VTA putative dopamine (DA) neurons in females susceptible to stress but not resilient ones. This hyperactivity was fully reversed by a single administration of ketamine. In virally-identified brain circuits of both susceptible and resilient females, we found a hypercholinergic tone to the VTA arising from the laterodorsal tegmentum. Application of puffs of nicotine revealed a decreased sensitivity of DA neurons in resilient mice when compared to naive or susceptible ones. The in vivo acute administration of the positive allosteric modulator for α7 nicotinic acetylcholine receptors (nAChRs) not only increased susceptibility to stress by enhancing activity of VTA DA neurons, but also triggered a switch in phenotype from resilient to susceptible. Our data unravel dysregulations of VTA DA neurons activity exclusively in females exhibiting depressive-like symptoms and identify VTA nAChRs as key molecular substrates that exacerbate susceptibility to stress.
Collapse
|
27
|
Joëls M, de Kloet ER. Sleeping off stress. Science 2022; 377:27-28. [PMID: 35771905 DOI: 10.1126/science.adc9782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Social defeat activates midbrain cells, promoting sleep and reducing anxiety in mice.
Collapse
Affiliation(s)
- Marian Joëls
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - E Ronald de Kloet
- Leiden Academic Center for Drug Research, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| |
Collapse
|
28
|
Zhao F, Wang K, Wen Y, Chen X, Liu H, Qi F, Fu Y, Zhu J, Guan S, Liu Z. Contribution of hippocampal BDNF/CREB signaling pathway and gut microbiota to emotional behavior impairment induced by chronic unpredictable mild stress during pregnancy in rats offspring. PeerJ 2022; 10:e13605. [PMID: 35769142 PMCID: PMC9235812 DOI: 10.7717/peerj.13605] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/26/2022] [Indexed: 01/17/2023] Open
Abstract
Background Numerous studies have shown that exposure to prenatal maternal stress (PMS) is associated with various psychopathological outcomes of offspring. The accumulating evidence linking bacteria in the gut and neurons in the brain (the microbiota-gut-brain axis) has been aconsensus; however, there is a lack of research on the involvement mechanism of gut microbiota in the regulation of the BDNF/CREB signaling pathway in the hippocampus of prenatally stressed offspring. Methods Pregnant rats were subjected to chronic unpredictable mild stress (CUMS) to establish the prenatal maternal stress model. The body weight was measured and the behavioral changes were recorded. Offspring were tested to determine emotional state using sucrose preference test (SPT), open-field test (OFT) and suspended tail test (STT). Gut microbiota was evaluated by sequencing the microbial 16S rRNA V3-V4 region, and the interactive analysis of bacterial community structure and diversity was carried out. The expression of hippocampal BDNF, TrkB and CREB mRNA and proteins were respectively measured using RT-PCR and Western blotting. Results Prenatal maternal stress increased maternal plasma corticosterone levels, slowed maternal weight gain and caused depression-like behaviors (all P < 0.05). In offspring, prenatal maternal stress increased plasma corticosterone levels (P < 0.05) and emotional behavior changes (depression-like state) were observed (P < 0.05). The species abundance, diversity and composition of the offspring's gut microbiota changed after the maternal stress during pregnancy (P < 0.05). Compared with the control group's offspring, the species abundance of Lactobacillaceae was dropped, while the abundance of the Muribaculaceae species abundance was risen. Concurrent, changes in the hippocampal structure of the offspring and decreases in expression of BDNF/CREB signaling were noted (P < 0.05). Conclusions Prenatal maternal stress leads to high corticosterone status and abnormal emotion behavior of offspring, which may be associated with the abnormal BDNF/CREB signaling in hippocampus of offspring caused by the change of gut microbiota composition.
Collapse
Affiliation(s)
- Feng Zhao
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, Ningxia, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Public Health, Chongqing Medical University, Chongqing, China, Chongqing Medical University, Chongqing, China
| | - Kai Wang
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yujun Wen
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xiaohui Chen
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Hongya Liu
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Faqiu Qi
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Youjuan Fu
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jiashu Zhu
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Suzhen Guan
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, Ningxia, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Zhihong Liu
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
29
|
SWI/SNF chromatin remodeler complex within the reward pathway is required for behavioral adaptations to stress. Nat Commun 2022; 13:1807. [PMID: 35379786 PMCID: PMC8980038 DOI: 10.1038/s41467-022-29380-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 02/22/2022] [Indexed: 01/01/2023] Open
Abstract
Enduring behavioral changes upon stress exposure involve changes in gene expression sustained by epigenetic modifications in brain circuits, including the mesocorticolimbic pathway. Brahma (BRM) and Brahma Related Gene 1 (BRG1) are ATPase subunits of the SWI/SNF complexes involved in chromatin remodeling, a process essential to enduring plastic changes in gene expression. Here, we show that in mice, social defeat induces changes in BRG1 nuclear distribution. The inactivation of the Brg1/Smarca4 gene within dopamine-innervated regions or the constitutive inactivation of the Brm/Smarca2 gene leads to resilience to repeated social defeat and decreases the behavioral responses to cocaine without impacting midbrain dopamine neurons activity. Within striatal medium spiny neurons, Brg1 gene inactivation reduces the expression of stress- and cocaine-induced immediate early genes, increases levels of heterochromatin and at a global scale decreases chromatin accessibility. Altogether these data demonstrate the pivotal function of SWI/SNF complexes in behavioral and transcriptional adaptations to salient environmental challenges. Repeated exposure to social stressors in rodents results in behavioural changes. Here the authors show that behavioural adaptations to stress are associated with nuclear organization changes through SWI/SNF chromatin remodeler in specific neuronal populations of the mesolimbic system.
Collapse
|
30
|
Morel C, Montgomery SE, Li L, Durand-de Cuttoli R, Teichman EM, Juarez B, Tzavaras N, Ku SM, Flanigan ME, Cai M, Walsh JJ, Russo SJ, Nestler EJ, Calipari ES, Friedman AK, Han MH. Midbrain projection to the basolateral amygdala encodes anxiety-like but not depression-like behaviors. Nat Commun 2022; 13:1532. [PMID: 35318315 PMCID: PMC8940900 DOI: 10.1038/s41467-022-29155-1] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
Anxiety disorders are complex diseases, and often co-occur with depression. It is as yet unclear if a common neural circuit controls anxiety-related behaviors in both anxiety-alone and comorbid conditions. Here, utilizing the chronic social defeat stress (CSDS) paradigm that induces singular or combined anxiety- and depressive-like phenotypes in mice, we show that a ventral tegmental area (VTA) dopamine circuit projecting to the basolateral amygdala (BLA) selectively controls anxiety- but not depression-like behaviors. Using circuit-dissecting ex vivo electrophysiology and in vivo fiber photometry approaches, we establish that expression of anxiety-like, but not depressive-like, phenotypes are negatively correlated with VTA → BLA dopamine neuron activity. Further, our optogenetic studies demonstrate a causal link between such neuronal activity and anxiety-like behaviors. Overall, these data establish a functional role for VTA → BLA dopamine neurons in bi-directionally controlling anxiety-related behaviors not only in anxiety-alone, but also in anxiety-depressive comorbid conditions in mice.
