1
|
Payet JM, Baratta MV, Christianson JP, Lowry CA, Hale MW. Modulation of dorsal raphe nucleus connectivity and serotonergic signalling to the insular cortex in the prosocial effects of chronic fluoxetine. Neuropharmacology 2025; 272:110406. [PMID: 40081797 DOI: 10.1016/j.neuropharm.2025.110406] [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: 10/28/2024] [Revised: 01/22/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Long-term exposure to fluoxetine and other selective serotonin reuptake inhibitors alters social and anxiety-related behaviours, including social withdrawal, which is a symptom of several neuropsychiatric disorders. Adaptive changes in serotonergic neurotransmission likely mediate this delayed effect, although the exact mechanisms are still unclear. Here we investigated the functional circuitry underlying the biphasic effects of fluoxetine on social approach-avoidance behaviour and explored the place of serotonergic dorsal raphe nucleus (DR) ensembles in this network, using c-Fos-immunoreactivity as a correlate of activity. Graph theory-based network analysis revealed changes in patterns of functional connectivity and identified neuronal populations in the insular cortex (IC) and serotonergic populations in the DR as central targets to the prosocial effects of chronic fluoxetine. To determine the role of serotonergic projections to the IC, a retrograde tracer was micro-injected in the IC prior to fluoxetine treatment and social behaviour testing. Chronic fluoxetine increased c-Fos immunoreactivity in insula-projecting neurons of the rostral, ventral part of the DR (DRV). Using a virally delivered Tet-Off platform for temporally-controlled marking of neuronal activation, we observed that chronic fluoxetine may affect social behaviour by influencing independent but interconnected populations of serotonergic DR ensembles. These findings suggest that sustained fluoxetine exposure causes adaptive changes in functional connectivity due to altered serotonergic neurotransmission in DR projection targets, and the increased serotonergic signalling to the IC likely mediates some of the therapeutic effects of fluoxetine on social behaviour.
Collapse
Affiliation(s)
- Jennyfer M Payet
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Michael V Baratta
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80301, USA
| | - John P Christianson
- Department of Psychology and Neuroscience, Boston College, Chestnut Hill, MA, 02467, USA
| | - Christopher A Lowry
- Department of Integrative Physiology, Center for Neuroscience, and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Matthew W Hale
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
2
|
Valentine C, Mitcheltree H, Sjövall IAK, Khalil MH. Architecturally Mediated Allostasis and Neurosustainability: A Proposed Theoretical Framework for the Impact of the Built Environment on Neurocognitive Health. Brain Sci 2025; 15:201. [PMID: 40002534 PMCID: PMC11853682 DOI: 10.3390/brainsci15020201] [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: 01/04/2025] [Revised: 02/02/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
The global rise in mental health-related disorders represents a significant health and wellbeing challenge, imposing a substantial social and economic burden on individuals, communities, and healthcare systems. According to the World Health Organization, one in four people globally will be affected by mental or neurological disorders at some point in their lives, highlighting a significant global health concern that warrants carefully considered and innovative responses. While mental health challenges arise from complex, multifaceted factors, emerging research indicates that the built environment-the architecture of our homes, workplaces, and public spaces-may exert a critical but underappreciated influence on mental health outcomes. This paper outlines a novel theoretical framework for how visual stressors in the built environment might trigger neurophysiological stress responses via the HPA and SAM axes, potentially contributing over time to allostatic load. In this paper, it is proposed that chronic physiological strain can alter neuroplastic processes and neurogenesis in key brain regions-such as the hippocampus, prefrontal cortex (PFC), anterior cingulate cortex (ACC), and amygdala-thereby affecting cognitive health, emotional regulation, and overall mental wellbeing. Drawing on the principle of neurosustainability, this paper suggests that long-term exposure to stress-inducing environments may create feedback loops, particularly involving the amygdala, that have downstream effects on other brain areas and may be linked to adverse mental health outcomes such as depression. By presenting this framework, this paper aims to inspire further inquiry and applied experimental research into the intersection of neurophysiology, mental health, and the built environment, with a particular emphasis on rigorous testing and validation of the proposed mechanisms, that may then be translated into practical architectural design strategies for supporting health and wellbeing. In doing so, it is hoped that this work may contribute to a more holistic approach to improving mental health that integrates the creation of nurturing, resilient spaces into the broader public health agenda.
Collapse
Affiliation(s)
- Cleo Valentine
- Department of Architecture, University of Cambridge, Cambridge CB2 1PX, UK; (H.M.); (M.H.K.)
| | - Heather Mitcheltree
- Department of Architecture, University of Cambridge, Cambridge CB2 1PX, UK; (H.M.); (M.H.K.)
| | | | - Mohamed Hesham Khalil
- Department of Architecture, University of Cambridge, Cambridge CB2 1PX, UK; (H.M.); (M.H.K.)
| |
Collapse
|
3
|
Gimbel BA, Wozniak JR, Mueller BA, Tuominen KA, Ernst AM, Anthony ME, de Water E, Roediger DJ. Regional hippocampal thinning and gyrification abnormalities and associated cognition in children with prenatal alcohol exposure. J Neurodev Disord 2025; 17:5. [PMID: 39910445 PMCID: PMC11796126 DOI: 10.1186/s11689-025-09595-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/30/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) impacts hippocampal structure and function, contributing to deficits in memory and decision-making in affected individuals. Here, we evaluate hippocampal anomalies in children with PAE and an unexposed comparison group using advanced MRI methods that characterize hippocampal curvature and thickness. METHODS Participants, ages 8 to 16 years, included children with PAE (n = 48) and an unexposed comparison group (n = 46) who underwent a dysmorphology exam, neuropsychological assessment, and an MRI scan. Height, weight, head circumference, and dysmorphic facial features were evaluated. Of those with PAE, 4.2% had fetal alcohol syndrome (FAS), 22.9% had partial FAS, and 72.9% had alcohol-related neurodevelopmental disorder. Neuropsychological testing included measures of intelligence and memory functioning. T1-weighted anatomical data were processed with the Hippunfold pipeline, which "unfolds" the complex hippocampal structure onto a template surface and provides measures of thickness and gyrification/curvature at each vertex. Permutation Analysis of Linear Models (PALM) was used to test for group differences (PAE vs. comparison) in hippocampal thickness and gyrification at each vertex and also to assess correlations with cognitive functioning. RESULTS There were significant regional differences in thickness and gyrification across bilateral hippocampi, with the PAE group showing substantially thinner tissue and less curvature than the comparison group, especially in CA1 and subiculum regions. For those with PAE, thinner subicular tissue (bilateral) was associated with lower IQ. Also in the PAE group, lower episodic memory performance was associated with thinness in the right hippocampus, especially in the subiculum region. There were no significant regional hippocampal patterns that were associated with cognitive functioning for individuals in the unexposed comparison group. CONCLUSIONS We used a novel MRI method to evaluate hippocampal structure in children with PAE and an unexposed comparison group. The data suggest that PAE disrupts hippocampal development, impacting both the early-stage folding of the structure and its ultimate thickness. The data also demonstrate that these developmental anomalies have functional consequences in terms of core memory functions as well as global intellectual functioning in children with PAE.
Collapse
Affiliation(s)
- Blake A Gimbel
- Nationwide Children's Hospital, Columbus, USA
- The Ohio State University, Columbus, USA
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Grillo AR. Polygene by environment interactions predicting depressive outcomes. Am J Med Genet B Neuropsychiatr Genet 2025; 198:e33000. [PMID: 39012198 DOI: 10.1002/ajmg.b.33000] [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: 05/01/2023] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024]
Abstract
Depression is a major public health problem with a continued need to uncover its etiology. Current models of depression contend that gene-by-environment (G × E) interactions influence depression risk, and further, that depression is polygenic. Thus, recent models have emphasized two polygenic approaches: a hypothesis-driven multilocus genetic profile score (MGPS; "MGPS × E") and a polygenic risk score (PRS; "PRS × E") derived from genome-wide association studies (GWAS). This review for the first time synthesizes current knowledge on polygene by environment "P × E" interaction research predicting primarily depression-related outcomes, and in brief, neurobiological outcomes. The "environment" of focus in this project is stressful life events. It further discusses findings in the context of differential susceptibility and diathesis-stress theories-two major theories guiding G × E work. This synthesis indicates that, within the MGPS literature, polygenic scores based on the serotonin system, the HPA axis, or across multiple systems, interact with environmental stress exposure to predict outcomes at multiple levels of analyses and most consistently align with differential susceptibility theory. Depressive outcomes are the most studied, but neuroendocrine, and neuroimaging findings are observed as well. By contrast, vast methodological differences between GWAS-based PRS studies contribute to mixed findings that yield inconclusive results.
Collapse
Affiliation(s)
- Alessandra R Grillo
- Department of Psychology, University of North Carolina, Greensboro, North Carolina, USA
| |
Collapse
|
5
|
Ontiveros-Araiza LF. The Neurobehavioral State hypothesis. Biosystems 2025; 247:105361. [PMID: 39521269 DOI: 10.1016/j.biosystems.2024.105361] [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: 06/07/2024] [Revised: 11/02/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Since the early attempts to understand the brain made by Greek philosophers more than 2000 years ago, one of the main questions in neuroscience has been how the brain perceives all the stimuli in the environment and uses this information to implement a response. Recent hypotheses of the neural code rely on the existence of an ideal observer, whether on specific areas of the cerebral cortex or distributed network composed of cortical and subcortical elements. The Neurobehavioral State hypothesis stipulates that neurons are in a quasi-stable state due to the dynamic interaction of their molecular components. This increases their computational capabilities and electrophysiological behavior further than a binary active/inactive state. Together, neuronal populations across the brain learn to identify and associate internal and external stimuli with actions and emotions. Furthermore, such associations can be stored through the regulation of neuronal components as new quasi-stable states. Using this framework, behavior arises as the result of the dynamic interaction between internal and external stimuli together with previously established quasi-stable states that delineate the behavioral response. Finally, the Neurobehavioral State hypothesis is firmly grounded on present evidence of the complex dynamics within the brain, from the molecular to the network level, and avoids the need for a central observer by proposing the brain configures itself through experience-driven associations.
Collapse
Affiliation(s)
- Luis Fernando Ontiveros-Araiza
- Department of Cognitive Neuroscience, Division of Neuroscience, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, Coyoacán, 04510, Mexico City, Mexico.
| |
Collapse
|
6
|
Maximino C. Biocultural Aspects of Mental Distress: Expanding the Biomedical Model Towards an Integrative Biopsychosocial Understanding of Disorder. Integr Psychol Behav Sci 2024; 59:5. [PMID: 39725806 DOI: 10.1007/s12124-024-09869-1] [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] [Accepted: 11/20/2024] [Indexed: 12/28/2024]
Abstract
To produce a theoretical approach about the relations between neuroscience and psychopathology that expands beyond the biomedical model to include a non-reductionist, enactive, and biocultural perspective. An integrative review, drawing from the biocultural approach from Anthropology, is used to produce examples from epigenetics, neuroplasticity, and functional neuroanatomy. A biocultural approach points to a brain that is highly plastic, reinforcing a much more complex model in which biological vulnerabilities and the historical-cultural environment co-construct each other. The examples given seem to point to the pressing need for a critical expansion of reductionist models of psychopathology. Importantly, the cultural-historical environment to which we refer is not a set of neutral social relations to which individuals are homogeneously exposed, such that aspects that are usually studied under the social determinants of health and disease (poverty, discrimination, violence, and other factors that represent sources of control, production, and distribution of material resources, ideology, and power) need to be incorporated in adequate biopsychosocial models of mental distress.
Collapse
Affiliation(s)
- Caio Maximino
- Laboratório de Neurociências e Comportamento, Faculdade de Psicologia, Instituto de Estudos em Saúde e Biológicas, Universidade Federal do Sul e Sudeste do Pará, Av. dos Ipês, S/N, Marabá, PA, 68500-000, Brazil.
| |
Collapse
|
7
|
Sudimac S, Kühn S. Can a nature walk change your brain? Investigating hippocampal brain plasticity after one hour in a forest. ENVIRONMENTAL RESEARCH 2024; 262:119813. [PMID: 39155041 DOI: 10.1016/j.envres.2024.119813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/09/2024] [Accepted: 08/16/2024] [Indexed: 08/20/2024]
Abstract
In cities, the incidence of mental disorders is higher, while visits to nature have been reported to benefit mental health and brain function. However, there is a lack of knowledge about how exposure to natural and urban environments affects brain structure. To explore the causal relationship between exposure to these environments and the hippocampal formation, 60 participants were sent on a one hour walk in either a natural (forest) or an urban environment (busy street), and high-resolution hippocampal imaging was performed before and after the walks. We found that the participants who walked in the forest had an increase in subiculum volume, a hippocampal subfield involved in stress response inhibition, while no change was observed after the urban walk. However, this result did not withstand Bonferroni correction for multiple comparisons. Furthermore, the increase in subiculum volume after the forest walk was associated with a decrease in self-reported rumination. These results indicate that visits to nature can lead to observable alterations in brain structure, with potential benefits for mental health and implications for public health and urban planning policies.
