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
|
Oubraim S, Hausknecht K, Micov V, Shen RY, Haj-Dahmane S. Chemogenetic inhibition of prefrontal cortex inputs to dorsal raphe reduces anxiety behaviors in male rat model of fetal alcohol spectrum disorder. Sci Rep 2025; 15:14397. [PMID: 40275074 PMCID: PMC12022358 DOI: 10.1038/s41598-025-99181-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
Prenatal ethanol exposure (PE) causes Fetal Alcohol Spectrum Disorders (FASD), characterized by cognitive, behavioral, and emotional deficits, including anxiety and depression. PE-induced alteration in the function of dorsal raphe nucleus (DRN) serotonin (5-HT) neurons is thought to be major contributing factor for increased anxiety. However, the precise neuronal circuits involved are unknown. Using electrophysiology, optogenetics, chemogenetics, and behavioral approaches, we find that PE preferentially potentiates medial prefrontal cortex (mPFC) glutamatergic inputs, but not lateral habenula (LHb), to DRN 5-HT neurons projecting to mPFC. Additionally, PE also increases the strength of LHb but not mPFC excitatory inputs to DRN 5-HT neurons projecting to central amygdala (Ce). This input and target selective effect of PE was mediated by a circuit-specific increase in nitric oxide (NO) signaling. Importantly, chemogenetic inhibition of mPFC-DRN neuronal circuit blunted anxiety-like behaviors in PE rats. As such, our results unraveled the DRN neuronal circuitries affected by PE, which gate FASD-induced anxiety-like behaviors.
Collapse
Affiliation(s)
- Saida Oubraim
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, USA
| | - Kathryn Hausknecht
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, USA
| | - Veronika Micov
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, USA
| | - Roh-Yu Shen
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, USA
- University at Buffalo Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 1021 Main Street, Buffalo, NY, 14203, USA
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, USA.
- University at Buffalo Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 1021 Main Street, Buffalo, NY, 14203, USA.
| |
Collapse
|
3
|
Kilpatrick LA, Church A, Meriwether D, Mahurkar-Joshi S, Li VW, Sohn J, Reist J, Labus JS, Dong T, Jacobs JP, Naliboff BD, Chang L, Mayer EA. Differential brainstem connectivity according to sex and menopausal status in healthy male and female individuals. Biol Sex Differ 2025; 16:25. [PMID: 40251694 PMCID: PMC12007138 DOI: 10.1186/s13293-025-00709-4] [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: 08/07/2024] [Accepted: 04/04/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Brainstem nuclei play a critical role in both ascending monoaminergic modulation of cortical function and arousal, and in descending bulbospinal pain modulation. Even though sex-related differences in the function of both systems have been reported in animal models, a complete understanding of sex differences, as well as menopausal effects, in brainstem connectivity in humans is lacking. This study evaluated resting-state connectivity of the dorsal raphe nucleus, right and left locus coeruleus complex (LCC), and periaqueductal gray (PAG) according to sex and menopausal status in healthy individuals. In addition, relationships between systemic estrogen levels and brainstem-network connectivity were examined in a subset of participants. METHODS Resting-state fMRI was performed in 47 healthy male (age, 31.2 ± 8.0 years), 53 healthy premenopausal female (age, 24.7 ± 7.3 years; 22 in the follicular phase, 31 in the luteal phase), and 20 postmenopausal female participants (age, 54.6 ± 7.2 years). Permutation Analysis of Linear Models (5000 permutations) was used to evaluate differences in brainstem-network connectivity according to sex and menopausal status, controlling for age. In 10 males and 17 females (9 premenopausal; 8 postmenopausal), estrogen and estrogen metabolite levels in plasma and stool were determined by liquid chromatography-mass spectrometry/mass spectrometry. Relationships between estrogen levels and brainstem-network connectivity were evaluated by partial least squares analysis. RESULTS Left LCC-executive control network connectivity showed an overall sex difference (p = 0.02), with higher connectivity in females than in males; however, this was mainly due to differences between males and premenopausal females (p = 0.008). Additional sex differences were dependent on menopausal status: PAG-default mode network (DMN) connectivity was higher in postmenopausal females than in males (p = 0.04), and PAG-sensorimotor network (SMN) connectivity was higher in premenopausal females than in males (p = 0.03) and postmenopausal females (p = 0.007). Notably, higher free 2-hydroxyestrone levels in stool were reliably associated with higher PAG-SMN and PAG-DMN connectivity in premenopausal females (p < 0.01). CONCLUSIONS Healthy females show higher brainstem-network connectivity involved in cognitive control, sensorimotor function, and self-relevant processes than males, dependent on their menopausal status. Further, 2-hydroxyestrone, implicated in pain, may modulate PAG connectivity in premenopausal females. These findings may relate to differential vulnerabilities to chronic stress-sensitive disorders at different life stages.