Collapse
Affiliation(s)
- Carole Morel
- grid.59734.3c0000 0001 0670 2351Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Sarah E. Montgomery
- grid.59734.3c0000 0001 0670 2351Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Long Li
- grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Romain Durand-de Cuttoli
- grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Emily M. Teichman
- grid.59734.3c0000 0001 0670 2351Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Barbara Juarez
- grid.59734.3c0000 0001 0670 2351Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.412623.00000 0000 8535 6057Department of Psychiatry and Behavioral Sciences, University of Washington Medical Center, Seattle, WA USA ,grid.412623.00000 0000 8535 6057Department of Pharmacology, University of Washington Medical Center, Seattle, WA USA
| | - Nikos Tzavaras
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Microscopy Core, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Stacy M. Ku
- grid.59734.3c0000 0001 0670 2351Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Meghan E. Flanigan
- grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.10698.360000000122483208Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC USA
| | - Min Cai
- grid.59734.3c0000 0001 0670 2351Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Jessica J. Walsh
- grid.59734.3c0000 0001 0670 2351Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.10698.360000000122483208Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ,grid.10698.360000000122483208Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Scott J. Russo
- grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Eric J. Nestler
- grid.59734.3c0000 0001 0670 2351Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Erin S. Calipari
- grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.152326.10000 0001 2264 7217Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN USA
| | - Allyson K. Friedman
- grid.59734.3c0000 0001 0670 2351Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.257167.00000 0001 2183 6649Department of Biological Science, Hunter College at the City University of New York, New York, NY USA
| | - Ming-Hu Han
- grid.59734.3c0000 0001 0670 2351Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Friedman Brain Institute, Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.458489.c0000 0001 0483 7922Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong China
| |
Collapse
|
31
|
Faure P, Fayad SL, Solié C, Reynolds LM. Social Determinants of Inter-Individual Variability and Vulnerability: The Role of Dopamine. Front Behav Neurosci 2022; 16:836343. [PMID: 35386723 PMCID: PMC8979673 DOI: 10.3389/fnbeh.2022.836343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Individuals differ in their traits and preferences, which shape their interactions, their prospects for survival and their susceptibility to diseases. These correlations are well documented, yet the neurophysiological mechanisms underlying the emergence of distinct personalities and their relation to vulnerability to diseases are poorly understood. Social ties, in particular, are thought to be major modulators of personality traits and psychiatric vulnerability, yet the majority of neuroscience studies are performed on rodents in socially impoverished conditions. Rodent micro-society paradigms are therefore key experimental paradigms to understand how social life generates diversity by shaping individual traits. Dopamine circuitry is implicated at the interface between social life experiences, the expression of essential traits, and the emergence of pathologies, thus proving a possible mechanism to link these three concepts at a neuromodulatory level. Evaluating inter-individual variability in automated social testing environments shows great promise for improving our understanding of the link between social life, personality, and precision psychiatry – as well as elucidating the underlying neurophysiological mechanisms.
Collapse
|
32
|
Feng Y, Fan H, Liang X, Wang X, Gao G, Gun S. Environmental enrichment changes rabbits' behavior, serum hormone level and further affects cecal microbiota. PeerJ 2022; 10:e13068. [PMID: 35287345 PMCID: PMC8917805 DOI: 10.7717/peerj.13068] [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: 12/15/2020] [Accepted: 02/15/2022] [Indexed: 01/12/2023] Open
Abstract
Many studies have shown that stress is associated with gut microbiota. Environmental enrichment (EE) could reduce stress in farm animals; however, limited information is available on the microbial community composition in rabbits raised with or without EE. This study aimed to identify EE influences on the behavior, serum hormonal levels, and cecal microbiota of rabbits. Two hundred Rex rabbits were segregated randomly within four cohorts (n = 50); reared for 76 d within standardized enclosures (non-enriched) or within cages containing a willow-stick (WS), rubber-duck (RD), or a can of beans (CB). The rabbits' ingestive, rest, locomotion, exploratory, grooming, and abnormal behavior were observed. The serum hormone levels for rabbits were measured, and cecal specimens were sequencedfrom the V3-V4 region using 16S rRNA amplicons. Environmental enrichment increased feeding and drinking time, promoted exploratory behavior, and reduced abnormal behavior in rabbits. Insulin-like growth factor 1(IGF-1) levels of the enriched cohorts were elevated in comparison to the control cohort. Serum cortisol level for CB cohort was markedly reduced in comparison to the control cohort (p < 0.05), while dopamine levels for CB cohort peaked. Further, we found that EE mainly affected the dominant microbiota. Several families, such as Erysipelotrichaceae, Tannerellaceae, Enterobacteriaceae, Burkholderiaceae, and Prevotellaceae were markedly reduced within the CB cohort. Bacteria such as Alloprevotella, Bifidobacterium, Enterobacteriaceae, Parabacteroides, and Erysipelatoclostridium were identified as having negative associations with the presence of serum cortisol. EE influenced rabbit behavior and serum hormonal levels, and CB enrichment was the most suitable for rabbits. Further, cecal microbiota composition and diversity were affected by CB enrichment. These findings suggested that CB could be considered for use in rabbit husbandry.
Collapse
Affiliation(s)
- Yang Feng
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Huimei Fan
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xue Liang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Xiaofeng Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Guoyan Gao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Shuangbao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| |
Collapse
|
33
|
Pomrenze MB, Paliarin F, Maiya R. Friend of the Devil: Negative Social Influences Driving Substance Use Disorders. Front Behav Neurosci 2022; 16:836996. [PMID: 35221948 PMCID: PMC8866771 DOI: 10.3389/fnbeh.2022.836996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/13/2022] [Indexed: 11/20/2022] Open
Abstract
Substance use disorders in humans have significant social influences, both positive and negative. While prosocial behaviors promote group cooperation and are naturally rewarding, distressing social encounters, such as aggression exhibited by a conspecific, are aversive and can enhance the sensitivity to rewarding substances, promote the acquisition of drug-taking, and reinstate drug-seeking. On the other hand, withdrawal and prolonged abstinence from drugs of abuse can promote social avoidance and suppress social motivation, accentuating drug cravings and facilitating relapse. Understanding how complex social states and experiences modulate drug-seeking behaviors as well as the underlying circuit dynamics, such as those interacting with mesolimbic reward systems, will greatly facilitate progress on understanding triggers of drug use, drug relapse and the chronicity of substance use disorders. Here we discuss some of the common circuit mechanisms underlying social and addictive behaviors that may underlie their antagonistic functions. We also highlight key neurochemicals involved in social influences over addiction that are frequently identified in comorbid psychiatric conditions. Finally, we integrate these data with recent findings on (±)3,4-methylenedioxymethamphetamine (MDMA) that suggest functional segregation and convergence of social and reward circuits that may be relevant to substance use disorder treatment through the competitive nature of these two types of reward. More studies focused on the relationship between social behavior and addictive behavior we hope will spur the development of treatment strategies aimed at breaking vicious addiction cycles.