Collapse
Affiliation(s)
- Sonja Sudimac
- Max Planck Institute for Human Development, Center for Environmental Neuroscience, Lentzeallee 94, 14195, Berlin, Germany.
| | - Simone Kühn
- Max Planck Institute for Human Development, Center for Environmental Neuroscience, Lentzeallee 94, 14195, Berlin, Germany; University Medical Center Hamburg-Eppendorf, Department of Psychiatry and Psychotherapy, Martinistr. 52, 20251, Hamburg, Germany; Max Planck UCL Centre for Computational Psychiatry and Ageing Research Berlin, Germany and London, UK, Lentzeallee 94, 14195, Berlin, Germany
| |
Collapse
|
8
|
Shafiek MS, Mekky RY, Nassar NN, El-Yamany MF, Rabie MA. Vortioxetine ameliorates experimental autoimmune encephalomyelitis model of multiple sclerosis in mice via activation of PI3K/Akt/CREB/BDNF cascade and modulation of serotonergic pathway signaling. Eur J Pharmacol 2024; 982:176929. [PMID: 39181226 DOI: 10.1016/j.ejphar.2024.176929] [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: 05/22/2024] [Revised: 08/01/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Multiple sclerosis (MS) is a chronic condition characterized by immune cell infiltration and cytokine overproduction that led to myelin sheath inflammatory assaults, thus causing axonal destruction. The former consequently provokes motor impairment and psychological disorders. Markedly, depression is one of the most prevalent lifelong comorbidities that negatively impacts the quality of life in MS patients. Vortioxetine (VTX), a multi-modal molecule prescribed to manage depression and anxiety disorder, additionally, it displays a promising neuroprotective properties against neurodegenerative diseases such as Alzheimer's and Parkinson's. To this end, the present study investigated the potential therapeutic efficacy of VTX against experimental autoimmune encephalomyelitis (EAE) model of MS in mice. Notably, treatment with VTX significantly ameliorated EAE-induced motor disability, as evident by enhanced performance in open field, rotarod and grip strength tests, alongside a reduction in immobility time during the forced swimming test, indicating a mitigation of the depressive-like behavior; outcomes that were corroborated with histological examinations and biochemical analyses. Mechanistically, VTX enhanced serotonin levels by inhibiting both serotonin transporter (SERT) and indoleamine 2,3-dioxygenase (IDO) enzyme, thereby promoting the activation of serotonin 1A (5-HT1A) receptor. The latter triggered the stimulation of phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) cascade that entailed activation/phosphorylation of cAMP response element-binding protein (CREB). This activation increased brain derived neurotrophic factor (BDNF) and myelin basic protein (MBP) contents that mitigated demyelination in the corpus callosum. Furthermore, VTX suppressed phospho serine 536 nuclear factor kappa B (pS536 NF-κB p65) activity and reduced tumor necrosis factor-alpha (TNF-α) production. The results underscore VTX's beneficial effects on disease severity in EAE model of MS in mice by amending both inflammatory and neurodegenerative components of MS progression.
Collapse
Affiliation(s)
- Marwa S Shafiek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | - Radwa Y Mekky
- Department of Pharmacology and Toxicology, October University for Modern Science and Arts (MSA), Giza, 12622, Egypt
| | - Noha N Nassar
- Department of Pharmacology and Toxicology, October University for Modern Science and Arts (MSA), Giza, 12622, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt.
| |
Collapse
|
9
|
Ishikawa A, Takanuma T, Hashimoto N, Tsudzuki M. Association between Temperament and Stress-related Gene Expression in Day-old Chickens. J Poult Sci 2024; 61:2024022. [PMID: 39130209 PMCID: PMC11310665 DOI: 10.2141/jpsa.2024022] [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: 06/19/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024] Open
Abstract
Stress in day-old chickens from commercial hatcheries is associated with problematic behavior in adult animals. Recently, we developed a new behavioral handling test for day-old chickens and demonstrated that it assessed temperament differences between seven breeds of native Japanese and Western chickens. In this study, we used 2-day-old male chicks from five of the above breeds to investigate the relationship between temperament and mRNA levels of three stress-related genes (nuclear receptor subfamily 3 group C member 1 (NR3C1), cytochrome P450 family 11 subfamily A member 1, and hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 1) involved in the hypothalamic-pituitary-adrenal axis. Principal component analysis of 10 behavioral traits for the handling test revealed that the Fayoumi breed and Hiroshima line of the Chabo breed, both of which exhibited boisterous temperament, clustered separately from the other breeds. Only NR3C1 expression showed a significant positive correlation with two behavioral traits (general vocalization and approaching the wall), and a negative correlation with movement. These results suggest that the complex temperament of day-old chickens is regulated, in part, by stress-related genes along the hypothalamic-pituitary-adrenal axis.
Collapse
Affiliation(s)
- Akira Ishikawa
- Graduate School of Bioagricultural Sciences, Nagoya
University, Nagoya 464-8601, Japan
| | - Tomoka Takanuma
- Graduate School of Bioagricultural Sciences, Nagoya
University, Nagoya 464-8601, Japan
| | - Norikazu Hashimoto
- Livestock Experiment Station, Wakayama Prefecture,
Hidaka-Gun 644-1111, Japan
| | - Masaoki Tsudzuki
- Graduate School of Integrated Sciences for Life, Hiroshima
University, Higashi-Hiroshima 739-8525, Japan
| |
Collapse
|
10
|
Armio RL, Laurikainen H, Ilonen T, Walta M, Sormunen E, Tolvanen A, Salokangas RKR, Koutsouleris N, Tuominen L, Hietala J. Longitudinal study on hippocampal subfields and glucose metabolism in early psychosis. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:66. [PMID: 39085221 PMCID: PMC11291638 DOI: 10.1038/s41537-024-00475-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 05/11/2024] [Indexed: 08/02/2024]
Abstract
Altered hippocampal morphology and metabolic pathology, but also hippocampal circuit dysfunction, are established phenomena seen in psychotic disorders. Thus, we tested whether hippocampal subfield volume deficits link with deviations in glucose metabolism commonly seen in early psychosis, and whether the glucose parameters or subfield volumes change during follow-up period using one-year longitudinal study design of 78 first-episode psychosis patients (FEP), 48 clinical high-risk patients (CHR) and 83 controls (CTR). We also tested whether hippocampal morphology and glucose metabolism relate to clinical outcome. Hippocampus subfields were segmented with Freesurfer from 3T MRI images and parameters of glucose metabolism were determined in fasting plasma samples. Hippocampal subfield volumes were consistently lower in FEPs, and findings were more robust in non-affective psychoses, with strongest decreases in CA1, molecular layer and hippocampal tail, and in hippocampal tail of CHRs, compared to CTRs. These morphometric differences remained stable at one-year follow-up. Both non-diabetic CHRs and FEPs had worse glucose parameters compared to CTRs at baseline. We found that, insulin levels and insulin resistance increased during the follow-up period only in CHR, effect being largest in the CHRs converting to psychosis, independent of exposure to antipsychotics. The worsening of insulin resistance was associated with deterioration of function and symptoms in CHR. The smaller volume of hippocampal tail was associated with higher plasma insulin and insulin resistance in FEPs, at the one-year follow-up. Our longitudinal study supports the view that temporospatial hippocampal subfield volume deficits are stable near the onset of first psychosis, being more robust in non-affective psychoses, but less prominent in the CHR group. Specific subfield defects were related to worsening glucose metabolism during the progression of psychosis, suggesting that hippocampus is part of the circuits regulating aberrant glucose metabolism in early psychosis. Worsening of glucose metabolism in CHR group was associated with worse clinical outcome measures indicating a need for heightened clinical attention to metabolic problems already in CHR.
Collapse
Grants
- Turun Yliopistollisen Keskussairaalan Koulutus- ja Tutkimussäätiö (TYKS-säätiö)
- Alfred Kordelinin Säätiö (Alfred Kordelin Foundation)
- Finnish Cultural Foundation | Varsinais-Suomen Rahasto (Varsinais-Suomi Regional Fund)
- Suomalainen Lääkäriseura Duodecim (Finnish Medical Society Duodecim)
- Turun Yliopisto (University of Turku)
- This work was supported by funding for the VAMI-project (Turku University Hospital, state research funding, no. P3848), partly supported by EU FP7 grants (PRONIA, grant a # 602152 and METSY grant #602478). Dr. Armio received personal funding from Doctoral Programme in Clinical Research at the University of Turku, grants from State Research Funding, Turunmaa Duodecim Society, Finnish Psychiatry Research Foundation, Finnish University Society of Turku (Valto Takala Foundation), Tyks-foundation, The Finnish Medical Foundation (Maija and Matti Vaskio fund), University of Turku, The Alfred Kordelin Foundation, Finnish Cultural Foundation (Terttu Enckell fund and Ritva Helminen fund) and The Alfred Kordelin foundation. Further, Dr. Tuominen received personal grant from Sigrid Juselius and Orion research foundation and NARSAD Young Investigator Grant from the Brain & Behavior Research Foundation.
- This work was supported by funding for the VAMI-project (Turku University Hospital, state research funding, no. P3848), partly supported by EU FP7 grants (PRONIA, grant a # 602152 and METSY grant #602478). Dr. Tuominen received personal grant from Sigrid Juselius and Orion research foundation and NARSAD Young Investigator Grant from the Brain & Behavior Research Foundation.
Collapse
Affiliation(s)
- Reetta-Liina Armio
- PET Centre, Turku University Hospital, 20520, Turku, Finland.
- Department of Psychiatry, University of Turku, 20700, Turku, Finland.
- Department of Psychiatry, Turku University Hospital, 20520, Turku, Finland.
| | - Heikki Laurikainen
- PET Centre, Turku University Hospital, 20520, Turku, Finland
- Department of Psychiatry, University of Turku, 20700, Turku, Finland
- Department of Psychiatry, Turku University Hospital, 20520, Turku, Finland
| | - Tuula Ilonen
- Department of Psychiatry, University of Turku, 20700, Turku, Finland
| | - Maija Walta
- PET Centre, Turku University Hospital, 20520, Turku, Finland
- Department of Psychiatry, University of Turku, 20700, Turku, Finland
- Department of Psychiatry, Turku University Hospital, 20520, Turku, Finland
| | - Elina Sormunen
- PET Centre, Turku University Hospital, 20520, Turku, Finland
- Department of Psychiatry, University of Turku, 20700, Turku, Finland
- Department of Psychiatry, Turku University Hospital, 20520, Turku, Finland
| | - Arvi Tolvanen
- Department of Psychiatry, University of Turku, 20700, Turku, Finland
| | | | - Nikolaos Koutsouleris
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilian University, D-80336, Munich, Germany
| | - Lauri Tuominen
- Department of Psychiatry, Turku University Hospital, 20520, Turku, Finland
- The Royal's Institute of Mental Health Research, University of Ottawa, Ottawa, ON, Canada
- Department of Psychiatry, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jarmo Hietala
- PET Centre, Turku University Hospital, 20520, Turku, Finland
- Department of Psychiatry, University of Turku, 20700, Turku, Finland
- Department of Psychiatry, Turku University Hospital, 20520, Turku, Finland
| |
Collapse
|
11
|
Hasaniani N, Mostafa Rahimi S, Akbari M, Sadati F, Pournajaf A, Rostami-Mansoor S. The Role of Intestinal Microbiota and Probiotics Supplementation in Multiple Sclerosis Management. Neuroscience 2024; 551:31-42. [PMID: 38777135 DOI: 10.1016/j.neuroscience.2024.05.013] [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: 02/01/2024] [Revised: 04/26/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024]
Abstract
Multiple sclerosis (MS) is a neurological autoimmune disorder predominantly afflicting young adults. The etiology of MS is intricate, involving a variety of environmental and genetic factors. Current research increasingly focuses on the substantial contribution of gut microbiota in MS pathogenesis. The commensal microbiota resident within the intestinal milieu assumes a central role within the intricate network recognized as the gut-brain axis (GBA), wielding beneficial impact in neurological and psychological facets. As a result, the modulation of gut microbiota is considered a pivotal aspect in the management of neural disorders, including MS. Recent investigations have unveiled the possibility of using probiotic supplements as a promising strategy for exerting a positive impact on the course of MS. This therapeutic approach operates through several mechanisms, including the reinforcement of gut epithelial integrity, augmentation of the host's resistance against pathogenic microorganisms, and facilitation of mucosal immunomodulatory processes. The present study comprehensively explains the gut microbiome's profound influence on the central nervous system (CNS). It underscores the pivotal role played by probiotics in forming the immune system and modulating neurotransmitter function. Furthermore, the investigation elucidates various instances of probiotic utilization in MS patients, shedding light on the potential therapeutic advantages afforded by this intervention.
Collapse
Affiliation(s)
- Nima Hasaniani
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Seyed Mostafa Rahimi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Marziyeh Akbari
- Department of Genetics, Faculty of Science, Shahrekord University, Shahrekord, Iran
| | - Fahimeh Sadati
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Abazar Pournajaf
- Biomedical and Microbial Advanced Technologies (BMAT) Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Sahar Rostami-Mansoor
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
12
|
Ysbæk-Nielsen AT. Exploring volumetric abnormalities in subcortical L-HPA axis structures in pediatric generalized anxiety disorder. Nord J Psychiatry 2024; 78:402-410. [PMID: 38573199 DOI: 10.1080/08039488.2024.2335980] [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: 05/15/2023] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Pediatric generalized anxiety disorder (GAD) is debilitating and increasingly prevalent, yet its etiology remains unclear. Some believe the disorder to be propagated by chronic dysregulation of the limbic-hypothalamic-pituitary-adrenal (L-HPA) axis, but morphometric studies of implicated subcortical areas have been largely inconclusive. Recognizing that certain subcortical subdivisions are more directly involved in L-HPA axis functioning, this study aims to detect specific abnormalities in these critical areas. METHODS Thirty-eight MRI scans of preschool children with (n = 15) and without (n = 23) GAD underwent segmentation and between-group volumetric comparisons of the basolateral amygdala (BLA), ventral hippocampal subiculum (vSC), and mediodorsal medial magnocellular (MDm) area of the thalamus. RESULTS Children with GAD displayed significantly larger vSC compared to healthy peers, F(1, 31) = 6.50, pFDR = .048. On average, children with GAD presented with larger BLA and MDm, Fs(1, 31) ≥ 4.86, psFDR ≤ .054. Exploratory analyses revealed right-hemispheric lateralization of all measures, most notably the MDm, F(1, 31) = 8.13, pFDR = .024, the size of which scaled with symptom severity, r = .83, pFDR = .033. CONCLUSION The BLA, vSC, and MDm are believed to be involved in the regulation of anxiety and stress, both individually and collectively through the excitation and inhibition of the L-HPA axis. All were found to be enlarged in children with GAD, perhaps reflecting hypertrophy related to hyperexcitability, or early neuronal overgrowth. Longitudinal studies should investigate the relationship between these early morphological differences and the long-term subcortical atrophy previously observed.
Collapse
|
13
|
Jensen DEA, Ebmeier KP, Suri S, Rushworth MFS, Klein-Flügge MC. Nuclei-specific hypothalamus networks predict a dimensional marker of stress in humans. Nat Commun 2024; 15:2426. [PMID: 38499548 PMCID: PMC10948785 DOI: 10.1038/s41467-024-46275-y] [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: 06/02/2023] [Accepted: 02/21/2024] [Indexed: 03/20/2024] Open
Abstract
The hypothalamus is part of the hypothalamic-pituitary-adrenal axis which activates stress responses through release of cortisol. It is a small but heterogeneous structure comprising multiple nuclei. In vivo human neuroimaging has rarely succeeded in recording signals from individual hypothalamus nuclei. Here we use human resting-state fMRI (n = 498) with high spatial resolution to examine relationships between the functional connectivity of specific hypothalamic nuclei and a dimensional marker of prolonged stress. First, we demonstrate that we can parcellate the human hypothalamus into seven nuclei in vivo. Using the functional connectivity between these nuclei and other subcortical structures including the amygdala, we significantly predict stress scores out-of-sample. Predictions use 0.0015% of all possible brain edges, are specific to stress, and improve when using nucleus-specific compared to whole-hypothalamus connectivity. Thus, stress relates to connectivity changes in precise and functionally meaningful subcortical networks, which may be exploited in future studies using interventions in stress disorders.