Collapse
Affiliation(s)
- Lisa A Kilpatrick
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Arpana Church
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA
| | - David Meriwether
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Swapna Mahurkar-Joshi
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Vince W Li
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jessica Sohn
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Juliana Reist
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jennifer S Labus
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA
- Brain Research Institute, Gonda (Goldschmied) Neuroscience and Genetics Research Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Tien Dong
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Jonathan P Jacobs
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Bruce D Naliboff
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Lin Chang
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA.
| | - Emeran A Mayer
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Falconi-Sobrinho LL, Fonseca-Rodrigues D, da Silva ML, Coimbra NC, Pinto-Ribeiro F. Neuroanatomical and neurochemical substrates mediating fear-induced antinociception: A systematic review of rodent preclinical studies. Neurosci Biobehav Rev 2025; 168:105959. [PMID: 39613200 DOI: 10.1016/j.neubiorev.2024.105959] [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: 06/03/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
Fear-induced antinociception (FIA), an instinctive defensive response producing pain suppression in stressful and/or dangerous situations, has been the subject of extensive research to elucidate the mechanisms involved in triggering and controlling pain during emotional disorders. In this systematic review, we synthesized pre-clinical studies that demonstrated the neural hodology and the neurochemical bases of FIA in laboratory animals. The literature search in PubMed, Web of Science, Science Direct, and Scopus, from inception up to July 2022, retrieved 797 articles from which 50 studies were included in this review. This review highlights key encephalic regions implicated in the modulation of FIA, such as the prefrontal cortex, the amygdaloid complex, the hippocampus, the hypothalamus, the corpora quadrigemina, the periaqueductal gray matter, and some reticular formation nuclei. FIA-related neural pathways, neurotransmitters and neuromodulators such as glutamatergic, serotonergic, norepinephrine, GABAergic, nitrergic, opioidergic and endocannabinoid connections across these encephalic regions were also addressed. Understanding these neural circuits and molecular neural mediation sheds light on the complex interplay between fear, anxiety, and pain modulation, offering potential avenues for therapeutic interventions targeting pain management in the context of heightened emotional states.
Collapse
Affiliation(s)
- Luiz Luciano Falconi-Sobrinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Gualtar Campus, Braga 4710-057, Portugal; ICVS/3B's-PT Government Associate Laboratory, Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Guimarães, Portugal; Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (USP), Av. Bandeirantes 3900, Ribeirão Preto, São Paulo 14049-900, Brazil; NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil; Postgraduate Program in Biosciences Applied to Health (PPGB), Federal University of Alfenas (UNIFAL), Alfenas, Minas Gerais, Brazil; Institute of Biomedical Sciences, Federal University of Alfenas (UNIFAL), Alfenas, Brazil
| | - Diana Fonseca-Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Gualtar Campus, Braga 4710-057, Portugal; ICVS/3B's-PT Government Associate Laboratory, Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Guimarães, Portugal
| | - Marcelo Lourenço da Silva
- Postgraduate Program in Biosciences Applied to Health (PPGB), Federal University of Alfenas (UNIFAL), Alfenas, Minas Gerais, Brazil; Institute of Biomedical Sciences, Federal University of Alfenas (UNIFAL), Alfenas, Brazil
| | - Norberto Cysne Coimbra
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (USP), Av. Bandeirantes 3900, Ribeirão Preto, São Paulo 14049-900, Brazil; NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil.