Collapse
Affiliation(s)
- Matthew B. Pomrenze
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
- *Correspondence: Matthew B. Pomrenze Rajani Maiya
| | - Franciely Paliarin
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Rajani Maiya
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- *Correspondence: Matthew B. Pomrenze Rajani Maiya
| |
Collapse
|
34
|
Liang S, Wu Y, Hanxiaoran L, Greenshaw AJ, Li T. Anhedonia in Depression and Schizophrenia: Brain Reward and Aversion Circuits. Neuropsychiatr Dis Treat 2022; 18:1385-1396. [PMID: 35836582 PMCID: PMC9273831 DOI: 10.2147/ndt.s367839] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/28/2022] [Indexed: 11/23/2022] Open
Abstract
Anhedonia, which is defined as markedly diminished interest or pleasure, is a prominent symptom of psychiatric disorders, most notably major depressive disorder (MDD) and schizophrenia. Anhedonia is considered a transdiagnostic symptom that is associated with deficits in neural reward and aversion functions. Here, we review the characteristics of anhedonia in depression and schizophrenia as well as shared or disorder-specific anhedonia-related alterations in reward and aversion pathways of the brain. In particular, we highlight that anhedonia is characterized by impairments in anticipatory pleasure and integration of reward-related information in MDD, whereas anhedonia in schizophrenia is associated with neurocognitive deficits in representing the value of rewards. Dysregulation of the frontostriatal circuit and mesocortical and mesolimbic circuit systems may be the transdiagnostic neurobiological basis of reward and aversion impairments underlying anhedonia in these two disorders. Blunted aversion processing in depression and relatively strong aversion in schizophrenia are primarily attributed to the dysfunction of the habenula, insula, amygdala, and anterior cingulate cortex. Furthermore, patients with schizophrenia appear to exhibit greater abnormal activation and extended functional coupling than those with depression. From a transdiagnostic perspective, understanding the neural mechanisms underlying anhedonia in patients with psychiatric disorders may help in the development of more targeted and efficacious treatment and intervention strategies.
Collapse
Affiliation(s)
- Sugai Liang
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310013, People's Republic of China
| | - Yue Wu
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310013, People's Republic of China
| | - Li Hanxiaoran
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310013, People's Republic of China
| | - Andrew J Greenshaw
- Department of Psychiatry, University of Alberta, Edmonton, AB, T6G 2B7, Canada
| | - Tao Li
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310013, People's Republic of China
| |
Collapse
|
35
|
Allichon MC, Ortiz V, Pousinha P, Andrianarivelo A, Petitbon A, Heck N, Trifilieff P, Barik J, Vanhoutte P. Cell-Type-Specific Adaptions in Striatal Medium-Sized Spiny Neurons and Their Roles in Behavioral Responses to Drugs of Abuse. Front Synaptic Neurosci 2022; 13:799274. [PMID: 34970134 PMCID: PMC8712310 DOI: 10.3389/fnsyn.2021.799274] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022] Open
Abstract
Drug addiction is defined as a compulsive pattern of drug-seeking- and taking- behavior, with recurrent episodes of abstinence and relapse, and a loss of control despite negative consequences. Addictive drugs promote reinforcement by increasing dopamine in the mesocorticolimbic system, which alters excitatory glutamate transmission within the reward circuitry, thereby hijacking reward processing. Within the reward circuitry, the striatum is a key target structure of drugs of abuse since it is at the crossroad of converging glutamate inputs from limbic, thalamic and cortical regions, encoding components of drug-associated stimuli and environment, and dopamine that mediates reward prediction error and incentive values. These signals are integrated by medium-sized spiny neurons (MSN), which receive glutamate and dopamine axons converging onto their dendritic spines. MSN primarily form two mostly distinct populations based on the expression of either DA-D1 (D1R) or DA-D2 (D2R) receptors. While a classical view is that the two MSN populations act in parallel, playing antagonistic functional roles, the picture seems much more complex. Herein, we review recent studies, based on the use of cell-type-specific manipulations, demonstrating that dopamine differentially modulates dendritic spine density and synapse formation, as well as glutamate transmission, at specific inputs projecting onto D1R-MSN and D2R-MSN to shape persistent pathological behavioral in response to drugs of abuse. We also discuss the identification of distinct molecular events underlying the detrimental interplay between dopamine and glutamate signaling in D1R-MSN and D2R-MSN and highlight the relevance of such cell-type-specific molecular studies for the development of innovative strategies with potential therapeutic value for addiction. Because drug addiction is highly prevalent in patients with other psychiatric disorders when compared to the general population, we last discuss the hypothesis that shared cellular and molecular adaptations within common circuits could explain the co-occurrence of addiction and depression. We will therefore conclude this review by examining how the nucleus accumbens (NAc) could constitute a key interface between addiction and depression.
Collapse
Affiliation(s)
- Marie-Charlotte Allichon
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Vanesa Ortiz
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Paula Pousinha
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Andry Andrianarivelo
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Anna Petitbon
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Nicolas Heck
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Pierre Trifilieff
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Jacques Barik
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Peter Vanhoutte
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| |
Collapse
|
36
|
Chakraborty P, Chattarji S, Jeanneteau F. A salience hypothesis of stress in PTSD. Eur J Neurosci 2021; 54:8029-8051. [PMID: 34766390 DOI: 10.1111/ejn.15526] [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: 07/10/2021] [Revised: 09/13/2021] [Accepted: 10/30/2021] [Indexed: 11/30/2022]
Abstract
Attention to key features of contexts and things is a necessary tool for all organisms. Detecting these salient features of cues, or simply, salience, can also be affected by exposure to traumatic stress, as has been widely reported in individuals suffering from post-traumatic stress disorder (PTSD). Interestingly, similar observations have been robustly replicated across many animal models of stress as well. By using evidence from such rodent stress paradigms, in the present review, we explore PTSD through the lens of salience processing. In this context, we propose that interaction between the neurotrophin brain-derived neurotrophic factor (BDNF) and glucocorticoids determines the long lasting cellular and behavioural consequences of stress salience. We also describe the dual effect of glucocorticoid therapy in the amelioration of PTSD symptoms. Finally, by integrating in vivo observations at multiple scales of plasticity, we propose a unifying hypothesis that pivots on a crucial role of glucocorticoid signalling in dynamically orchestrating stress salience.