Collapse
Affiliation(s)
- Daria E A Jensen
- Department of Experimental Psychology, University of Oxford, Tinsley Building, Mansfield Road, Oxford, OX1 3TA, UK.
- Wellcome Centre for Integrative Neuroimaging (WIN), Centre for Functional MRI of the Brain (FMRIB, University of Oxford, Nuffield Department of Clinical Neurosciences, Level 6, West Wing, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, UK.
- Clinic of Cognitive Neurology, University Medical Center Leipzig and Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstrasse 1a, 04103, Leipzig, Germany.
| | - Klaus P Ebmeier
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, UK
| | - Sana Suri
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, UK
- Wellcome Centre for Integrative Neuroimaging (WIN), Oxford Centre for Human Brain Activity (OHBA), University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, UK
| | - Matthew F S Rushworth
- Department of Experimental Psychology, University of Oxford, Tinsley Building, Mansfield Road, Oxford, OX1 3TA, UK
- Wellcome Centre for Integrative Neuroimaging (WIN), Centre for Functional MRI of the Brain (FMRIB, University of Oxford, Nuffield Department of Clinical Neurosciences, Level 6, West Wing, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Miriam C Klein-Flügge
- Department of Experimental Psychology, University of Oxford, Tinsley Building, Mansfield Road, Oxford, OX1 3TA, UK.
- Wellcome Centre for Integrative Neuroimaging (WIN), Centre for Functional MRI of the Brain (FMRIB, University of Oxford, Nuffield Department of Clinical Neurosciences, Level 6, West Wing, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, UK.
| |
Collapse
|
14
|
Buo C, Bearss RJ, Novak AG, Anello AE, Dakin JJ, Piet R. Serotonin stimulates female preoptic area kisspeptin neurons via activation of type 2 serotonin receptors in mice. Front Endocrinol (Lausanne) 2023; 14:1212854. [PMID: 37900129 PMCID: PMC10602649 DOI: 10.3389/fendo.2023.1212854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023] Open
Abstract
Background The neuroendocrine control of ovulation is orchestrated by neuronal circuits that ultimately drive the release of gonadotropin-releasing hormone (GnRH) from the hypothalamus to trigger the preovulatory surge in luteinizing hormone (LH) secretion. While estrogen feedback signals are determinant in triggering activation of GnRH neurons, through stimulation of afferent kisspeptin neurons in the rostral periventricular area of the third ventricle (RP3VKISS1 neurons), many neuropeptidergic and classical neurotransmitter systems have been shown to regulate the LH surge. Among these, several lines of evidence indicate that the monoamine neurotransmitter serotonin (5-HT) has an excitatory, permissive, influence over the generation of the surge, via activation of type 2 5-HT (5-HT2) receptors. The mechanisms through which this occurs, however, are not well understood. We hypothesized that 5-HT exerts its influence on the surge by stimulating RP3VKISS1 neurons in a 5-HT2 receptor-dependent manner. Methods We tested this using kisspeptin neuron-specific calcium imaging and electrophysiology in brain slices obtained from male and female mice. Results We show that exogenous 5-HT reversibly increases the activity of the majority of RP3VKISS1 neurons. This effect is more prominent in females than in males, is likely mediated directly at RP3VKISS1 neurons and requires activation of 5-HT2 receptors. The functional impact of 5-HT on RP3VKISS1 neurons, however, does not significantly vary during the estrous cycle. Conclusion Taken together, these data suggest that 5-HT2 receptor-mediated stimulation of RP3VKISS1 neuron activity might be involved in mediating the influence of 5-HT on the preovulatory LH surge.
Collapse
Affiliation(s)
- Carrie Buo
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Robin J. Bearss
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Alyssa G. Novak
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Anna E. Anello
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Jordan J. Dakin
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Richard Piet
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| |
Collapse
|
15
|
Khairuddin S, Lim WL, Aquili L, Tsui KC, Tse ACK, Jayalath S, Varma R, Sharp T, Benazzouz A, Steinbusch H, Blokland A, Temel Y, Lim LW. Prelimbic Cortical Stimulation Induces Antidepressant-like Responses through Dopaminergic-Dependent and -Independent Mechanisms. Cells 2023; 12:1449. [PMID: 37296570 PMCID: PMC10253143 DOI: 10.3390/cells12111449] [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: 04/05/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 06/12/2023] Open
Abstract
High-frequency stimulation (HFS) is a promising therapy for patients with depression. However, the mechanisms underlying the HFS-induced antidepressant-like effects on susceptibility and resilience to depressive-like behaviors remain obscure. Given that dopaminergic neurotransmission has been found to be disrupted in depression, we investigated the dopamine(DA)-dependent mechanism of the antidepressant-like effects of HFS of the prelimbic cortex (HFS PrL). We performed HFS PrL in a rat model of mild chronic unpredictable stress (CUS) together with 6-hydroxydopamine lesioning in the dorsal raphe nucleus (DRN) and ventral tegmental area (VTA). Animals were assessed for anxiety, anhedonia, and behavioral despair. We also examined levels of corticosterone, hippocampal neurotransmitters, neuroplasticity-related proteins, and morphological changes in dopaminergic neurons. We found 54.3% of CUS animals exhibited decreased sucrose consumption and were designated as CUS-susceptible, while the others were designated CUS-resilient. HFS PrL in both the CUS-susceptible and CUS-resilient animals significantly increased hedonia, reduced anxiety, decreased forced swim immobility, enhanced hippocampal DA and serotonin levels, and reduced corticosterone levels when compared with the respective sham groups. The hedonic-like effects were abolished in both DRN- and VTA-lesioned groups, suggesting the effects of HFS PrL are DA-dependent. Interestingly, VTA-lesioned sham animals had increased anxiety and forced swim immobility, which was reversed by HFS PrL. The VTA-lesioned HFS PrL animals also had elevated DA levels, and reduced p-p38 MAPK and NF-κB levels when compared to VTA-lesioned sham animals. These findings suggest that HFS PrL in stressed animals leads to profound antidepressant-like responses possibly through both DA-dependent and -independent mechanisms.
Collapse
Affiliation(s)
- Sharafuddin Khairuddin
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wei Ling Lim
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Biological Sciences, Sunway University, Bandar Sunway, Petaling Jaya 47500, Malaysia
| | - Luca Aquili
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Discipline of Psychology, College of Health and Education, Murdoch University, Perth 6150, Australia
| | - Ka Chun Tsui
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Anna Chung-Kwan Tse
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shehani Jayalath
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ruhani Varma
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Trevor Sharp
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Abdelhamid Benazzouz
- CNRS UMR5293, Institute of Neurodegenerative Diseases, University de Bordeaux, 33000 Bordeaux, France
| | - Harry Steinbusch
- Department of Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Arjan Blokland
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Yasin Temel
- Department of Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Neurosurgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Lee Wei Lim
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Biological Sciences, Sunway University, Bandar Sunway, Petaling Jaya 47500, Malaysia
| |
Collapse
|
16
|
Bessei W, Tetens J, Bennewitz J, Falker-Gieske C, Hofmann T, Piepho HP. Disturbed circadian rhythm of locomotor activity of pullets is related to feather pecking in laying hens. Poult Sci 2023; 102:102548. [PMID: 36907128 PMCID: PMC10024181 DOI: 10.1016/j.psj.2023.102548] [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: 11/30/2022] [Revised: 01/16/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
Various aspects of activity, such as spontaneous activity, explorative activity, activity in open-field tests, and hyperactivity syndrome have been explored as causal factors of feather pecking in laying hens, with no clear results. In all previous studies, mean values of activity over different time intervals were used as criteria. Incidental observation of alternated oviposition time in lines selected for high (HFP) and low feather pecking (LFP), supported by a recent study which showed differentially expressed genes related to the circadian clock in the same lines, led to the hypothesis that feather pecking may be related to a disturbed diurnal activity rhythm. Hence activity recordings of a previous generation of these lines have been reanalyzed. Data sets of a total of 682 pullets of 3 subsequent hatches of HFP, LFP, and an unselected control line (CONTR) were used. Locomotor activity was recorded in pullets housed in groups of mixed lines in a deep litter pen on 7 consecutive 13-h light phases, using a radio-frequency identification antenna system. The number of approaches to the antenna system was recorded as a measure of locomotor activity and analyzed using a generalized linear mixed model including hatch, line, time of day and the interactions of hatch × time of day and line × time of day as fixed effects. Significant effects were found for time and the interaction line × time of day but not for line. All lines showed a bimodal pattern of diurnal activity. The peak activity of the HFP in the morning was lower than that of the LFP and CONTR. In the afternoon peak all lines differed with the highest mean in the LFP followed by CONTR and HFP. The present results provide support for the hypothesis that a disturbed circadian clock plays a role in the development of feather pecking.
Collapse
Affiliation(s)
- Werner Bessei
- Institute of Animal Science, University of Hohenheim, Garbenstr. 17, 70599 Stuttgart, Germany.
| | - Jens Tetens
- Department of Animal Sciences, Georg-August-University, Burckhardtweg 2, 37077, Göttingen, Germany
| | - Jörn Bennewitz
- Institute of Animal Science, University of Hohenheim, Garbenstr. 17, 70599 Stuttgart, Germany
| | - Clemens Falker-Gieske
- Department of Animal Sciences, Georg-August-University, Burckhardtweg 2, 37077, Göttingen, Germany
| | - Tanja Hofmann
- Institute of Animal Science, University of Hohenheim, Garbenstr. 17, 70599 Stuttgart, Germany
| | - Hans-Peter Piepho
- Institute of Crop Science, University of Hohenheim, Fruwirthstr. 23, 70599 Stuttgart, Germany
| |
Collapse
|
17
|
Urban MM, Stingl MR, Meinhardt MW. Mini-review: The neurobiology of treating substance use disorders with classical psychedelics. Front Neurosci 2023; 17:1156319. [PMID: 37139521 PMCID: PMC10149865 DOI: 10.3389/fnins.2023.1156319] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/23/2023] [Indexed: 05/05/2023] Open
Abstract
The potential of psychedelics to persistently treat substance use disorders is known since the 1960s. However, the biological mechanisms responsible for their therapeutic effects have not yet been fully elucidated. While it is known that serotonergic hallucinogens induce changes in gene expression and neuroplasticity, particularly in prefrontal regions, theories on how specifically this counteracts the alterations that occur in neuronal circuitry throughout the course of addiction are largely unknown. This narrative mini-review endeavors to synthesize well-established knowledge from addiction research with findings and theories regarding the neurobiological effects of psychedelics to give an overview of the potential mechanisms that underlie the treatment of substance use disorders with classical hallucinogenic compounds and point out gaps in the current understanding.
Collapse
Affiliation(s)
- Marvin M. Urban
- Interdisciplinary Center for Neurosciences, University of Heidelberg, Heidelberg, Germany
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, United States
- *Correspondence: Marvin M. Urban,
| | - Moritz R. Stingl
- Interdisciplinary Center for Neurosciences, University of Heidelberg, Heidelberg, Germany
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, United States
| | - Marcus W. Meinhardt
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
18
|
Cole AB, Montgomery K, Bale TL, Thompson SM. What the hippocampus tells the HPA axis: Hippocampal output attenuates acute stress responses via disynaptic inhibition of CRF+ PVN neurons. Neurobiol Stress 2022; 20:100473. [PMID: 35982732 PMCID: PMC9379952 DOI: 10.1016/j.ynstr.2022.100473] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/11/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022] Open
Abstract
The hippocampus exerts inhibitory feedback on the release of glucocorticoids. Because the major hippocampal efferent projections are excitatory, it has been hypothesized that this inhibition is mediated by populations of inhibitory neurons in the hypothalamus or elsewhere. These regions would be excited by hippocampal efferents and project to corticotropin-releasing factor (CRF) cells in the paraventricular nucleus of the hypothalamus (PVN). A direct demonstration of the synaptic responses elicited by hippocampal outputs in PVN cells or upstream GABAergic interneurons has not been provided previously. Here, we used viral vectors to express channelrhodopsin (ChR) and enhanced yellow fluorescent protein (EYFP) in pyramidal cells in the ventral hippocampus (vHip) in mice expressing tdTomato in GABA- or CRF-expressing neurons. We observed dense innervation of the bed nucleus of the stria terminalis (BNST) by labeled vHip axons and sparse labeling within the PVN. Using whole-cell voltage-clamp recording in parasagittal brain slices containing the BNST and PVN, photostimulation of vHip terminals elicited rapid excitatory postsynaptic currents (EPSCs) and longer-latency inhibitory postsynaptic currents (IPSCs) in both CRF+ and GAD + cells. The ratio of synaptic excitation and inhibition was maintained in CRF + cells during 20 Hz stimulus trains. Photostimulation of hippocampal afferents to the BNST and PVN in vivo inhibited the rise in blood glucocorticoid levels produced by acute restraint stress. We thus provide functional evidence suggesting that hippocampal output to the BNST contributes to a net inhibition of the hypothalamic-pituitary axis, providing further mechanistic insights into this process using methods with enhanced spatial and temporal resolution.
Collapse
Affiliation(s)
- Anthony B. Cole
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Medical Scientist Training Program, Departments of University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Kristen Montgomery
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Tracy L. Bale
- Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Psychiatry, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Scott M. Thompson
- Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Psychiatry, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD, USA
| |
Collapse
|
19
|
Vrshek-Schallhorn S, Corneau GM, Grillo AR, Sapuram VR, Plieger T, Reuter M. Additive serotonergic genetic sensitivity and cortisol reactivity to lab-based social evaluative stress: Influence of severity across two samples. Psychoneuroendocrinology 2022; 142:105767. [PMID: 35525123 DOI: 10.1016/j.psyneuen.2022.105767] [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: 09/17/2021] [Revised: 02/28/2022] [Accepted: 04/10/2022] [Indexed: 11/29/2022]
Abstract
Prior work demonstrates that an additive serotonergic multilocus genetic profile score (MGPS) predicts amplified risk for depression following significant life stress, and that it interacts with elevations in the cortisol awakening response to predict depression. The serotonin system and HPA-axis have bidirectional influence, but whether this MGPS predicts acute cortisol reactivity, which might then serve as a mechanism for depression, is unknown. Our prior work suggests that depression risk factors predict blunted cortisol reactivity to explicit negative evaluative lab-based stress. Thus, we hypothesized that a 4-variant serotonergic MGPS (three SNPs from the original 5-variant version plus 5HTTLPR) would predict blunted cortisol reactivity to explicit negative evaluative stress versus a control. In Sample 1, growth curve modeling showed that the MGPS predicted heightened cortisol reactivity (p = 0.0001) in an explicitly negative evaluative Trier Social Stress Test variant (TSST) versus a control condition among non-depressed emerging adults (N = 152; 57% female). In Sample 2, 125 males completed the Socially Evaluative Cold Pressor Test (SECPT), an ambiguously negative evaluative manipulation; findings displayed a similar pattern but did not reach statistical significance (ps.075-.091). A participant-level meta-analysis of the two samples demonstrated a significant effect of negative evaluation severity, such that the MGPS effect size on reactivity increased linearly from control to SECPT to an explicitly negative evaluative TSST. Findings indicate that this MGPS contributes to sensitivity to social threat and that cortisol dysregulation in the context of social stress may be one mechanism by which this MGPS contributes to depression.