| | - Filipa Pinto-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Gualtar Campus, Braga 4710-057, Portugal; ICVS/3B's-PT Government Associate Laboratory, Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Guimarães, Portugal.
| |
Collapse
|
5
|
Dudhabhate BB, Awathale SN, Choudhary AG, Subhedar NK, Kokare DM. Deep brain stimulation targeted at lateral hypothalamus-medial forebrain bundle reverses depressive-like symptoms and related cognitive deficits in rat: Role of serotoninergic system. Neuroscience 2024; 556:96-113. [PMID: 39103042 DOI: 10.1016/j.neuroscience.2024.07.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/12/2024] [Accepted: 07/31/2024] [Indexed: 08/07/2024]
Abstract
The aim of the study is to understand the rationale behind the application of deep brain stimulation (DBS) in the treatment of depression. Male Wistar rats, rendered depressive with chronic unpredictable mild stress (CUMS) were implanted with electrode in the lateral hypothalamus-medial forebrain bundle (LH-MFB) and subjected to deep brain stimulation (DBS) for 4 h each day for 14 days. DBS rats, as well as controls, were screened for a range of parameters indicative of depressive state. Symptomatic features noticed in CUMS rats like the memory deficit, anhedonia, reduction in body weight and 5-hydroxytryptamine (5-HT) and 5-hydroxyindoleacetic acid (5-HIAA) levels in mPFC and elevated plasma corticosterone were reversed in rats subjected to DBS. DBS arrested CUMS induced degeneration of 5-HT cells in interfascicular region of dorsal raphe nucleus (DRif) and fibers in LH-MFB and induced dendritic proliferation in mPFC neurons. MFB is known to serve as a major conduit for the DRif-mPFC serotoninergic pathway. While the density of serotonin fibers in the LH-MFB circuit was reduced in CUMS, it was upregulated in DBS-treated rats. Furthermore, microinjection of 5-HT1A receptor antagonist, WAY100635 into mPFC countered the positive effects of DBS like the antidepressant and memory-enhancing action. In this background, we suggest that DBS at LH-MFB may exercise positive effect in depressive rats via upregulation of the serotoninergic system. While these data drawn from the experiments on rat provide meaningful clues, we suggest that further studies aimed at understanding the usefulness of DBS at LH-MFB in humans may be rewarding.
Collapse
Affiliation(s)
- Biru B Dudhabhate
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India
| | - Sanjay N Awathale
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule 424 001, Maharashtra, India
| | - Amit G Choudhary
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India
| | - Nishikant K Subhedar
- Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pune 411 008, India
| | - Dadasaheb M Kokare
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India.