Collapse
Affiliation(s)
- Prabahan Chakraborty
- Institut de Genomique Fonctionnelle, University of Montpellier, Inserm, CNRS, Montpellier, 34090, France.,Tata Institute of Fundamental Research, National Centre for Biological Sciences, Bellary Road, Bangalore, 560065, India
| | - Sumantra Chattarji
- Tata Institute of Fundamental Research, National Centre for Biological Sciences, Bellary Road, Bangalore, 560065, India.,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.,Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Freddy Jeanneteau
- Institut de Genomique Fonctionnelle, University of Montpellier, Inserm, CNRS, Montpellier, 34090, France
| |
Collapse
|
37
|
Parker KN, Donovan MH, Smith K, Noble-Haeusslein LJ. Traumatic Injury to the Developing Brain: Emerging Relationship to Early Life Stress. Front Neurol 2021; 12:708800. [PMID: 34484104 PMCID: PMC8416304 DOI: 10.3389/fneur.2021.708800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/22/2021] [Indexed: 12/01/2022] Open
Abstract
Despite the high incidence of brain injuries in children, we have yet to fully understand the unique vulnerability of a young brain to an injury and key determinants of long-term recovery. Here we consider how early life stress may influence recovery after an early age brain injury. Studies of early life stress alone reveal persistent structural and functional impairments at adulthood. We consider the interacting pathologies imposed by early life stress and subsequent brain injuries during early brain development as well as at adulthood. This review outlines how early life stress primes the immune cells of the brain and periphery to elicit a heightened response to injury. While the focus of this review is on early age traumatic brain injuries, there is also a consideration of preclinical models of neonatal hypoxia and stroke, as each further speaks to the vulnerability of the brain and reinforces those characteristics that are common across each of these injuries. Lastly, we identify a common mechanistic trend; namely, early life stress worsens outcomes independent of its temporal proximity to a brain injury.
Collapse
Affiliation(s)
- Kaila N. Parker
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Michael H. Donovan
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Kylee Smith
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Linda J. Noble-Haeusslein
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
38
|
Lei Y, Cui Q, Yang G, Piao L, Inoue A, Wu H, Li X, Kuzuya M, Cheng XW. Statins Mitigate Stress-Related Vascular Aging and Atherosclerosis in apoE-Deficient Mice Fed High Fat-Diet: The Role of Glucagon-Like Peptide-1/Adiponectin Axis. Front Cell Dev Biol 2021; 9:687868. [PMID: 34368136 PMCID: PMC8335539 DOI: 10.3389/fcell.2021.687868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/04/2021] [Indexed: 01/30/2023] Open
Abstract
Objectives Exposure to chronic psychosocial stress is a risk factor for atherosclerotic cardiovascular diseases. Given that the 3-hydroxy-3-methylglutaryl-coenzyme reductase inhibitor statins prevent atherogenesis, we evaluated whether pitavastatin prevents chronic stress- and high fat diet-induced vascular senescence and atherogenesis in apolipoprotein E-deficient (ApoE–/–) mice, with a special focus on glucagon-like peptide-1 (GLP-1)/adiponectin (APN) axis. Methods and Results 6-week-old ApoE–/– mice loaded a high-fat diet were randomly assigned into non-stress (n = 12) and stress (n = 13) groups for 12 weeks. Non-stress control mice were left undisturbed. Chronic stress accelerated high fat diet-induce arterial senescence and atherosclerotic plaque growth. The chronic stress lowered the levels of circulating GLP-1 as well as adipose and plasma APN. As compared with the stress alone mice, the pitavastatin-treated mice had reduced macrophage infiltration, elastin fragments, and increased plaque collagen volume, and lowered levels of osteopontin, toll-like receptor-2/-4, macrophage chemoattractant protein-1, C-X-C chemokine receptor-4, p47phox, p47phox, gp91phox, cathepsins S, p16, and p21, mRNAs and/or proteins. Pitavastatin increased plasma GLP-1 and APN levels and suppressed matrix metalloproteinase-2/-9 gene expressions and activities in the aortas. Finally, the protective effect of pitavastatin was abrogated by APN blocking. Conclusion These findings suggested that the pitavastatin-mediated pleiotropic vasculoprotective effects are likely attributable, at least in part, to the elevation of GLP-1 and APN levels and the inhibition of diet-induced plaque inflammation, oxidative stress, and proteolysis in ApoE–/– mice received chronic stress conditions.
Collapse
Affiliation(s)
- Yanna Lei
- Department of Intensive Care Unit, Yanbian University Hospital, Yanjin, China
| | - Qingsong Cui
- Department of Intensive Care Unit, Yanbian University Hospital, Yanjin, China
| | - Guang Yang
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanjin, China
| | - Limei Piao
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanjin, China
| | - Aiko Inoue
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hongxian Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiang Li
- Department of Intensive Care Unit, Yanbian University Hospital, Yanjin, China
| | - Masafumi Kuzuya
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Xian Wu Cheng
- Department of Intensive Care Unit, Yanbian University Hospital, Yanjin, China.,Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
39
|
Contesse T, Broussot L, Fofo H, Vanhoutte P, Fernandez SP, Barik J. Dopamine and glutamate receptors control social stress-induced striatal ERK1/2 activation. Neuropharmacology 2021; 190:108534. [PMID: 33781778 DOI: 10.1016/j.neuropharm.2021.108534] [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: 12/07/2020] [Revised: 03/03/2021] [Accepted: 03/12/2021] [Indexed: 11/27/2022]
Abstract
Stress has been acknowledged as one of the main risk factors for the onset of psychiatric disorders. Social stress is the most common type of stressor encountered in our daily lives. Uncovering the molecular determinants of the effect of stress on the brain would help understanding the complex maladaptations that contribute to pathological stress-related mental states. We examined molecular changes in the reward system following social defeat stress in mice, as increasing evidence implicates this system in sensing stressful stimuli. Following acute or chronic social defeat stress, the activation (i.e. phosphorylation) of extracellular signal-regulated kinases ERK1 and ERK2 (pERK1/2), markers of synaptic plasticity, was monitored in sub-regions of the reward system. We employed pharmacological antagonists and inhibitory DREADD to dissect the sequence of events controlling pERK1/2 dynamics. The nucleus accumbens (NAc) showed marked increases in pERK1/2 following both acute and chronic social stress compared to the dorsal striatum. Increases in pERK1/2 required dopamine D1 receptors and GluN2B-containing NMDA receptors. Paraventricular thalamic glutamatergic inputs to the NAc are required for social stress-induced pERK1/2. The molecular adaptations identified here could contribute to the long-lasting impact of stress on the brain and may be targeted to counteract stress-related psychopathologies.