Collapse
Affiliation(s)
| | - Gail M Corneau
- Department of Psychology, University of North Carolina at Greensboro, USA
| | | | - Vaibhav R Sapuram
- Department of Psychology, University of North Carolina at Greensboro, USA
| | - Thomas Plieger
- Department of Psychology and Center for Economics & Neuroscience, Bonn University, Germany
| | - Martin Reuter
- Department of Psychology and Center for Economics & Neuroscience, Bonn University, Germany
| |
Collapse
|
20
|
Urien L, Cohen S, Howard S, Yakimov A, Nordlicht R, Bauer EP. Aversive Contexts Reduce Activity in the Ventral Subiculum- BNST Pathway. Neuroscience 2022; 496:129-140. [PMID: 35724771 PMCID: PMC9329270 DOI: 10.1016/j.neuroscience.2022.06.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 06/08/2022] [Accepted: 06/12/2022] [Indexed: 01/25/2023]
Abstract
Many anxiety disorders can be characterized by abnormalities in detecting and learning about threats, and the inability to reduce fear responses in non-threatening environments. PTSD may be the most representative of context processing pathology, as intrusive memories are experienced in "safe" contexts. The ventral subiculum (vSUB), the main output of the ventral hippocampus, encodes environmental cues and is critical for context processing. The bed nucleus of the stria terminalis (BNST) contributes to anxiety-like behaviors as well as context fear conditioning. Given the important roles of the BNST and the vSUB in these anxiety and fear-related behaviors, and the anatomical connections between the two brain regions, the major aims of this study were to characterize the anatomy and function of the vSUB-BNST pathway. First, using the retrograde tracer cholera toxin, we mapped the topographical arrangement of the vSUB-BNST pathway. Dual retrograde tracing experiments revealed neurons projecting to the BNST and those projecting to the basolateral amygdala are distinct populations. Second, we assessed whether activity in this pathway, as indexed by FOS immunohistochemistry, was modulated by context fear conditioning. Our data reveal less activation of the vSUB-BNST pathway in both males and females in aversive contexts and the greatest activation when animals explored a neutral familiar context. In addition, the vSUB of females contained fewer GABAergic neurons compared to males. These findings suggest that the vSUB-BNST pathway is involved in eliciting appropriate responses to contexts.
Collapse
Affiliation(s)
- Louise Urien
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Stacey Cohen
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Sophia Howard
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Alexandrina Yakimov
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Rachel Nordlicht
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Elizabeth P Bauer
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States.
| |
Collapse
|
21
|
Philippe TJ, Bao L, Koblanski ME, Viau V. Sex Differences in Serotonin 5-HT 1A Receptor Responses to Repeated Restraint Stress in Adult Male and Female Rats. Int J Neuropsychopharmacol 2022; 25:863-876. [PMID: 35904324 PMCID: PMC9593217 DOI: 10.1093/ijnp/pyac046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/15/2022] [Accepted: 07/23/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Male and female rats were exposed to repeated restraint to determine how changes in serotonin (5-hydroxytryptamine; 5-HT) 1A receptors associate with stress hypothalamic-pituitary-adrenal (HPA) axis habituation. METHODS In response to 2-hour episodes of restraint, repeated daily for 5 consecutive days, males and females displayed reliable declines in HPA output, indicated by diminished adrenocorticotropin and corticosterone secretory responses. Using the 5-HT 1A receptor agonist 8-hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT) as a pharmacological challenge for inducing hypothermia and elevations in plasma corticosterone, males displayed sensitized hypothermal responses after repeated restraint, whereas corticosterone responses to 8-OH-DPAT were enhanced in both sexes following single or repeated exposure. RESULTS Only males showed elevations in 5-HT 1A receptor G-protein coupling responses in the dorsal raphe after repeated restraint, whereas only females showed an increase in 5-HT 1A receptor responses in the hippocampus following single or repeated exposure. G-protein coupling responses within both regions correlated positively with 5-HT 1A receptor binding capacity. Thus, despite expressing similar capacities for stress HPA axis habituation, males and females emerged from repeated restraint to show region-specific changes in 5-HT 1A receptor function that may be explained, at least in part, by changes in receptor availability. CONCLUSIONS Based on the hypothermal and corticosteroid responses to 8-OH-DPAT, the present data suggest that stress habituation is met by an increase in the sensitivity of presynaptic 5-HT 1A receptors in males and by an increase in the sensitivity of a population of postsynaptic receptors in both sexes.
Collapse
Affiliation(s)
- Tristan J Philippe
- Department of Cellular and Physiological Sciences, Neuroscience Program, University of British Columbia, Vancouver, Canada
| | - Lexia Bao
- Department of Cellular and Physiological Sciences, Neuroscience Program, University of British Columbia, Vancouver, Canada
| | - Maya E Koblanski
- Department of Cellular and Physiological Sciences, Neuroscience Program, University of British Columbia, Vancouver, Canada
| | - Victor Viau
- Correspondence: Victor Viau, PhD, Department of Cellular and Physiological Sciences, Life Sciences Centre, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada ()
| |
Collapse
|
22
|
Kovac M, Vladimirovna Ippolitova T, Pozyabin S, Aliev R, Lobanova V, Drakul N, S. Rutland C. Equine Stress: Neuroendocrine Physiology and Pathophysiology. Vet Med Sci 2022. [DOI: 10.5772/intechopen.105045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
This review presents new aspects to understanding the neuroendocrine regulation of equine stress responses, and their influences on the physiological, pathophysiological, and behavioral processes. Horse management, in essence, is more frequently confirmed by external and internal stress factors, than in other domestic animals. Regardless of the nature of the stimulus, the equine stress response is an effective and highly conservative set of interconnected relationships designed to maintain physiological integrity even in the most challenging circumstances (e.g., orthopedic injuries, abdominal pain, transport, competitions, weaning, surgery, and inflammation). The equine stress response is commonly a complementary homeostatic mechanism that provides protection (not an adaptation) when the body is disturbed or threatened. It activates numerous neural and hormonal networks to optimize metabolic, cardiovascular, musculoskeletal, and immunological functions. This review looks into the various mechanisms involved in stress responses, stress-related diseases, and assessment, prevention or control, and management of these diseases and stress. Stress-related diseases can not only be identified and assessed better, given the latest research and techniques but also prevented or controlled.
Collapse
|
23
|
Aoyama BB, Zanetti GG, Dias EV, Athié MCP, Lopes-Cendes I, Schwambach Vieira A. Transcriptomic analysis of dorsal and ventral subiculum after induction of acute seizures by electric stimulation of the perforant pathway in rats. Hippocampus 2022; 32:436-448. [PMID: 35343006 DOI: 10.1002/hipo.23417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 11/09/2022]
Abstract
Preconditioning is a mechanism in which injuries induced by non-lethal hypoxia or seizures trigger cellular resistance to subsequent events. Norwood et al., in a 2010 study, showed that an 8-h-long period of electrical stimulation of the perforant pathway in rats is required for the induction of hippocampal sclerosis. However, in order to avoid generalized seizures, status epilepticus (SE), and death, a state of resistance to seizures must be induced in the hippocampus by a preconditioning paradigm consisting of two daily 30-min stimulation periods. Due to the importance of the subiculum in the hippocampal formation, this study aims to investigate differential gene expression patterns in the dorsal and ventral subiculum using RNA-sequencing, after induction of a preconditioning protocol by electrical stimulation of the perforant pathway. The dorsal (dSub) and ventral (vSub) subiculum regions were collected by laser-microdissection 24 h after preconditioning protocol induction in rats. RNA sequencing was performed in a Hiseq 4000 platform, reads were aligned using the STAR and DESEq2 statistics package was used to estimate gene expression. We identified 1176 differentially expressed genes comparing control to preconditioned subiculum regions, 204 genes were differentially expressed in dSub and 972 in vSub. The gene ontology enrichment analysis showed that the most significant common enrichment pathway considering up-regulated genes in dSub and vSub was steroid metabolism. In contrast, the most significant enrichment pathway considering down-regulated genes in vSub was axon guidance. Our results indicate that preconditioning induces changes in the expression of genes related to synaptic reorganization, increased cholesterol metabolism, and astrogliosis in both dSub and vSub. Both regions also presented a decrease in the expression of genes related to glutamatergic transmission and an increase in expression of genes related to complement system activation and GABAergic transmission. The down-regulation of proapoptotic and axon guidance genes in the ventral subiculum suggests that preconditioning may induce a neuroprotective environment in this region.
Collapse
Affiliation(s)
- Beatriz B Aoyama
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - Gabriel G Zanetti
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - Elayne V Dias
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - Maria C P Athié
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil.,Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Iscia Lopes-Cendes
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil.,Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - André Schwambach Vieira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| |
Collapse
|
24
|
de Oliveira RP, de Andrade JS, Spina M, Chamon JV, Silva PHD, Werder AK, Ortolani D, Thomaz LDSC, Romariz S, Ribeiro DA, Longo BM, Spadari RC, Viana MDB, Melo-Thomas L, Céspedes IC, da Silva RCB. Clozapine prevented social interaction deficits and reduced c-Fos immunoreactivity expression in several brain areas of rats exposed to acute restraint stress. PLoS One 2022; 17:e0262728. [PMID: 35239670 PMCID: PMC8893644 DOI: 10.1371/journal.pone.0262728] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/04/2022] [Indexed: 11/21/2022] Open
Abstract
In the present study, we evaluate the effect of acute restraint stress (15 min) of male Wistar rats on social interaction measurements and c-Fos immunoreactivity (c-Fos-ir) expression, a marker of neuronal activity, in areas involved with the modulation of acute physical restraint in rats, i.e., the paraventricular nucleus of the hypothalamus (PVN), median raphe nucleus (MnR), medial prefrontal cortex (mPFC), cingulate prefrontal cortex (cPFC), nucleus accumbens (NaC), hippocampus (CA3), lateral septum (LS) and medial amygdala (MeA). We considered the hypothesis that restraint stress exposure could promote social withdrawal induced by the activation of the hypothalamic-pituitary-adrenocortical (HPA) axis, and increase c-Fos expression in these limbic forebrain areas investigated. In addition, we investigated whether pretreatment with the atypical antipsychotic clozapine (5 mg/kg; I.P.) could attenuate or block the effects of restraint on these responses. We found that restraint stress induced social withdrawal, and increased c-Fos-ir in these areas, demonstrating that a single 15 min session of physical restraint of rats effectively activated the HPA axis, representing an effective tool for the investigation of neuronal activity in brain regions sensitive to stress. Conversely, pretreatment with clozapine, prevented social withdrawal and reduced c-Fos expression. We suggest that treatment with clozapine exerted a preventive effect in the social interaction deficit, at least in part, by blocking the effect of restraint stress in brain regions that are known to regulate the HPA-axis, including the cerebral cortex, hippocampus, hypothalamus, septum and amygdala. Further experiments will be done to confirm this hypothesis.
Collapse
Affiliation(s)
| | - José Simões de Andrade
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | - Marianna Spina
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | - João Vítor Chamon
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | | | - Ana Keyla Werder
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | - Daniela Ortolani
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | | | - Simone Romariz
- Departamento de Fisiologia, Universidade Federal de São Paulo (UNIFESP/SP), São Paulo, Brazil
| | - Daniel Araki Ribeiro
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | - Beatriz Monteiro Longo
- Departamento de Fisiologia, Universidade Federal de São Paulo (UNIFESP/SP), São Paulo, Brazil
| | - Regina Célia Spadari
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | - Milena de Barros Viana
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | - Liana Melo-Thomas
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-Universityof Marburg, Marburg, Germany
- Marburg Center for Mind, Brain, and Behavior (MCMBB), Marburg, Marburg, Germany
| | - Isabel Cristina Céspedes
- Departamento de Morfologia e Genética, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Regina Cláudia Barbosa da Silva
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
- Instituto de Neurociências e Comportamento (INeC), Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
25
|
Mei J, Muller E, Ramaswamy S. Informing deep neural networks by multiscale principles of neuromodulatory systems. Trends Neurosci 2022; 45:237-250. [DOI: 10.1016/j.tins.2021.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/04/2021] [Accepted: 12/21/2021] [Indexed: 01/19/2023]
|
26
|
Martins DF, Viseux FJF, Salm DC, Ribeiro ACA, da Silva HKL, Seim LA, Bittencourt EB, Bianco G, Moré AOO, Reed WR, Mazzardo-Martins L. The role of the vagus nerve in fibromyalgia syndrome. Neurosci Biobehav Rev 2021; 131:1136-1149. [PMID: 34710514 DOI: 10.1016/j.neubiorev.2021.10.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 08/08/2021] [Accepted: 10/18/2021] [Indexed: 12/15/2022]
Abstract
Fibromyalgia (FM) syndrome is a common illness characterized by chronic widespread pain, sleep problems, fatigue, and cognitive difficulties. Dysfunctional neurotransmitter systems that influence the body's endogenous stress response systems are thought to underlie many of the major FM-related symptoms. A model of FM pathogenesis suggests biological and psychosocial variables interact to influence the genetic predisposition, but the precise mechanisms remain unclear. The Polyvagal Theory provides a theoretical framework from which to investigate potential biological mechanisms. The vagus nerve (VN) has anti-inflammatory properties via its afferent and efferent fibers. A low vagal tone (as assessed by low heart rate variability), has been observed in painful and inflammatory diseases, including FM, while the ventral branch of the VN is linked to emotional expression and social engagement. These anti-inflammatory and psychological (limbic system) properties of the VN may possess therapeutic potential in treating FM. This review paper summarizes the scientific literature regarding the potential role of the VN in transducing and/or therapeutically managing FM signs and symptoms.