| |
Collapse
|
6
|
Kilpatrick LA, Gupta A, Meriwether D, Mahurkar-Joshi S, Li VW, Sohn J, Reist J, Labus JS, Dong T, Jacobs JP, Naliboff BD, Chang L, Mayer EA. Differential brainstem connectivity according to sex and menopausal status in healthy men and women. RESEARCH SQUARE 2024:rs.3.rs-4875269. [PMID: 39184081 PMCID: PMC11343298 DOI: 10.21203/rs.3.rs-4875269/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Background Brainstem nuclei play a critical role in both ascending monoaminergic modulation of cortical function and arousal, and in descending bulbospinal pain modulation. Even though sex-related differences in the function of both systems have been reported in animal models, a complete understanding of sex differences, as well as menopausal effects, in brainstem connectivity in humans is lacking. This study evaluated resting-state connectivity of the dorsal raphe nucleus (DRN), right and left locus coeruleus complex (LCC), and periaqueductal gray (PAG) according to sex and menopausal status in healthy individuals. In addition, relationships between systemic estrogen levels and brainstem-network connectivity were examined in a subset of participants. Methods Resting-state fMRI was performed in 50 healthy men (age, 31.2 ± 8.0 years), 53 healthy premenopausal women (age, 24.7 ± 7.3 years; 22 in the follicular phase, 31 in the luteal phase), and 20 postmenopausal women (age, 54.6 ± 7.2 years). Permutation Analysis of Linear Models (5000 permutations) was used to evaluate differences in brainstem-network connectivity according to sex and menopausal status, controlling for age. In 10 men and 17 women (9 premenopausal; 8 postmenopausal), estrogen and estrogen metabolite levels in plasma and stool were determined by liquid chromatography-mass spectrometry/mass spectrometry. Relationships between estrogen levels and brainstem-network connectivity were evaluated by partial least squares analysis. Results Left LCC-executive control network (ECN) connectivity showed an overall sex difference (p = 0.02), with higher connectivity in women than in men; however, this was mainly due to differences between men and pre-menopausal women (p = 0.008). Additional sex differences were dependent on menopausal status: PAG-default mode network (DMN) connectivity was higher in postmenopausal women than in men (p = 0.04), and PAG-sensorimotor network (SMN) connectivity was higher in premenopausal women than in men (p = 0.03) and postmenopausal women (p = 0.007). Notably, higher free 2-hydroxyestrone levels in stool were associated with higher PAG-SMN and PAG-DMN connectivity in premenopausal women (p < 0.01). Conclusions Healthy women show higher brainstem-network connectivity involved in cognitive control, sensorimotor function, and self-relevant processes than men, dependent on their menopausal status. Further, 2-hydroxyestrone, implicated in pain, may modulate PAG connectivity in premenopausal women. These findings may relate to differential vulnerabilities to chronic stress-sensitive disorders at different life stages.
Collapse
|
7
|
Mitten EH, Souders A, Marron Fernandez de Velasco E, Wickman K. Stress-induced anxiety-related behavior in mice is driven by enhanced excitability of ventral tegmental area GABA neurons. Front Behav Neurosci 2024; 18:1425607. [PMID: 39086371 PMCID: PMC11288924 DOI: 10.3389/fnbeh.2024.1425607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction Stress and trauma are significant risk factors for many neuropsychiatric disorders and diseases, including anxiety disorders. Stress-induced anxiety symptoms have been attributed to enhanced excitability in circuits controlling fear, anxiety, and aversion. A growing body of evidence has implicated GABAergic neurons of the ventral tegmental area (VTA) in aversion processing and affective behavior. Methods We used an unpredictable footshock (uFS) model, together with electrophysiological and behavioral approaches, to investigate the role of VTA GABA neurons in anxiety-related behavior in mice. Results One day after a single uFS session, C57BL/6J mice exhibited elevated anxiety-related behavior and VTA GABA neuron excitability. The enhanced excitability of VTA GABA neurons was correlated with increased glutamatergic input and a reduction in postsynaptic signaling mediated via GABAA and GABAB receptors. Chemogenetic activation of VTA GABA neurons was sufficient to increase anxiety-related behavior in stress-naïve mice. In addition, chemogenetic inhibition of VTA GABA neurons suppressed anxiety-related behavior in mice exposed to uFS. Discussion These data show that VTA GABA neurons are an early substrate for stress-induced anxiety-related behavior in mice and suggest that approaches mitigating enhanced excitability of VTA GABA neurons may hold promise for the treatment of anxiety provoked by stress and trauma.