Collapse
Affiliation(s)
- Thomas Contesse
- Université Côte d'Azur, Nice, France; Institut de Pharmacologie Moléculaire & Cellulaire, CNRS UMR7275, Valbonne, France
| | - Loïc Broussot
- Université Côte d'Azur, Nice, France; Institut de Pharmacologie Moléculaire & Cellulaire, CNRS UMR7275, Valbonne, France
| | - Hugo Fofo
- Université Côte d'Azur, Nice, France; Institut de Pharmacologie Moléculaire & Cellulaire, CNRS UMR7275, Valbonne, France
| | - Peter Vanhoutte
- CNRS, UMR 8246, Neuroscience Paris Seine, F, 75005, Paris, France; INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, F, 75005, Paris, France; Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, F, 75005, Paris, France
| | - Sebastian P Fernandez
- Université Côte d'Azur, Nice, France; Institut de Pharmacologie Moléculaire & Cellulaire, CNRS UMR7275, Valbonne, France.
| | - Jacques Barik
- Université Côte d'Azur, Nice, France; Institut de Pharmacologie Moléculaire & Cellulaire, CNRS UMR7275, Valbonne, France.
| |
Collapse
|
40
|
HUZARD D, RAPPENEAU V, MEIJER OC, TOUMA C, ARANGO-LIEVANO M, GARABEDIAN MJ, JEANNETEAU F. Experience and activity-dependent control of glucocorticoid receptors during the stress response in large-scale brain networks. Stress 2021; 24:130-153. [PMID: 32755268 PMCID: PMC7907260 DOI: 10.1080/10253890.2020.1806226] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The diversity of actions of the glucocorticoid stress hormones among individuals and within organs, tissues and cells is shaped by age, gender, genetics, metabolism, and the quantity of exposure. However, such factors cannot explain the heterogeneity of responses in the brain within cells of the same lineage, or similar tissue environment, or in the same individual. Here, we argue that the stress response is continuously updated by synchronized neural activity on large-scale brain networks. This occurs at the molecular, cellular and behavioral levels by crosstalk communication between activity-dependent and glucocorticoid signaling pathways, which updates the diversity of responses based on prior experience. Such a Bayesian process determines adaptation to the demands of the body and external world. We propose a framework for understanding how the diversity of glucocorticoid actions throughout brain networks is essential for supporting optimal health, while its disruption may contribute to the pathophysiology of stress-related disorders, such as major depression, and resistance to therapeutic treatments.
Collapse
Affiliation(s)
- Damien HUZARD
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Virginie RAPPENEAU
- Department of Behavioural Biology, University of Osnabrück, Osnabrück, Germany
| | - Onno C. MEIJER
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Chadi TOUMA
- Department of Behavioural Biology, University of Osnabrück, Osnabrück, Germany
| | - Margarita ARANGO-LIEVANO
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
| | | | - Freddy JEANNETEAU
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
- Corresponding author:
| |
Collapse
|
41
|
Salberg S, Yamakawa GR, Griep Y, Bain J, Beveridge JK, Sun M, McDonald SJ, Shultz SR, Brady RD, Wright DK, Noel M, Mychasiuk R. Pain in the Developing Brain: Early Life Factors Alter Nociception and Neurobiological Function in Adolescent Rats. Cereb Cortex Commun 2021; 2:tgab014. [PMID: 34296160 PMCID: PMC8152853 DOI: 10.1093/texcom/tgab014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 01/09/2023] Open
Abstract
Although adverse early experiences prime individuals to be at increased risk for chronic pain, little research has examined the trauma–pain relationship in early life or the underlying mechanisms that drive pathology over time. Given that early experiences can potentiate the nociceptive response, this study aimed to examine the effects of a high-fat, high-sugar (HFHS) diet and early life stress (maternal separation [MS]) on pain outcomes in male and female adolescent rats. Half of the rats also underwent a plantar-incision surgery to investigate how the pain system responded to a mildly painful stimuli in adolescence. Compared with controls, animals that were on the HFHS diet, experienced MS, or had exposure to both, exhibited increased anxiety-like behavior and altered thermal and mechanical nociception at baseline and following the surgery. Advanced magnetic resonance imaging demonstrated that the HFHS diet and MS altered the maturation of the brain, leading to changes in brain volume and diffusivity within the anterior cingulate, amygdala, corpus callosum, nucleus accumbens, and thalamus, while also modifying the integrity of the corticospinal tracts. The effects of MS and HFHS diet were often cumulative, producing exacerbated pain sensitivity and increased neurobiological change. As early experiences are modifiable, understanding their role in pain may provide targets for early intervention/prevention.
Collapse
Affiliation(s)
- Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne 3004, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Monash University, Melbourne 3004, Australia
| | - Yannick Griep
- Behavioural Science Institute, Radboud University, Nijmegen 6525 GD, the Netherlands.,Division of Epidemiology, Stress Research Institute, Stockholm University, Stockholm 114 19, Sweden
| | - Jesse Bain
- Department of Neuroscience, Monash University, Melbourne 3004, Australia
| | - Jaimie K Beveridge
- Department of Psychology, University of Calgary, Calgary T2N 1N4, Canada
| | - Mujun Sun
- Department of Neuroscience, Monash University, Melbourne 3004, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne 3004, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne 3086, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne 3004, Australia
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne 3004, Australia
| | - David K Wright
- Department of Neuroscience, Monash University, Melbourne 3004, Australia
| | - Melanie Noel
- Department of Psychology, University of Calgary, Calgary T2N 1N4, Canada
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne 3004, Australia.,Department of Psychology, University of Calgary, Calgary T2N 1N4, Canada
| |
Collapse
|
42
|
Tiwari D, Gupta P. Nuclear Receptors in Asthma: Empowering Classical Molecules Against a Contemporary Ailment. Front Immunol 2021; 11:594433. [PMID: 33574813 PMCID: PMC7870687 DOI: 10.3389/fimmu.2020.594433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/09/2020] [Indexed: 02/06/2023] Open
Abstract
The escalation in living standards and adoption of 'Western lifestyle' has an allied effect on the increased allergy and asthma burden in both developed and developing countries. Current scientific reports bespeak an association between allergic diseases and metabolic dysfunction; hinting toward the critical requirement of organized lifestyle and dietary habits. The ubiquitous nuclear receptors (NRs) translate metabolic stimuli into gene regulatory signals, integrating diet inflences to overall developmental and physiological processes. As a consequence of such promising attributes, nuclear receptors have historically been at the cutting edge of pharmacy world. This review discusses the recent findings that feature the cardinal importance of nuclear receptors and how they can be instrumental in modulating current asthma pharmacology. Further, it highlights a possible future employment of therapy involving dietary supplements and synthetic ligands that would engage NRs and aid in eliminating both asthma and linked comorbidities. Therefore, uncovering new and evolving roles through analysis of genomic changes would represent a feasible approach in both prevention and alleviation of asthma.