Collapse
Affiliation(s)
- Daniel F Martins
- Experimental Neuroscience Laboratory (LaNEx), Physiotherapy Graduate Course, University of Southern Santa Catarina, Palhoça, SC, Brazil; Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, SC, Brazil.
| | - Frederic J F Viseux
- Laboratoire d'Automatique, de Mécanique et d'Informatique industrielle et Humaine (LAMIH), UMR CNRS 8201, Université Polytechnique des Hauts-de-France, Valenciennes, France; Centre d'Evaluation et de Traitement de la Douleur (CETD), Hôpital Jean Bernard, Centre Hospitalier de Valenciennes, F-59322 Valenciennes, France
| | - Daiana C Salm
- Experimental Neuroscience Laboratory (LaNEx), Physiotherapy Graduate Course, University of Southern Santa Catarina, Palhoça, SC, Brazil; Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, SC, Brazil
| | - Anny Caroline Avelino Ribeiro
- Experimental Neuroscience Laboratory (LaNEx), Physiotherapy Graduate Course, University of Southern Santa Catarina, Palhoça, SC, Brazil
| | - Helen Kassiana Lopes da Silva
- Experimental Neuroscience Laboratory (LaNEx), Physiotherapy Graduate Course, University of Southern Santa Catarina, Palhoça, SC, Brazil
| | - Lynsey A Seim
- Hospital Internal Medicine, 4500 San Pablo Road, Mayo Clinic, Jacksonville, FL, USA
| | | | - Gianluca Bianco
- Research Laboratory of Posturology and Neuromodulation RELPON, Department of Human Neuroscience, Sapienza University, Rome, Italy; Istituto di Formazione in Agopuntura e Neuromodulazione IFAN, Rome, Italy
| | - Ari Ojeda Ocampo Moré
- Integrative Medicine and Acupuncture Service, University Hospital, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - William R Reed
- Department of Physical Therapy, University of Alabama at Birmingham, Birmingham, AL, USA; Rehabilitation Science Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Leidiane Mazzardo-Martins
- Postgraduate Program in Neuroscience, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
27
|
Sobczak F, Pais-Roldán P, Takahashi K, Yu X. Decoding the brain state-dependent relationship between pupil dynamics and resting state fMRI signal fluctuation. eLife 2021; 10:e68980. [PMID: 34463612 PMCID: PMC8460262 DOI: 10.7554/elife.68980] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/27/2021] [Indexed: 01/19/2023] Open
Abstract
Pupil dynamics serve as a physiological indicator of cognitive processes and arousal states of the brain across a diverse range of behavioral experiments. Pupil diameter changes reflect brain state fluctuations driven by neuromodulatory systems. Resting-state fMRI (rs-fMRI) has been used to identify global patterns of neuronal correlation with pupil diameter changes; however, the linkage between distinct brain state-dependent activation patterns of neuromodulatory nuclei with pupil dynamics remains to be explored. Here, we identified four clusters of trials with unique activity patterns related to pupil diameter changes in anesthetized rat brains. Going beyond the typical rs-fMRI correlation analysis with pupil dynamics, we decomposed spatiotemporal patterns of rs-fMRI with principal component analysis (PCA) and characterized the cluster-specific pupil-fMRI relationships by optimizing the PCA component weighting via decoding methods. This work shows that pupil dynamics are tightly coupled with different neuromodulatory centers in different trials, presenting a novel PCA-based decoding method to study the brain state-dependent pupil-fMRI relationship.
Collapse
Affiliation(s)
- Filip Sobczak
- Translational Neuroimaging and Neural Control Group, High Field Magnetic Resonance Department, Max Planck Institute for Biological CyberneticsTübingenGermany
- Graduate Training Centre of Neuroscience, International Max Planck Research School, University of TuebingenTuebingenGermany
| | - Patricia Pais-Roldán
- Translational Neuroimaging and Neural Control Group, High Field Magnetic Resonance Department, Max Planck Institute for Biological CyberneticsTübingenGermany
- Institute of Neuroscience and Medicine 4, Medical Imaging Physics, Forschungszentrum JülichJülichGermany
| | - Kengo Takahashi
- Translational Neuroimaging and Neural Control Group, High Field Magnetic Resonance Department, Max Planck Institute for Biological CyberneticsTübingenGermany
- Graduate Training Centre of Neuroscience, International Max Planck Research School, University of TuebingenTuebingenGermany
| | - Xin Yu
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical SchoolCharlestown, MassachusettsUnited States
| |
Collapse
|
28
|
Vinterstare J, Brönmark C, Nilsson PA, Langerhans RB, Berglund O, Örjes J, Brodin T, Fick J, Hulthén K. Antipredator phenotype in crucian carp altered by a psychoactive drug. Ecol Evol 2021; 11:9435-9446. [PMID: 34306633 PMCID: PMC8293787 DOI: 10.1002/ece3.7762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 01/01/2023] Open
Abstract
Predator-inducible defenses constitute a widespread form of adaptive phenotypic plasticity, and such defenses have recently been suggested linked with the neuroendocrine system. The neuroendocrine system is a target of endocrine disruptors, such as psychoactive pharmaceuticals, which are common aquatic contaminants. We hypothesized that exposure to an antidepressant pollutant, fluoxetine, influences the physiological stress response in our model species, crucian carp, affecting its behavioral and morphological responses to predation threat. We examined short- and long-term effects of fluoxetine and predator exposure on behavior and morphology in crucian carp. Seventeen days of exposure to a high dose of fluoxetine (100 µg/L) resulted in a shyer phenotype, regardless of the presence/absence of a pike predator, but this effect disappeared after long-term exposure. Fluoxetine effects on morphological plasticity were context-dependent as a low dose (1 µg/L) only influenced crucian carp body shape in pike presence. A high dose of fluoxetine strongly influenced body shape regardless of predator treatment. Our results highlight that environmental pollution by pharmaceuticals could disrupt physiological regulation of ecologically important inducible defenses.
Collapse
Affiliation(s)
- Jerker Vinterstare
- Department of BiologyAquatic Ecology Unit, Ecology BuildingLund UniversityLundSweden
| | - Christer Brönmark
- Department of BiologyAquatic Ecology Unit, Ecology BuildingLund UniversityLundSweden
| | - P. Anders Nilsson
- Department of BiologyAquatic Ecology Unit, Ecology BuildingLund UniversityLundSweden
| | - R. Brian Langerhans
- Department of Biological Sciences and W.M. Keck Center for Behavioral BiologyNorth Carolina State UniversityRaleighNCUSA
| | - Olof Berglund
- Department of BiologyAquatic Ecology Unit, Ecology BuildingLund UniversityLundSweden
| | - Jennie Örjes
- Department of BiologyAquatic Ecology Unit, Ecology BuildingLund UniversityLundSweden
| | - Tomas Brodin
- Department of Wildlife, Fish and Environmental StudiesSwedish University of Agricultural Sciences (SLU) – UmeåUmeåSweden
| | - Jerker Fick
- Department of ChemistryUmeå UniversityUmeåSweden
| | - Kaj Hulthén
- Department of BiologyAquatic Ecology Unit, Ecology BuildingLund UniversityLundSweden
| |
Collapse
|
29
|
Stamoula E, Siafis S, Dardalas I, Ainatzoglou A, Matsas A, Athanasiadis T, Sardeli C, Stamoulas K, Papazisis G. Antidepressants on Multiple Sclerosis: A Review of In Vitro and In Vivo Models. Front Immunol 2021; 12:677879. [PMID: 34093579 PMCID: PMC8173210 DOI: 10.3389/fimmu.2021.677879] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/06/2021] [Indexed: 01/03/2023] Open
Abstract
Background Increased prevalence of depression has been observed among patients with multiple sclerosis (MS) and correlated with the elevated levels of proinflammatory cytokines and the overall deregulation of monoaminergic neurotransmitters that these patients exhibit. Antidepressants have proved effective not only in treating depression comorbid to MS, but also in alleviating numerous MS symptoms and even minimizing stress-related relapses. Therefore, these agents could prospectively prove beneficial as a complementary MS therapy. Objective This review aims at illustrating the underlying mechanisms involved in the beneficial clinical effects of antidepressants observed in MS patients. Methods Through a literature search we screened and comparatively assessed papers on the effects of antidepressant use both in vitro and in vivo MS models, taking into account a number of inclusion and exclusion criteria. Results In vitro studies indicated that antidepressants promote neural and glial cell viability and differentiation, reduce proinflammatory cytokines and exert neuroprotective activity by eliminating axonal loss. In vivo studies confirmed that antidepressants delayed disease onset and alleviated symptoms in Experimental Autoimmune Encephalomyelitis (EAE), the most prevalent animal model of MS. Further, antidepressant agents suppressed inflammation and restrained demyelination by decreasing immune cell infiltration of the CNS. Conclusion Antidepressants were efficient in tackling numerous aspects of disease pathophysiology both in vitro and in vivo models. Given that several antidepressants have already proved effective in clinical trials on MS patients, the inclusion of such agents in the therapeutic arsenal of MS should be seriously considered, following an individualized approach to minimize the adverse events of antidepressants in MS patients.
Collapse
Affiliation(s)
- Eleni Stamoula
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Spyridon Siafis
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Dardalas
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alexandra Ainatzoglou
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alkis Matsas
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Chrysanthi Sardeli
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Georgios Papazisis
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
30
|
Rizk MM, Herzog S, Dugad S, Stanley B. Suicide Risk and Addiction: The Impact of Alcohol and Opioid Use Disorders. CURRENT ADDICTION REPORTS 2021; 8:194-207. [PMID: 33747710 PMCID: PMC7955902 DOI: 10.1007/s40429-021-00361-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2021] [Indexed: 01/05/2023]
Abstract
Purpose of Review Suicide is a major public health concern and a leading cause of death in the US. Alcohol and opioid use disorders (AUD/OUD) significantly increase risk for suicidal ideation, attempts, and death, and are the two most frequently implicated substances in suicide risk. We provide a brief overview of shared risk factors and pathways in the pathogenesis of AUD/OUD and suicidal thoughts and behaviors. We also review clinical recommendations on inpatient care, pharmacotherapy, and psychotherapeutic interventions for people with AUD/OUD and co-occurring suicidal ideation and behavior. Recent Findings Among people with an underlying vulnerability to risk-taking and impulsive behaviors, chronic alcohol intoxication can increase maladaptive coping behaviors and hinder self-regulation, thereby increasing the risk of suicide. Additionally, chronic opioid use can result in neurobiological changes that lead to increases in negative affective states, jointly contributing to suicide risk and continued opioid use. Despite significantly elevated suicide risk in individuals with AUD/OUD, there is a dearth of research on pharmacological and psychosocial interventions for co-occurring AUD/OUD and suicidal ideation and behavior. Summary Further research is needed to understand the effects of alcohol and opioid use on suicide risk, as well as address notable gaps in the literature on psychosocial and pharmacological interventions to lower risk for suicide among individuals with AUD/OUD.
Collapse
Affiliation(s)
- Mina M. Rizk
- Department of Psychiatry, Columbia University Irving Medical Center, Columbia University, New York, NY USA
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, 1051 Riverside Dr., Unit 42, New York, NY 10032 USA
- Department of Psychiatry, Faculty of Medicine, Minia University, Egypt, Egypt
| | - Sarah Herzog
- Department of Psychiatry, Columbia University Irving Medical Center, Columbia University, New York, NY USA
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, 1051 Riverside Dr., Unit 42, New York, NY 10032 USA
| | - Sanjana Dugad
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, 1051 Riverside Dr., Unit 42, New York, NY 10032 USA
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Barbara Stanley
- Department of Psychiatry, Columbia University Irving Medical Center, Columbia University, New York, NY USA
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, 1051 Riverside Dr., Unit 42, New York, NY 10032 USA
| |
Collapse
|
31
|
Steinbusch HWM, Dolatkhah MA, Hopkins DA. Anatomical and neurochemical organization of the serotonergic system in the mammalian brain and in particular the involvement of the dorsal raphe nucleus in relation to neurological diseases. PROGRESS IN BRAIN RESEARCH 2021; 261:41-81. [PMID: 33785137 DOI: 10.1016/bs.pbr.2021.02.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The brainstem is a neglected brain area in neurodegenerative diseases, including Alzheimer's and Parkinson's disease, frontotemporal lobar degeneration and autonomic dysfunction. In Depression, several observations have been made in relation to changes in one particular the Dorsal Raphe Nucleus (DRN) which also points toward as key area in various age-related and neurodevelopmental diseases. The DRN is further thought to be related to stress regulated processes and cognitive events. It is involved in neurodegeneration, e.g., amyloid plaques, neurofibrillary tangles, and impaired synaptic transmission in Alzheimer's disease as shown in our autopsy findings. The DRN is a phylogenetically old brain area, with projections that reach out to a large number of regions and nuclei of the central nervous system, particularly in the forebrain. These ascending projections contain multiple neurotransmitters. One of the main reasons for the past and current interest in the DRN is its involvement in depression, and its main transmitter serotonin. The DRN also points toward the increased importance and focus of the brainstem as key area in various age-related and neurodevelopmental diseases. This review describes the morphology, ascending projections and the complex neurotransmitter nature of the DRN, stressing its role as a key research target into the neural bases of depression.
Collapse
Affiliation(s)
- Harry W M Steinbusch
- Department of Cellular Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands; Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology-DGIST, Daegu, South Korea.
| | | | - David A Hopkins
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
32
|
Babicola L, Ventura R, D'Addario SL, Ielpo D, Andolina D, Di Segni M. Long term effects of early life stress on HPA circuit in rodent models. Mol Cell Endocrinol 2021; 521:111125. [PMID: 33333214 DOI: 10.1016/j.mce.2020.111125] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/23/2020] [Accepted: 12/10/2020] [Indexed: 01/06/2023]
Abstract
Adaptation to environmental challenges represents a critical process for survival, requiring the complex integration of information derived from both external cues and internal signals regarding current conditions and previous experiences. The Hypothalamic-pituitary-adrenal axis plays a central role in this process inducing the activation of a neuroendocrine signaling cascade that affects the delicate balance of activity and cross-talk between areas that are involved in sensorial, emotional, and cognitive processing such as the hippocampus, amygdala, Prefrontal Cortex, Ventral Tegmental Area, and dorsal raphe. Early life stress, especially early critical experiences with caregivers, influences the functional and structural organization of these areas, affects these processes in a long-lasting manner and may result in long-term maladaptive and psychopathological outcomes, depending on the complex interaction between genetic and environmental factors. This review summarizes the results of studies that have modeled this early postnatal stress in rodents during the first 2 postnatal weeks, focusing on the long-term effects on molecular and structural alteration in brain areas involved in Hypothalamic-pituitary-adrenal axis function. Moreover, a brief investigation of epigenetic mechanisms and specific genetic targets mediating the long-term effects of these early environmental manipulations and at the basis of differential neurobiological and behavioral effects during adulthood is provided.
Collapse
Affiliation(s)
- Lucy Babicola
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy
| | - Rossella Ventura
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy.
| | - Sebastian Luca D'Addario
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy; Behavioral Neuroscience PhD Programme, Sapienza University, Piazzale Aldo Moro 5, 00184, Rome, Italy
| | - Donald Ielpo
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy; Behavioral Neuroscience PhD Programme, Sapienza University, Piazzale Aldo Moro 5, 00184, Rome, Italy
| | - Diego Andolina
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy
| | - Matteo Di Segni
- IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy.
| |
Collapse
|
33
|
Grizzell JA, Clarity TT, Graham NB, Dulka BN, Cooper MA. Activity of a vmPFC-DRN Pathway Corresponds With Resistance to Acute Social Defeat Stress. Front Neural Circuits 2020; 14:50. [PMID: 33177993 PMCID: PMC7596355 DOI: 10.3389/fncir.2020.00050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 07/13/2020] [Indexed: 11/15/2022] Open
Abstract
The ventromedial prefrontal cortex (vmPFC) plays a critical role in stress resilience through top-down inhibition of key stress-sensitive limbic and hindbrain structures, including the dorsal raphe nucleus (DRN). In a model of experience-dependent stress resistance, socially dominant Syrian hamsters display fewer signs of anxiety following acute social defeat when compared to subordinate or control counterparts. Further, dominants activate vmPFC neurons to a greater degree during stress than do subordinates and become stress-vulnerable following pharmacological inhibition of the vmPFC. Dominants also display fewer stress-activated DRN neurons than subordinates do, suggesting that dominance experience gates activation of vmPFC neurons that inhibit the DRN during social defeat stress. To test whether social dominance alters stress-induced activity of a vmPFC-DRN pathway, we injected a retrograde tracer, cholera toxin B (CTB), into the DRN of dominant, subordinate, and control hamsters and used a dual-label immunohistochemical approach to identify vmPFC neurons co-labeled with CTB and the defeat-induced expression of an immediate early gene, cFos. Results indicate that dominant hamsters display more cFos+ and dual-labeled cells in layers V/VI of infralimbic and prelimbic subregions of the vmPFC compared to other animals. Furthermore, vmPFC-DRN activation corresponded directly with proactive behavioral strategies during defeat, which is indicative of stress resilience. Together, results suggest that recruiting the vmPFC-DRN pathway during acute stress corresponds with resistance to the effects of social defeat in dominant hamsters. Overall, these findings indicate that a monosynaptic vmPFC-DRN pathway can be engaged in an experience-dependent manner, which has implications for behavioral interventions aimed at alleviating stress-related psychopathologies.
Collapse
Affiliation(s)
- J Alex Grizzell
- Department of Psychology, NeuroNET Research Center, The University of Tennessee, Knoxville, Knoxville, TN, United States.,Department of Psychology and Neuroscience, Center for Neuroscience Research, University of Colorado Boulder, Boulder, CO, United States
| | - Thomas T Clarity
- Department of Psychology, NeuroNET Research Center, The University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Nate B Graham
- Department of Psychology, NeuroNET Research Center, The University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Brooke N Dulka
- Department of Psychology, NeuroNET Research Center, The University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Matthew A Cooper
- Department of Psychology, NeuroNET Research Center, The University of Tennessee, Knoxville, Knoxville, TN, United States
| |
Collapse
|
34
|
Zhang SR, Wu JL, Chen H, Luo R, Chen WJ, Tang LJ, Li XW, Yang JM, Gao TM. ErbB4 knockdown in serotonergic neurons in the dorsal raphe induces anxiety-like behaviors. Neuropsychopharmacology 2020; 45:1698-1706. [PMID: 31905370 PMCID: PMC7419508 DOI: 10.1038/s41386-020-0601-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 12/12/2019] [Accepted: 12/17/2019] [Indexed: 12/20/2022]
Abstract
There is a close relationship between serotonergic (5-HT) activity and anxiety. ErbB4, a receptor tyrosine kinase, is expressed in 5-HT neurons. However, whether ErbB4 regulates 5-HT neuronal function and anxiety-related behaviors is unclear. Here, using transgenic and viral approaches, we show that mice with ErbB4 deficiency in 5-HT neurons exhibit heightened anxiety-like behavior and impaired fear extinction, possibly due to an increased excitability of 5-HT neurons in the dorsal raphe nucleus (DRN). Notably, the chemogenetic inhibition of 5-HT neurons in the DRN of ErbB4 mutant mice rescues anxiety-like behaviors. Altogether, our results unravel a previously unknown role of ErbB4 signaling in the regulation of DRN 5-HT neuronal function and anxiety-like behaviors, providing novel insights into the treatment of anxiety disorders.
Collapse
Affiliation(s)
- Sheng-Rong Zhang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jian-Lin Wu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hao Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Rong Luo
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wen-Jun Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Li-Juan Tang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiao-Wen Li
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jian-Ming Yang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
35
|
Fenske L, Concato AC, Vanin AP, Tamagno WA, de Oliveira Sofiatti JR, Treichel H, da Rosa JGS, Barcellos LJG, Kaizer RR. 17-α-Ethinylestradiol modulates endocrine and behavioral responses to stress in zebrafish. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:29341-29351. [PMID: 32440876 DOI: 10.1007/s11356-020-09318-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 05/14/2020] [Indexed: 06/11/2023]
Abstract
The synthetic estrogen, 17-α-ethinylestradiol (EE2), present in contraceptive pills, is an endocrine-disrupting chemical (EDC) that can be found in the aquatic environment. We examined the impacts of EE2 on zebrafish behavioral and physiological responses through the novel tank test (NTT), which measures anxiety-like behavior; the mirror-induced aggression (MIA) test, which measures aggressiveness; and the social preference test (SPT), which measures social cohesion. The steroid hormone levels were also measured. Here, we show that exposure to EE2 impairs stress responses by regulating the levels of specific hormones and eliciting an anxiolytic response, increasing aggression, and reducing social preference in zebrafish. In nature, these changes in behavior compromise reproduction and anti-predator behaviors, which, in turn, affects species survival. The maintenance of an intact behavioral repertoire in zebrafish is essential for their survival. Thus, our results point to the danger of environmental contamination with EE2 as it may alter the dynamics of the prey-predator relationship.
Collapse
Affiliation(s)
- Lurian Fenske
- Programa de Pós-Graduação em Ciência e Tecnologia Ambiental, Universidade Federal da Fronteira Sul, Rodovia RS 135, Km 72, Erechim, RS, 99700-970, Brazil
| | - Ani Carla Concato
- Programa de Pós-Graduação em Ciência e Tecnologia Ambiental, Universidade Federal da Fronteira Sul, Rodovia RS 135, Km 72, Erechim, RS, 99700-970, Brazil
- Instituto Federal de Educação, Ciência e Tecnologia do Rio Grande do Sul, Campus Sertão, Rodovia RS 135, Km 25, Sertão, RS, 99170-000, Brazil
| | - Ana Paula Vanin
- Instituto Federal de Educação, Ciência e Tecnologia do Rio Grande do Sul, Campus Sertão, Rodovia RS 135, Km 25, Sertão, RS, 99170-000, Brazil
| | - Wagner Antonio Tamagno
- Instituto Federal de Educação, Ciência e Tecnologia do Rio Grande do Sul, Campus Sertão, Rodovia RS 135, Km 25, Sertão, RS, 99170-000, Brazil
| | - Jéssica Reis de Oliveira Sofiatti
- Programa de Pós-Graduação em Ciência e Tecnologia Ambiental, Universidade Federal da Fronteira Sul, Rodovia RS 135, Km 72, Erechim, RS, 99700-970, Brazil
| | - Helen Treichel
- Programa de Pós-Graduação em Ciência e Tecnologia Ambiental, Universidade Federal da Fronteira Sul, Rodovia RS 135, Km 72, Erechim, RS, 99700-970, Brazil
| | | | - Leonardo José Gil Barcellos
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
- Curso de Medicina Veterinária, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
- Programa de Pós-Graduação em Bioexperimentação, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
- Programa de Pós-Graduação em Ciências Ambientais, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - Rosilene R Kaizer
- Programa de Pós-Graduação em Ciência e Tecnologia Ambiental, Universidade Federal da Fronteira Sul, Rodovia RS 135, Km 72, Erechim, RS, 99700-970, Brazil.
- Instituto Federal de Educação, Ciência e Tecnologia do Rio Grande do Sul, Campus Sertão, Rodovia RS 135, Km 25, Sertão, RS, 99170-000, Brazil.
| |
Collapse
|
36
|
Abstract
The brain serotonin systems participate in numerous aspects of reward processing, although it remains elusive how exactly serotonin signals regulate neural computation and reward-related behavior. The application of optogenetics and imaging techniques during the last decade has provided many insights. Here, we review recent progress on the organization and physiology of the dorsal raphe serotonin neurons and the relationships between their activity and behavioral functions in the context of reward processing. We also discuss several interesting theories on serotonin's function and how these theories may be reconciled by the possibility that serotonin, acting in synergy with coreleased glutamate, tracks and calculates the so-called beneficialness of the current state to guide an animal's behavior in dynamic environments.
Collapse
Affiliation(s)
- Zhixiang Liu
- National Institute of Biological Sciences, Beijing 102206, China
| | - Rui Lin
- National Institute of Biological Sciences, Beijing 102206, China
| | - Minmin Luo
- National Institute of Biological Sciences, Beijing 102206, China
- School of Life Sciences, Tsinghua University, Beijing 100081, China
- Chinese Institute for Brain Research, Beijing 102206, China
| |
Collapse
|
37
|
Gorlova A, Ortega G, Waider J, Bazhenova N, Veniaminova E, Proshin A, Kalueff AV, Anthony DC, Lesch KP, Strekalova T. Stress-induced aggression in heterozygous TPH2 mutant mice is associated with alterations in serotonin turnover and expression of 5-HT6 and AMPA subunit 2A receptors. J Affect Disord 2020; 272:440-451. [PMID: 32553388 DOI: 10.1016/j.jad.2020.04.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/31/2020] [Accepted: 04/17/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND The contribution of gene-environment interactions that lead to excessive aggression is poorly understood. Environmental stressors and mutations of the gene encoding tryptophan hydroxylase-2 (TPH2) are known to influence aggression. For example, TPH2 null mutant mice (Tph2-/-) are naturally highly aggressive, while heterozygous mice (Tph2+/-) lack a behavioral phenotype and are considered endophenotypically normal. Here we sought to discover whether an environmental stressor would affect the phenotype of the genetically 'susceptible' heterozygous mice (Tph2+/-). METHODS Tph2+/- male mice or Tph2+/+ controls were subjected to a five-day long rat exposure stress paradigm. Brain serotonin metabolism and the expression of selected genes encoding serotonin receptors, AMPA receptors, and stress markers were studied. RESULTS Stressed Tph2+/- mice displayed increased levels of aggression and social dominance, whereas Tph2+/+ animals became less aggressive and less dominant. Brain tissue concentrations of serotonin, its precursor hydroxytryptophan and its metabolite 5-hydroxyindoleacetic acid were significantly altered in all groups in the prefrontal cortex, striatum, amygdala, hippocampus and dorsal raphe after stress. Compared to non-stressed animals, the concentration of 5-hydroxytryptophan was elevated in the amygdala though decreased in the other brain structures. The overexpression of the AMPA receptor subunit, GluA2, and downregulation of 5-HT6 receptor, as well as overexpression of c-fos and glycogen-synthase-kinase-3β (GSK3-β), were found in most structures of the stressed Tph2+/- mice. LIMITATIONS Rescue experiments would help to verify causal relationships of reported changes. CONCLUSIONS The interaction of a partial TPH2 gene deficit with stress results in pathological aggression and molecular changes, and suggests that the presence of genetic susceptibility can augment aggression in seemingly resistant phenotypes.
Collapse
Affiliation(s)
- Anna Gorlova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| | - Gabriela Ortega
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Jonas Waider
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Natalia Bazhenova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands; Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Ekaterina Veniaminova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| | - Andrey Proshin
- PK Anokhin Research Institute of Normal Physiology, Moscow
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China with Ural Federal University, Ekaterinburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University and Almazov Medical Research Center, Institute of Experimental Medicine, St. Petersburg Russia
| | - Daniel C Anthony
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Department of Pharmacology, Oxford University, Oxford, United Kingdom
| | - Klaus-Peter Lesch
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands; Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Tatyana Strekalova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands; Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany; Institute of General Pathology and Pathophysiology, Moscow, Russia.
| |
Collapse
|
38
|
Serotonergic innervation of the auditory midbrain: dorsal raphe subregions differentially project to the auditory midbrain in male and female mice. Brain Struct Funct 2020; 225:1855-1871. [DOI: 10.1007/s00429-020-02098-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 06/06/2020] [Indexed: 01/12/2023]
|
39
|
Ho TC, Gutman B, Pozzi E, Grabe HJ, Hosten N, Wittfeld K, Völzke H, Baune B, Dannlowski U, Förster K, Grotegerd D, Redlich R, Jansen A, Kircher T, Krug A, Meinert S, Nenadic I, Opel N, Dinga R, Veltman DJ, Schnell K, Veer I, Walter H, Gotlib IH, Sacchet MD, Aleman A, Groenewold NA, Stein DJ, Li M, Walter M, Ching CRK, Jahanshad N, Ragothaman A, Isaev D, Zavaliangos‐Petropulu A, Thompson PM, Sämann PG, Schmaal L. Subcortical shape alterations in major depressive disorder: Findings from the ENIGMA major depressive disorder working group. Hum Brain Mapp 2020; 43:341-351. [PMID: 32198905 PMCID: PMC8675412 DOI: 10.1002/hbm.24988] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/01/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
Alterations in regional subcortical brain volumes have been investigated as part of the efforts of an international consortium, ENIGMA, to identify reliable neural correlates of major depressive disorder (MDD). Given that subcortical structures are comprised of distinct subfields, we sought to build significantly from prior work by precisely mapping localized MDD‐related differences in subcortical regions using shape analysis. In this meta‐analysis of subcortical shape from the ENIGMA‐MDD working group, we compared 1,781 patients with MDD and 2,953 healthy controls (CTL) on individual measures of shape metrics (thickness and surface area) on the surface of seven bilateral subcortical structures: nucleus accumbens, amygdala, caudate, hippocampus, pallidum, putamen, and thalamus. Harmonized data processing and statistical analyses were conducted locally at each site, and findings were aggregated by meta‐analysis. Relative to CTL, patients with adolescent‐onset MDD (≤ 21 years) had lower thickness and surface area of the subiculum, cornu ammonis (CA) 1 of the hippocampus and basolateral amygdala (Cohen's d = −0.164 to −0.180). Relative to first‐episode MDD, recurrent MDD patients had lower thickness and surface area in the CA1 of the hippocampus and the basolateral amygdala (Cohen's d = −0.173 to −0.184). Our results suggest that previously reported MDD‐associated volumetric differences may be localized to specific subfields of these structures that have been shown to be sensitive to the effects of stress, with important implications for mapping treatments to patients based on specific neural targets and key clinical features.