Collapse
Affiliation(s)
| | - Pawan Gupta
- Department of Molecular Biology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
43
|
Zhou Q, Ding W, Qian Z, Jiang G, Sun C, Xu K. Chronic Unpredictable Mild Stress Accelerates the Growth of Bladder Cancer in a Xenograft Mouse Model. Psychol Res Behav Manag 2020; 13:1289-1297. [PMID: 33380846 PMCID: PMC7767701 DOI: 10.2147/prbm.s288983] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/10/2020] [Indexed: 01/17/2023] Open
Abstract
Objective Chronic psychological stress is common in patients with bladder cancer. An increasing number of evidence demonstrated that psychiatric disorder leads to worse prognostic outcomes in bladder cancer. This study was to investigate the effects of chronic psychological stress on the growth of bladder cancer and its potential mechanisms. Methods A xenograft mouse model was established by subcutaneously implanting the human bladder cancer cell line T24 into nude mice. All of the tumor-bearing mice (N=20) were randomly separated into two groups. Mice in the control group were subjected to normal feeding conditions, while in another group, a chronic unpredictable mild stress (CUMS) model was established, in which mice were exposed to various types of stressors. Various analyses were performed on parameters including the tumor volume, tumor weight, expression of Caspase-3 and VEGF, proportion of Ki-67 positive cells (Ki-67 index), microvessel density (MVD) and serum concentrations of epinephrine and cortisol. Results In the CUMS group, the growth of transplanted tumors was distinctly accelerated, with the weight of removed tumors at the end of experiment increased by 34.07% compared to that of the control. Serum levels of epinephrine and cortisol determined by ELISA were significantly increased by CUMS. Immunohistochemistry and Western blot analysis showed that the expression of Caspase-3 was downregulated, whereas the expression of VEGF was upregulated in the CUMS group. Meanwhile, CUMS could increase the Ki-67 index and MVD. Conclusion Our research supports the hypothesis that CUMS could affect the growth of bladder cancer in nude mice, indicating that the intervention of chronic psychological stress may be a possible therapeutic strategy for bladder cancer.
Collapse
Affiliation(s)
- Qidong Zhou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Weihong Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Zhiyu Qian
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Guangliang Jiang
- Department of Urology, Ruijin Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Chuanyu Sun
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Ke Xu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
44
|
Weger M, Alpern D, Cherix A, Ghosal S, Grosse J, Russeil J, Gruetter R, de Kloet ER, Deplancke B, Sandi C. Mitochondrial gene signature in the prefrontal cortex for differential susceptibility to chronic stress. Sci Rep 2020; 10:18308. [PMID: 33110158 PMCID: PMC7591539 DOI: 10.1038/s41598-020-75326-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial dysfunction was highlighted as a crucial vulnerability factor for the development of depression. However, systemic studies assessing stress-induced changes in mitochondria-associated genes in brain regions relevant to depression symptomatology remain scarce. Here, we performed a genome-wide transcriptomic study to examine mitochondrial gene expression in the prefrontal cortex (PFC) and nucleus accumbens (NAc) of mice exposed to multimodal chronic restraint stress. We identified mitochondria-associated gene pathways as most prominently affected in the PFC and with lesser significance in the NAc. A more detailed mitochondrial gene expression analysis revealed that in particular mitochondrial DNA-encoded subunits of the oxidative phosphorylation complexes were altered in the PFC. The comparison of our data with a reanalyzed transcriptome data set of chronic variable stress mice and major depression disorder subjects showed that the changes in mitochondrial DNA-encoded genes are a feature generalizing to other chronic stress-protocols as well and might have translational relevance. Finally, we provide evidence for changes in mitochondrial outputs in the PFC following chronic stress that are indicative of mitochondrial dysfunction. Collectively, our work reinforces the idea that changes in mitochondrial gene expression are key players in the prefrontal adaptations observed in individuals with high behavioral susceptibility and resilience to chronic stress.
Collapse
Affiliation(s)
- Meltem Weger
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.,Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Daniel Alpern
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.,Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland
| | - Antoine Cherix
- Laboratory for Functional and Metabolic Imaging, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.,Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, England, UK
| | - Sriparna Ghosal
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Jocelyn Grosse
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Julie Russeil
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Rolf Gruetter
- Laboratory for Functional and Metabolic Imaging, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - E Ronald de Kloet
- Departement of Endocrinology and Metabolic Disease, Leiden University Medical Center, Leiden, The Netherlands
| | - Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.,Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.
| |
Collapse
|
45
|
Keugong Wado E, Kubicki M, Ngatanko AHH, Léa Blondelle KD, Jorelle Linda D, Roland RN, Balbine K, Lamshoeft M, Assongalem AE, Foyet HS. Anxiolytic and antidepressant effects of Ziziphus mucronata hydromethanolic extract in male rats exposed to unpredictable chronic mild stress: Possible mechanisms of actions. JOURNAL OF ETHNOPHARMACOLOGY 2020; 260:112987. [PMID: 32446929 DOI: 10.1016/j.jep.2020.112987] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/04/2020] [Accepted: 05/16/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ziziphus mucronata (ZM) is used traditionally in the treatment of mood and depression. However, no existing scientific data is confirming this traditional claim. AIM OF THE STUDY The present study was planned to investigate the anxiolytic and antidepressant-like effects of this plant in a stressed-induced depression model in rats. MATERIALS AND METHODS Depressive-like behaviors were induced by exposing rats to different stress paradigms daily for 30 days. A sucrose preference test was performed to assess anhedonia in rats. Anxiety and depression-related behavior were assessed. The oxidative parameters (lipid peroxidation, SOD and catalase activities) were evaluated. Pindolol and Flumazenil were also used to assess the mechanism of action of ZM extract. RESULTS The results showed that chronic administration of ZM (150, 300, and 600 mg/kg, p.o., 30 days) and imipramine treatment (20 mg/kg, p.o, 30 days) remarkably (P < 0.05) reversed the UCMS-induced behavioral changes observed in stress vehicle treated rats by reducing sucrose preference, decreased the immobility period in the FST and latency in NSF. Besides, ZM (300 and 600 mg/kg, p.o., 30 days) raised the percentages of time spent and number of open arms entries as well as the number of transitions. Also, ZM (300 mg/kg, (P < 0.05) decreased lipid peroxidation and increased both SOD and catalase activities (300 and 600 mg/kg, (P < 0.05)). These aforementioned behavioral indices were also completely nullified by pindolol a β-adrenoceptors blocker and 5-HT 1A/1B receptor antagonist but not by flumazenil, a benzodiazepine receptors antagonist. CONCLUSION ZM improved symptoms of anxiety and depression in behavioral despair paradigm in chronically stressed rats. The observed effects could be due to its capacities to restore the antioxidant status, and probably the modulation of monoamines transmissions.