Collapse
Affiliation(s)
- Tiffany C. Ho
- Department of Psychiatry & Weill Institute for Neurosciences San Francisco California USA
- Department of Psychiatry & Behavioral Sciences Stanford University Stanford California USA
- Department of Psychology Stanford University Stanford California USA
| | - Boris Gutman
- Department of Biomedical Engineering Illinois Institute of Technology Chicago Illinois USA
| | - Elena Pozzi
- Melbourne Neuropsychiatry Centre, Department of Psychiatry The University of Melbourne & Melbourne Health Melbourne Australia
- Orygen, The National Centre of Excellence in Youth Mental Health Parkville Australia
| | - Hans J. Grabe
- Department of Psychiatry and Psychotherapy University Medicine Greifswald Germany
- German Centre of Neurodegenerative Diseases (DZNE) site Greifswald/Rostock Germany
| | - Norbert Hosten
- Department of Diagnostic Radiology and Neuroradiology University Medicine Greifswald Germany
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy University Medicine Greifswald Germany
- German Centre of Neurodegenerative Diseases (DZNE) site Greifswald/Rostock Germany
| | - Henry Völzke
- Institute for Community Medicine University Medicine Greifswald Germany
| | - Bernhard Baune
- Department of Psychiatry University of Münster Münster Germany
- Department of Psychiatry, Melbourne Medical School The University of Melbourne Melbourne Australia
| | - Udo Dannlowski
- Department of Psychiatry University of Münster Münster Germany
| | | | | | - Ronny Redlich
- Department of Psychiatry University of Münster Münster Germany
| | - Andreas Jansen
- Department of Psychiatry Philipps‐University Marburg Germany
| | - Tilo Kircher
- Department of Psychiatry Philipps‐University Marburg Germany
| | - Axel Krug
- Department of Psychiatry Philipps‐University Marburg Germany
| | - Susanne Meinert
- Department of Psychiatry University of Münster Münster Germany
| | - Igor Nenadic
- Department of Psychiatry Philipps‐University Marburg Germany
| | - Nils Opel
- Department of Psychiatry University of Münster Münster Germany
| | - Richard Dinga
- Department of Psychiatry, Amsterdam University Medical Centers VU University Medical Center, GGZ inGeest, Amsterdam Neuroscience Amsterdam The Netherlands
| | - Dick J. Veltman
- Department of Psychiatry, Amsterdam University Medical Centers VU University Medical Center, GGZ inGeest, Amsterdam Neuroscience Amsterdam The Netherlands
| | - Knut Schnell
- Department of Psychiatry and Psychotherapy University Medical Center Göttingen Göttingen Germany
| | - Ilya Veer
- Division of Mind and Brain Research, Department of Psychiatry and Psychotherapy CCM, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
| | - Henrik Walter
- Division of Mind and Brain Research, Department of Psychiatry and Psychotherapy CCM, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
| | - Ian H. Gotlib
- Department of Psychology Stanford University Stanford California USA
| | - Matthew D. Sacchet
- Department of Psychiatry & Behavioral Sciences Stanford University Stanford California USA
- McLean Hospital and Department of Psychiatry Harvard Medical School Belmont Massachusetts USA
| | - André Aleman
- University of Groningen, University Medical Center Groningen, Department of Neuroscience Groningen The Netherlands
| | - Nynke A. Groenewold
- University of Groningen, University Medical Center Groningen, Department of Neuroscience Groningen The Netherlands
- University of Groningen, University Medical Center Groningen Interdisciplinary Center Psychopathology and Emotion Regulation (ICPE) Groningen The Netherlands
| | - Dan J. Stein
- Department of Psychiatry and Mental Health University of Cape Town South Africa
| | - Meng Li
- Max Planck Institute for Biological Cybernetics Tuebingen Germany
| | - Martin Walter
- Department of Psychiatry University Tuebingen Germany
| | - Christopher R. K. Ching
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute Keck USC School of Medicine California USA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute Keck USC School of Medicine California USA
| | - Anjanibhargavi Ragothaman
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute Keck USC School of Medicine California USA
| | - Dmitry Isaev
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute Keck USC School of Medicine California USA
| | - Artemis Zavaliangos‐Petropulu
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute Keck USC School of Medicine California USA
| | - Paul M. Thompson
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute Keck USC School of Medicine California USA
| | | | - Lianne Schmaal
- Orygen, The National Centre of Excellence in Youth Mental Health Parkville Australia
- Centre for Youth Mental Health The University of Melbourne Melbourne Australia
| |
Collapse
|
40
|
Abstract
Neurons that synthesize and release 5-hydroxytryptamine (5-HT; serotonin) express a core set of genes that establish and maintain this neurotransmitter phenotype and distinguish these neurons from other brain cells. Beyond a shared 5-HTergic phenotype, these neurons display divergent cellular properties in relation to anatomy, morphology, hodology, electrophysiology and gene expression, including differential expression of molecules supporting co-transmission of additional neurotransmitters. This diversity suggests that functionally heterogeneous subtypes of 5-HT neurons exist, but linking subsets of these neurons to particular functions has been technically challenging. We discuss recent data from molecular genetic, genomic and functional methods that, when coupled with classical findings, yield a reframing of the 5-HT neuronal system as a conglomeration of diverse subsystems with potential to inspire novel, more targeted therapies for clinically distinct 5-HT-related disorders.
Collapse
|
41
|
Ma L, Hu P, Zhang J, Cui W, Zhao X. Purpurin exerted antidepressant-like effects on behavior and stress axis reactivity: evidence of serotonergic engagement. Psychopharmacology (Berl) 2020; 237:887-899. [PMID: 31900524 DOI: 10.1007/s00213-019-05422-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 12/03/2019] [Indexed: 12/25/2022]
Abstract
RATIONALE AND OBJECTIVES Major depression represents a significant public health problem worldwide, and effective regimen is lacking. The present study investigated the antidepressant-like effects of purpurin, a natural anthraquinone compound from Rubia tinctorum L., and explored the underlying mechanism(s). METHODS Forced swim test (FST) and tail suspension test (TST) were used to assess antidepressant-like effects of purpurin in mice. Effects of purpurin on neuroendocrine responsivity were evaluated at the level of corticosterone and ACTH following acute restraint stress and intracerebroventricular injection of corticotrophin-releasing-factor (CRF). Serotonergic mechanisms underlying purpurin antidepressant effect were explored using biochemical, neurochemical, and pharmacological paradigms. RESULTS Chronic purpurin treatment exerted in mice dose-dependently antidepressant-like effects on behavior and stress axis reactivity (n = 9-11 per group). The purpurin-triggered antidepressant-like effects are serotonergically dependent, since purpurin-treated mice showed escalated levels of brain serotonin and suppressed monoamine oxidase (MAO) activity (n = 8-11 per group). Consistently, chemical depletion of brain serotonin by p-chlorophenylalanine (PCPA) abolished the antidepressant-like effects of purpurin on behavior and stress axis responsivity (n = 9-10 per group). Moreover, the antidepressant effect by purpurin was preferentially counteracted by 1A-selective 5-HT receptor antagonist WAY-100635, but potentiated by 1A-selective agonist 8-OH-DPAT and sub-effective dose of serotonergic antidepressant fluoxetine (n = 9-11 per group), suggesting a crucial role for 5-HT1A related serotonergic system in mediating such purpurin antidepressant effect. CONCLUSION We have revealed the antidepressant-like effects of purpurin on both behavior and stress axis reactivity in mice, with serotonergic system that preferentially couples with 5-HT1A receptors being critically engaged.
Collapse
Affiliation(s)
- Li Ma
- Department of Neurology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, Hubei Province, China
| | - Pei Hu
- Department of Vasculocardiology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, Hubei Province, China
| | - Junfang Zhang
- Department of Pharmacology, Ningbo University, School of Medical Science, Ningbo, China
| | - Wugeng Cui
- Department of Pharmacology, Ningbo University, School of Medical Science, Ningbo, China.
| | - Xin Zhao
- Department of Pharmacology, Ningbo University, School of Medical Science, Ningbo, China.
| |
Collapse
|
42
|
Bajgarova Z, Bajgar A. The relationships among MAOA, COMT Val158Met, and 5-HTTLPR polymorphisms, newborn stress reactivity, and infant temperament. Brain Behav 2020; 10:e01511. [PMID: 31884721 PMCID: PMC7010585 DOI: 10.1002/brb3.1511] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/23/2019] [Accepted: 11/27/2019] [Indexed: 01/23/2023] Open
Abstract
INTRODUCTION Variance in hypothalamic-pituitary-adrenal (HPA) axis reactivity is considered to be one of the sources of differences in infant temperament. The cortisol enters into interactions with dopamine and serotonin, so it is expected that polymorphisms in genes coding monoamine metabolism influence both HPA axis reactivity and temperament. METHODS We therefore explore the relationship among 5-HTTLPR S/L, MAOA H/L, and COMT Val158Met polymorphisms, the stress reaction of newborn infants after a heel stick blood draw (measured by determining salivary cortisol at three time points), and temperament assessed at the age of 3 months using Rothbart's Infant Behavior Questionnaire-Revised (IBQ-R) with a sample of 84 infants. RESULTS The decrease in the salivary cortisol correlated with nine primary scales and all three secondary scales of IBQ-R. Children with a greater cortisol decrease were assessed as less susceptible to negative emotions, more extraverted, and more regulated. The polymorphisms that were observed were related both to the course of the stress reaction and to temperament. The 5-HTTLPR S allele was connected to higher scores for Negative Emotionality and lower scores for Orienting/Regulatory Capacity. The presence of the MAOA L allele predisposed its carriers to higher scores for Negative Emotionality, lower scores for Orienting/Regulatory Capacity, and a lower decrease in cortisol. The Met allele of COMT Val158Met polymorphism was connected to a higher Positive Affectivity/Surgency and Orienting/Regulatory Capacity and a greater cortisol decrease. CONCLUSIONS Contrary to previous studies referring mainly basal cortisol and its increase, the results of our study emphasize the importance of cortisol elimination in infant temperament. Another interesting finding was a higher cortisol increase, higher Distress to Limitations, Negative Emotionality, and Approach in MAOA LL homozygotes which are traditionally understood as more vulnerable toward early stress in developing later externalizing behavior.
Collapse
Affiliation(s)
- Zdenka Bajgarova
- Department of Pedagogy and Psychology, Faculty of Education, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Adam Bajgar
- Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic
| |
Collapse
|
43
|
Agrawal L, Korkutata M, Vimal SK, Yadav MK, Bhattacharyya S, Shiga T. Therapeutic potential of serotonin 4 receptor for chronic depression and its associated comorbidity in the gut. Neuropharmacology 2020; 166:107969. [PMID: 31982703 DOI: 10.1016/j.neuropharm.2020.107969] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/19/2022]
Abstract
The latest estimates from world health organization suggest that more than 450 million people are suffering from depression and other psychiatric conditions. Of these, 50-60% have been reported to have progression of gut diseases. In the last two decades, researchers introduced incipient physiological roles for serotonin (5-HT) receptors (5-HTRs), suggesting their importance as a potential pharmacological target in various psychiatric and gut diseases. A growing body of evidence suggests that 5-HT systems affect the brain-gut axis in depressive patients, which leads to gut comorbidity. Recently, preclinical trials of 5-HT4R agonists and antagonists were promising as antipsychotic and prokinetic agents. In the current review, we address the possible pharmacological role and contribution of 5-HT4R in the pathophysiology of chronic depression and associated gut abnormalities. Physiologically, during depression episodes, centers of the sympathetic and parasympathetic nervous system couple together with neuroendocrine systems to alter the function of hypothalamic-pituitary-adrenal (HPA) axis and enteric nervous system (ENS), which in turn leads to onset of gastrointestinal tract (GIT) disorders. Consecutively, the ENS governs a broad spectrum of physiological activities of gut, such as visceral pain and motility. During the stages of emotional stress, hyperactivity of the HPA axis alters the ENS response to physiological and noxious stimuli. Consecutively, stress-induced flare, swelling, hyperalgesia and altered reflexes in gut eventually lead to GIT disorders. In summary, the current review provides prospective information about the role and mechanism of 5-HT4R-based therapeutics for the treatment of depressive disorder and possible consequences for the gut via brain-gut axis interactions. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.