Collapse
Affiliation(s)
- Eglantine Keugong Wado
- Department of Biological Sciences, Faculty of Science, University of Maroua, P.O. Box: 814, Maroua, Cameroon.
| | - Michael Kubicki
- Research and Development Bayer Crop Science, 40789, Monheim, Germany
| | | | | | - Damo Jorelle Linda
- Department of Biological Sciences, Faculty of Science, University of Maroua, P.O. Box: 814, Maroua, Cameroon
| | - Rebe Nhouma Roland
- Department of Biological Sciences, Faculty of Science, University of Maroua, P.O. Box: 814, Maroua, Cameroon
| | - Kamleu Balbine
- Department of Biological Sciences, Faculty of Science, University of Maroua, P.O. Box: 814, Maroua, Cameroon
| | - Marc Lamshoeft
- Research and Development Bayer Crop Science, 40789, Monheim, Germany
| | - Acha Emmanuel Assongalem
- Department of Biomedical Science, Faculty of Health Sciences, University of Buea, P.O. Box: 63, Buea, Cameroon
| | - Harquin Simplice Foyet
- Department of Biological Sciences, Faculty of Science, University of Maroua, P.O. Box: 814, Maroua, Cameroon
| |
Collapse
|
46
|
Guzik TJ, Mohiddin SA, Dimarco A, Patel V, Savvatis K, Marelli-Berg FM, Madhur MS, Tomaszewski M, Maffia P, D’Acquisto F, Nicklin SA, Marian AJ, Nosalski R, Murray EC, Guzik B, Berry C, Touyz RM, Kreutz R, Wang DW, Bhella D, Sagliocco O, Crea F, Thomson EC, McInnes IB. COVID-19 and the cardiovascular system: implications for risk assessment, diagnosis, and treatment options. Cardiovasc Res 2020; 116:1666-1687. [PMID: 32352535 PMCID: PMC7197627 DOI: 10.1093/cvr/cvaa106] [Citation(s) in RCA: 920] [Impact Index Per Article: 184.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/11/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
The novel coronavirus disease (COVID-19) outbreak, caused by SARS-CoV-2, represents the greatest medical challenge in decades. We provide a comprehensive review of the clinical course of COVID-19, its comorbidities, and mechanistic considerations for future therapies. While COVID-19 primarily affects the lungs, causing interstitial pneumonitis and severe acute respiratory distress syndrome (ARDS), it also affects multiple organs, particularly the cardiovascular system. Risk of severe infection and mortality increase with advancing age and male sex. Mortality is increased by comorbidities: cardiovascular disease, hypertension, diabetes, chronic pulmonary disease, and cancer. The most common complications include arrhythmia (atrial fibrillation, ventricular tachyarrhythmia, and ventricular fibrillation), cardiac injury [elevated highly sensitive troponin I (hs-cTnI) and creatine kinase (CK) levels], fulminant myocarditis, heart failure, pulmonary embolism, and disseminated intravascular coagulation (DIC). Mechanistically, SARS-CoV-2, following proteolytic cleavage of its S protein by a serine protease, binds to the transmembrane angiotensin-converting enzyme 2 (ACE2) -a homologue of ACE-to enter type 2 pneumocytes, macrophages, perivascular pericytes, and cardiomyocytes. This may lead to myocardial dysfunction and damage, endothelial dysfunction, microvascular dysfunction, plaque instability, and myocardial infarction (MI). While ACE2 is essential for viral invasion, there is no evidence that ACE inhibitors or angiotensin receptor blockers (ARBs) worsen prognosis. Hence, patients should not discontinue their use. Moreover, renin-angiotensin-aldosterone system (RAAS) inhibitors might be beneficial in COVID-19. Initial immune and inflammatory responses induce a severe cytokine storm [interleukin (IL)-6, IL-7, IL-22, IL-17, etc.] during the rapid progression phase of COVID-19. Early evaluation and continued monitoring of cardiac damage (cTnI and NT-proBNP) and coagulation (D-dimer) after hospitalization may identify patients with cardiac injury and predict COVID-19 complications. Preventive measures (social distancing and social isolation) also increase cardiovascular risk. Cardiovascular considerations of therapies currently used, including remdesivir, chloroquine, hydroxychloroquine, tocilizumab, ribavirin, interferons, and lopinavir/ritonavir, as well as experimental therapies, such as human recombinant ACE2 (rhACE2), are discussed.
Collapse
Affiliation(s)
- Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Kraków, Poland
| | - Saidi A Mohiddin
- Barts Heart Center, St Bartholomew’s NHS Trust, London, UK
- William Harvey Institute Queen Mary University of London, London, UK
| | | | - Vimal Patel
- Barts Heart Center, St Bartholomew’s NHS Trust, London, UK
| | | | | | - Meena S Madhur
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, UK
| | - Pasquale Maffia
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | - Stuart A Nicklin
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Ali J Marian
- Department of Medicine, Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Ryszard Nosalski
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Kraków, Poland
| | - Eleanor C Murray
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Bartlomiej Guzik
- Jagiellonian University Medical College, Institute of Cardiology, Department of Interventional Cardiology; John Paul II Hospital, Krakow, Poland
| | - Colin Berry
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Reinhold Kreutz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institut für Klinische Pharmakologie und Toxikologie, Germany
| | - Dao Wen Wang
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - David Bhella
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, UK
| | - Orlando Sagliocco
- Emergency Department, Intensive Care Unit; ASST Bergamo Est Bolognini Hospital Bergamo, Italy
| | - Filippo Crea
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Emma C Thomson
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, UK
- Department of Infectious Diseases, Queen Elizabeth University Hospital, Glasgow, UK
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
47
|
Chaves RDC, Mallmann ASV, de Oliveira NF, Capibaribe VCC, da Silva DMA, Lopes IS, Valentim JT, Barbosa GR, de Carvalho AMR, Fonteles MMDF, Gutierrez SJC, Barbosa Filho JM, de Sousa FCF. The neuroprotective effect of Riparin IV on oxidative stress and neuroinflammation related to chronic stress-induced cognitive impairment. Horm Behav 2020; 122:104758. [PMID: 32304685 DOI: 10.1016/j.yhbeh.2020.104758] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Cognitive impairment is identified as one of the diagnostic criteria for major depressive disorder and can extensively affect the quality of life of patients. Based on these findings, this study aimed to investigate the possible effects of Riparin IV (Rip IV) on cognitive impairment induced by chronic administration of corticosterone in mice. METHODS Female Swiss mice were divided into four groups: control (Control), corticosterone (Cort), Riparin IV (Cort + Rip IV), and Fluvoxamine (Cort + Flu). Three groups were administered corticosterone (20 mg/kg) subcutaneously during the 22-day study, while the control group received only vehicle. After the 14th day, the groups were administered medications: Riparin IV (Rip IV), fluvoxamine (Flu), or distilled water, by gavage, 1 h after the subcutaneous injections. After treatment, mice underwent behavioral testing, and brain areas were removed for oxidative stress and cytokine content assays. RESULTS The results revealed that Cort-treated mice developed a cognitive impairment and exhibited a neuroinflammatory profile with an oxidative load and Th1/Th2 cytokine imbalance. Rip IV treatment significantly ameliorated the cognitive deficit induced by Cort and displayed a neuroprotective effect. CONCLUSION The antidepressant-like ability of Rip IV treatment against chronic Cort-induced stress may be due to its potential to mitigate inflammatory damage and oxidative stress. The antioxidant and anti-inflammatory effect observed indicates Rip IV as a possible drug for antidepressant treatment of non-responsive patients with severe and cognitive symptoms.