Collapse
Affiliation(s)
- Lokesh Agrawal
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1, 305-8577, Tennodai, Tsukuba, Ibaraki, Japan.
| | - Mustafa Korkutata
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Sunil Kumar Vimal
- Department of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Manoj Kumar Yadav
- School of Integrative and Global Majors, University of Tsukuba, 1-1-1, 305-8577, Tennodai, Tsukuba, Ibaraki, Japan; Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Sanjib Bhattacharyya
- Department of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Takashi Shiga
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1, 305-8577, Tennodai, Tsukuba, Ibaraki, Japan; Department of Neurobiology, Faculty of Medicine, University of Tsukuba,1-1-1, Tennodai, Tsukuba, 305-8577, Ibaraki, Japan.
| |
Collapse
|
44
|
Metzger M, Souza R, Lima LB, Bueno D, Gonçalves L, Sego C, Donato J, Shammah-Lagnado SJ. Habenular connections with the dopaminergic and serotonergic system and their role in stress-related psychiatric disorders. Eur J Neurosci 2019; 53:65-88. [PMID: 31833616 DOI: 10.1111/ejn.14647] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/28/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022]
Abstract
The habenula (Hb) is a phylogenetically old epithalamic structure differentiated into two nuclear complexes, the medial (MHb) and lateral habenula (LHb). After decades of search for a great unifying function, interest in the Hb resurged when it was demonstrated that LHb plays a major role in the encoding of aversive stimuli ranging from noxious stimuli to the loss of predicted rewards. Consistent with a role as an anti-reward center, aberrant LHb activity has now been identified as a key factor in the pathogenesis of major depressive disorder. Moreover, both MHb and LHb emerged as new players in the reward circuitry by primarily mediating the aversive properties of distinct drugs of abuse. Anatomically, the Hb serves as a bridge that links basal forebrain structures with monoaminergic nuclei in the mid- and hindbrain. So far, research on Hb has focused on the role of the LHb in regulating midbrain dopamine release. However, LHb/MHb are also interconnected with the dorsal (DR) and median (MnR) raphe nucleus. Hence, it is conceivable that some of the habenular functions are at least partly mediated by the complex network that links MHb/LHb with pontomesencephalic monoaminergic nuclei. Here, we summarize research about the topography and transmitter phenotype of the reciprocal connections between the LHb and ventral tegmental area-nigra complex, as well as those between the LHb and DR/MnR. Indirect MHb outputs via interpeduncular nucleus to state-setting neuromodulatory networks will also be commented. Finally, we discuss the role of specific LHb-VTA and LHb/MHb-raphe circuits in anxiety and depression.
Collapse
Affiliation(s)
- Martin Metzger
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rudieri Souza
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Leandro B Lima
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Debora Bueno
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luciano Gonçalves
- Department of Human Anatomy, Federal University of the Triângulo Mineiro, Uberaba, Brazil
| | - Chemutai Sego
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jose Donato
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sara J Shammah-Lagnado
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
45
|
Uribe-Mariño A, Castiblanco-Urbina MA, Falconi-Sobrinho LL, Dos Anjos-Garcia T, de Oliveira RC, Mendes-Gomes J, da Silva Soares R, Matthiesen M, Almada RC, de Oliveira R, Coimbra NC. The alpha- and beta-noradrenergic receptors blockade in the dorsal raphe nucleus impairs the panic-like response elaborated by medial hypothalamus neurons. Brain Res 2019; 1725:146468. [PMID: 31541642 DOI: 10.1016/j.brainres.2019.146468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/13/2019] [Accepted: 09/17/2019] [Indexed: 12/16/2022]
Abstract
Dorsal raphe nucleus (DRN) neurons are reciprocally connected to the locus coeruleus (LC) and send neural pathways to the medial hypothalamus (MH). The aim of this work was to investigate whether the blockade of α1-, α2- or β-noradrenergic receptors in the DRN or the inactivation of noradrenergic neurons in the LC modify defensive behaviours organised by MH neurons. For this purpose, Wistar male rats received microinjections of WB4101, RX821002, propranolol (α1-, α2- and β-noradrenergic receptor antagonists, respectively) or physiological saline in the DRN, followed 10 min later by MH GABAA receptor blockade. Other groups of animals received DSP-4 (a noradrenergic neurotoxin), physiological saline or only a needle insertion (sham group) into the LC, and 5 days later, bicuculline or physiological saline was administered in the MH. In all these cases, after MH treatment, the frequency and duration of defensive responses were recorded over 15 min. An anterograde neural tract tracer was also deposited in the DRN. DRN neurons send pathways to lateral and dorsomedial hypothalamus. Blockade of α1- and β-noradrenergic receptors in the DRN decreased escape reactions elicited by bicuculline microinjections in the MH. In addition, a significant increase in anxiety-like behaviours was observed after the blockade of α2-noradrenergic receptors in the DRN. LC pretreatment with DSP-4 decreased both anxiety- and panic attack-like behaviours evoked by GABAA receptor blockade in the MH. In summary, the present findings suggest that the norepinephrine-mediated system modulates defensive reactions organised by MH neurons at least in part via noradrenergic receptors recruitment on DRN neurons.
Collapse
Affiliation(s)
- Andrés Uribe-Mariño
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstraße 2-10, 80804 Munich, Germany
| | - Maria Angélica Castiblanco-Urbina
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; Institute of Developmental Genetics, Helmholtz-Zentrum München-German Research Centre for Environmental Health (GmbH), Ingolstaedter Landstrasse 1, Neuherberg, Germany
| | - Luiz Luciano Falconi-Sobrinho
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; Behavioural Neurosciences Institute (INeC), Avenida do Café, 2450, Ribeirão Preto 14220-030, São Paulo, Brazil
| | - Tayllon Dos Anjos-Garcia
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; Behavioural Neurosciences Institute (INeC), Avenida do Café, 2450, Ribeirão Preto 14220-030, São Paulo, Brazil
| | - Rithiele Cristina de Oliveira
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; Behavioural Neurosciences Institute (INeC), Avenida do Café, 2450, Ribeirão Preto 14220-030, São Paulo, Brazil
| | - Joyce Mendes-Gomes
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; Behavioural Neurosciences Institute (INeC), Avenida do Café, 2450, Ribeirão Preto 14220-030, São Paulo, Brazil
| | - Raimundo da Silva Soares
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil
| | - Melina Matthiesen
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil
| | - Rafael Carvalho Almada
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; Behavioural Neurosciences Institute (INeC), Avenida do Café, 2450, Ribeirão Preto 14220-030, São Paulo, Brazil
| | - Ricardo de Oliveira
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; Behavioural Neurosciences Institute (INeC), Avenida do Café, 2450, Ribeirão Preto 14220-030, São Paulo, Brazil; Health Sciences Institute, Mato Grosso Federal University Medical School (UFMT), Av. Alexandre Ferronato, 1200, Reserva 35, Setor industrial, 78550-000 Sinop, Mato Grosso, Brazil
| | - Norberto Cysne Coimbra
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto 14049-900, São Paulo, Brazil; Behavioural Neurosciences Institute (INeC), Avenida do Café, 2450, Ribeirão Preto 14220-030, São Paulo, Brazil.
| |
Collapse
|
46
|
Huang KW, Ochandarena NE, Philson AC, Hyun M, Birnbaum JE, Cicconet M, Sabatini BL. Molecular and anatomical organization of the dorsal raphe nucleus. eLife 2019; 8:e46464. [PMID: 31411560 PMCID: PMC6726424 DOI: 10.7554/elife.46464] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 08/13/2019] [Indexed: 12/20/2022] Open
Abstract
The dorsal raphe nucleus (DRN) is an important source of neuromodulators and has been implicated in a wide variety of behavioral and neurological disorders. The DRN is subdivided into distinct anatomical subregions comprised of multiple cell types, and its complex cellular organization has impeded efforts to investigate the distinct circuit and behavioral functions of its subdomains. Here we used single-cell RNA sequencing, in situ hybridization, anatomical tracing, and spatial correlation analysis to map the transcriptional and spatial profiles of cells from the mouse DRN. Our analysis of 39,411 single-cell transcriptomes revealed at least 18 distinct neuron subtypes and 5 serotonergic neuron subtypes with distinct molecular and anatomical properties, including a serotonergic neuron subtype that preferentially innervates the basal ganglia. Our study lays out the molecular organization of distinct serotonergic and non-serotonergic subsystems, and will facilitate the design of strategies for further dissection of the DRN and its diverse functions.
Collapse
Affiliation(s)
- Kee Wui Huang
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Nicole E Ochandarena
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Adrienne C Philson
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Minsuk Hyun
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Jaclyn E Birnbaum
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Marcelo Cicconet
- Image and Data Analysis CoreHarvard Medical SchoolBostonUnited States
| | - Bernardo L Sabatini
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
47
|
O’Mara SM, Aggleton JP. Space and Memory (Far) Beyond the Hippocampus: Many Subcortical Structures Also Support Cognitive Mapping and Mnemonic Processing. Front Neural Circuits 2019; 13:52. [PMID: 31447653 PMCID: PMC6692652 DOI: 10.3389/fncir.2019.00052] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/22/2019] [Indexed: 11/13/2022] Open
Abstract
Memory research remains focused on just a few brain structures-in particular, the hippocampal formation (the hippocampus and entorhinal cortex). Three key discoveries promote this continued focus: the striking demonstrations of enduring anterograde amnesia after bilateral hippocampal damage; the realization that synapses in the hippocampal formation are plastic e.g., when responding to short bursts of patterned stimulation ("long-term potentiation" or LTP); and the discovery of a panoply of spatially-tuned cells, principally surveyed in the hippocampal formation (place cells coding for position; head-direction cells, providing compass-like information; and grid cells, providing a metric for 3D space). Recent anatomical, behavioral, and electrophysiological work extends this picture to a growing network of subcortical brain structures, including the anterior thalamic nuclei, rostral midline thalamic nuclei, and the claustrum. There are, for example, spatially-tuned cells in all of these regions, including cells with properties similar to place cells of the hippocampus proper. These findings add new perspectives to what had been originally been proposed-but often overlooked-half a century ago: that damage to an extended network of structures connected to the hippocampal formation results in diencephalic amnesia. We suggest these new findings extend spatial signaling in the brain far beyond the hippocampal formation, with profound implications for theories of the neural bases of spatial and mnemonic functions.
Collapse
Affiliation(s)
- Shane M. O’Mara
- School of Psychology and Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - John P. Aggleton
- School of Psychology, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
48
|
Morris G, Berk M, Maes M, Carvalho AF, Puri BK. Socioeconomic Deprivation, Adverse Childhood Experiences and Medical Disorders in Adulthood: Mechanisms and Associations. Mol Neurobiol 2019; 56:5866-5890. [PMID: 30685844 PMCID: PMC6614134 DOI: 10.1007/s12035-019-1498-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 01/15/2019] [Indexed: 12/30/2022]
Abstract
Severe socioeconomic deprivation (SED) and adverse childhood experiences (ACE) are significantly associated with the development in adulthood of (i) enhanced inflammatory status and/or hypothalamic-pituitary-adrenal (HPA) axis dysfunction and (ii) neurological, neuroprogressive, inflammatory and autoimmune diseases. The mechanisms by which these associations take place are detailed. The two sets of consequences are themselves strongly associated, with the first set likely contributing to the second. Mechanisms enabling bidirectional communication between the immune system and the brain are described, including complex signalling pathways facilitated by factors at the level of immune cells. Also detailed are mechanisms underpinning the association between SED, ACE and the genesis of peripheral inflammation, including epigenetic changes to immune system-related gene expression. The duration and magnitude of inflammatory responses can be influenced by genetic factors, including single nucleotide polymorphisms, and by epigenetic factors, whereby pro-inflammatory cytokines, reactive oxygen species, reactive nitrogen species and nuclear factor-κB affect gene DNA methylation and histone acetylation and also induce several microRNAs including miR-155, miR-181b-1 and miR-146a. Adult HPA axis activity is regulated by (i) genetic factors, such as glucocorticoid receptor polymorphisms; (ii) epigenetic factors affecting glucocorticoid receptor function or expression, including the methylation status of alternative promoter regions of NR3C1 and the methylation of FKBP5 and HSD11β2; (iii) chronic inflammation and chronic nitrosative and oxidative stress. Finally, it is shown how severe psychological stress adversely affects mitochondrial structure and functioning and is associated with changes in brain mitochondrial DNA copy number and transcription; mitochondria can act as couriers of childhood stress into adulthood.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, P.O. Box 291, Geelong, Victoria, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Level 1 North, Main Block, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, 35 Poplar Rd, Parkville, Victoria, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - André F Carvalho
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Centre for Addiction & Mental Health (CAMH), Toronto, ON, Canada
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK.
| |
Collapse
|
49
|
Serotonergically dependent antidepressant-like activity on behavior and stress axis responsivity of acacetin. Pharmacol Res 2019; 146:104310. [DOI: 10.1016/j.phrs.2019.104310] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 04/27/2019] [Accepted: 06/10/2019] [Indexed: 01/22/2023]
|
50
|
Lieb MW, Weidner M, Arnold MR, Loupy KM, Nguyen KT, Hassell JE, Schnabel KS, Kern R, Day HEW, Lesch KP, Waider J, Lowry CA. Effects of maternal separation on serotonergic systems in the dorsal and median raphe nuclei of adult male Tph2-deficient mice. Behav Brain Res 2019; 373:112086. [PMID: 31319134 DOI: 10.1016/j.bbr.2019.112086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 06/18/2019] [Accepted: 07/13/2019] [Indexed: 12/13/2022]
Abstract
Previous studies have highlighted interactions between serotonergic systems and adverse early life experience as important gene x environment determinants of risk of stress-related psychiatric disorders. Evidence suggests that mice deficient in Tph2, the rate-limiting enzyme for brain serotonin synthesis, display disruptions in behavioral phenotypes relevant to stress-related psychiatric disorders. The aim of this study was to determine how maternal separation in wild-type, heterozygous, and Tph2 knockout mice affects mRNA expression of serotonin-related genes. Serotonergic genes studied included Tph2, the high-affinity, low-capacity, sodium-dependent serotonin transporter (Slc6a4), the serotonin type 1a receptor (Htr1a), and the corticosterone-sensitive, low-affinity, high-capacity sodium-independent serotonin transporter, organic cation transporter 3 (Slc22a3). Furthermore, we studied corticotropin-releasing hormone receptors 1 (Crhr1) and 2 (Crhr2), which play important roles in controlling serotonergic neuronal activity. For this study, offspring of Tph2 heterozygous dams were exposed to daily maternal separation for the first two weeks of life. Adult, male wild-type, heterozygous, and homozygous offspring were subsequently used for molecular analysis. Maternal separation differentially altered serotonergic gene expression in a genotype- and topographically-specific manner. For example, maternal separation increased Slc6a4 mRNA expression in the dorsal part of the dorsal raphe nucleus in Tph2 heterozygous mice, but not in wild-type or knockout mice. Overall, these data are consistent with the hypothesis that gene x environment interactions, including serotonergic genes and adverse early life experience, play an important role in vulnerability to stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Margaret W Lieb
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Magdalena Weidner
- Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany; Department of Psychiatry and Psychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands.
| | - Mathew R Arnold
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Kelsey M Loupy
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Kadi T Nguyen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - James E Hassell
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - K'Loni S Schnabel
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Raphael Kern
- Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany.
| | - Heidi E W Day
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany; Department of Psychiatry and Psychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.
| | - Jonas Waider
- Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany.
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Department of Physical Medicine & Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO 80045, USA; Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Aurora, CO 80045, USA.
| |
Collapse
|