Collapse
Affiliation(s)
- Raquell de Castro Chaves
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Auriana Serra Vasconcelos Mallmann
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Natália Ferreira de Oliveira
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Victor Celso Cavalcanti Capibaribe
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Daniel Moreira Alves da Silva
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Iardja Stéfane Lopes
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - José Tiago Valentim
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Giovanna Riello Barbosa
- Multi-User Facility, Drug Research and Development Center, Federal University of Ceará, Brazil
| | - Alyne Mara Rodrigues de Carvalho
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | | | - Stanley Juan Chavez Gutierrez
- Laboratory of Pharmaceutical Chemistry, Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, Piauí, Brazil
| | - José Maria Barbosa Filho
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutical Science, Federal University of Paraiba, João Pessoa, Paraiba, Brazil
| | - Francisca Cléa Florenço de Sousa
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil.
| |
Collapse
|
48
|
Duda P, Hajka D, Wójcicka O, Rakus D, Gizak A. GSK3β: A Master Player in Depressive Disorder Pathogenesis and Treatment Responsiveness. Cells 2020; 9:cells9030727. [PMID: 32188010 PMCID: PMC7140610 DOI: 10.3390/cells9030727] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/12/2020] [Accepted: 03/14/2020] [Indexed: 12/11/2022] Open
Abstract
Glycogen synthase kinase 3β (GSK3β), originally described as a negative regulator of glycogen synthesis, is a molecular hub linking numerous signaling pathways in a cell. Specific GSK3β inhibitors have anti-depressant effects and reduce depressive-like behavior in animal models of depression. Therefore, GSK3β is suggested to be engaged in the pathogenesis of major depressive disorder, and to be a target and/or modifier of anti-depressants’ action. In this review, we discuss abnormalities in the activity of GSK3β and its upstream regulators in different brain regions during depressive episodes. Additionally, putative role(s) of GSK3β in the pathogenesis of depression and the influence of anti-depressants on GSK3β activity are discussed.
Collapse
|
49
|
Carlton CN, Sullivan-Toole H, Ghane M, Richey JA. Reward Circuitry and Motivational Deficits in Social Anxiety Disorder: What Can Be Learned From Mouse Models? Front Neurosci 2020; 14:154. [PMID: 32174811 PMCID: PMC7054462 DOI: 10.3389/fnins.2020.00154] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 02/10/2020] [Indexed: 01/09/2023] Open
Abstract
Social anxiety disorder (SAD) is a common and serious psychiatric condition that typically emerges during adolescence and persists into adulthood if left untreated. Prevailing interventions focus on modulating threat and arousal systems but produce only modest rates of remission. This gap in efficacy suggests that most mainstream treatment concepts do not sufficiently target core processes involved in the onset and maintenance of SAD. This idea has further driven the development of new theoretical models that target dopamine (DA)-driven reward circuitry and motivational deficits that appear to be systematically altered in SAD. Most of the available data linking systemic alterations in DA neurobiology to SAD in humans, although abundant, remains at the level of correlational evidence. Accordingly, the purpose of this brief review is to critically evaluate the relevance of experimental work in rodent models that link details of DA function to symptoms of social anxiety. We conclude that, despite certain systematic limitations inherent in animal models, these approaches provide useful insights into human biomarkers of social anxiety including that (1) adolescence may serve as a critical period for the convergence of neurobiological and environmental factors that modify future expectations about social reward through experience dependent changes in DA-ergic circuitry, (2) females may show unique susceptibility to social anxiety symptoms when encountering relational instability that influences DA-related neural processes, and (3) separate from fear and arousal systems, the functional neurobiology of central DA systems contribute uniquely to susceptibility and maintenance of anhedonic factors relevant to human models of SAD.
Collapse
Affiliation(s)
- Corinne N Carlton
- Clinical Science Program, Department of Psychology, Virginia Tech, Blacksburg, VA, United States
| | - Holly Sullivan-Toole
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, United States
| | - Merage Ghane
- Clinical Science Program, Department of Psychology, Virginia Tech, Blacksburg, VA, United States
| | - John A Richey
- Clinical Science Program, Department of Psychology, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
50
|
Zanese M, Tomaselli G, Roullot-Lacarrière V, Moreau M, Bellocchio L, Grel A, Marsicano G, Sans N, Vallée M, Revest JM. Alpha technology: A powerful tool to detect mouse brain intracellular signaling events. J Neurosci Methods 2020; 332:108543. [PMID: 31830543 DOI: 10.1016/j.jneumeth.2019.108543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 11/15/2019] [Accepted: 12/05/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Phosphorylation by protein kinases is a fundamental molecular process involved in the regulation of signaling activities in living organisms. Understanding this complex network of phosphorylation, especially phosphoproteins, is a necessary step for grasping the basis of cellular pathophysiology. Studying brain intracellular signaling is a particularly complex task due to the heterogeneous complex nature of the brain tissue, which consists of many embedded structures. NEW METHOD Overcoming this degree of complexity requires a technology with a high throughput and economical in the amount of biological material used, so that a large number of signaling pathways may be analyzed in a large number of samples. We have turned to Alpha (Amplified Luminescent Proximity Homogeneous Assay) technology. COMPARISON WITH EXISTING METHOD Western blot is certainly the most commonly used method to measure the phosphorylation state of proteins. Even though Western blot is an accurate and reliable method for analyzing modifications of proteins, it is a time-consuming and large amounts of samples are required. Those two parameters are critical when the goal of the research is to comprehend multi-signaling proteic events so as to analyze several targets from small brain areas. RESULT Here we demonstrate that Alpha technology is particularly suitable for studying brain signaling pathways by allowing rapid, sensitive, reproducible and semi-quantitative detection of phosphoproteins from individual mouse brain tissue homogenates and from cell fractionation and synaptosomal preparations of mouse hippocampus. CONCLUSION Alpha technology represents a major experimental step forward in unraveling the brain phosphoprotein-related molecular mechanisms involved in brain-related disorders.
Collapse
Affiliation(s)
- Marion Zanese
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Giovanni Tomaselli
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Valérie Roullot-Lacarrière
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Maïté Moreau
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Luigi Bellocchio
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Agnès Grel
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Giovanni Marsicano
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Nathalie Sans
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Monique Vallée
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Jean-Michel Revest
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France.
| |
Collapse
|