Collapse
Affiliation(s)
- Eric H. Mitten
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Anna Souders
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| | | | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
8
|
Cheng J, Chen L, Zheng YN, Liu J, Zhang L, Zhang XM, Huang L, Yuan QL. Disfunction of dorsal raphe nucleus-hippocampus serotonergic-HTR3 transmission results in anxiety phenotype of Neuroplastin 65-deficient mice. Acta Pharmacol Sin 2024; 45:1393-1405. [PMID: 38528118 PMCID: PMC11192762 DOI: 10.1038/s41401-024-01252-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/26/2024] [Indexed: 03/27/2024]
Abstract
Anxiety disorders are the most common psychiatric condition, but the etiology of anxiety disorders remains largely unclear. Our previous studies have shown that neuroplastin 65 deficiency (NP65-/-) mice exhibit abnormal social and mental behaviors and decreased expression of tryptophan hydroxylase 2 (TPH2) protein. However, whether a causal relationship between TPH2 reduction and anxiety disorders exists needs to be determined. In present study, we found that replenishment of TPH2 in dorsal raphe nucleus (DRN) enhanced 5-HT level in the hippocampus and alleviated anxiety-like behaviors. In addition, injection of AAV-NP65 in DRN significantly increased TPH2 expression in DRN and hippocampus, and reduced anxiety-like behaviors. Acute administration of exogenous 5-HT or HTR3 agonist SR57227A in hippocampus mitigated anxiety-like behaviors in NP65-/- mice. Moreover, replenishment of TPH2 in DRN partly repaired the impairment of long-term potentiation (LTP) maintenance in hippocampus of NP65-/- mice. Finally, we found that loss of NP65 lowered transcription factors Lmx1b expression in postnatal stage and replenishment of NP65 in DRN reversed the decrease in Lmx1b expression of NP65-/- mice. Together, our findings reveal that NP65 deficiency induces anxiety phenotype by downregulating DRN-hippocampus serotonergic-HTR3 transmission. These studies provide a novel and insightful view about NP65 function, suggesting an attractive potential target for treatment of anxiety disorders.
Collapse
Affiliation(s)
- Jie Cheng
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ling Chen
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ya-Ni Zheng
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Juan Liu
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Lei Zhang
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Xiao-Ming Zhang
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Liang Huang
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Qiong-Lan Yuan
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
9
|
Livermore JJA, Skora LI, Adamatzky K, Garfinkel SN, Critchley HD, Campbell-Meiklejohn D. General and anxiety-linked influences of acute serotonin reuptake inhibition on neural responses associated with attended visceral sensation. Transl Psychiatry 2024; 14:241. [PMID: 38844469 PMCID: PMC11156930 DOI: 10.1038/s41398-024-02971-3] [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: 11/07/2023] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
Ordinary sensations from inside the body are important causes and consequences of our affective states and behaviour, yet the roles of neurotransmitters in interoceptive processing have been unclear. With a within-subjects design, this experiment tested the impacts of acute increases of endogenous extracellular serotonin on the neural processing of attended internal sensations and the links of these effects to anxiety using a selective serotonin reuptake inhibitor (SSRI) (20 mg CITALOPRAM) and a PLACEBO. Twenty-one healthy volunteers (fourteen female, mean age 23.9) completed the Visceral Interoceptive Attention (VIA) task while undergoing functional magnetic resonance imaging (fMRI) with each treatment. The VIA task required focused attention on the heart, stomach, or visual sensation. The relative neural interoceptive responses to heart sensation [heart minus visual attention] (heart-IR) and stomach sensation [stomach minus visual attention] (stomach-IR) were compared between treatments. Visual attention subtraction controlled for the general effects of CITALOPRAM on sensory processing. CITALOPRAM was associated with lower interoceptive processing in viscerosensory (the stomach-IR of bilateral posterior insular cortex) and integrative/affective (the stomach-IR and heart-IR of bilateral amygdala) components of interoceptive neural pathways. In anterior insular cortex, CITALOPRAM reductions of heart-IR depended on anxiety levels, removing a previously known association between anxiety and the region's response to attended heart sensation observed with PLACEBO. Preliminary post hoc analysis indicated that CITALOPRAM effects on the stomach-IR of the amygdalae corresponded to acute anxiety changes. This direct evidence of general and anxiety-linked serotonergic influence on neural interoceptive processes advances our understanding of interoception, its regulation, and anxiety.
Collapse
Affiliation(s)
| | - Lina I Skora
- School of Psychology, University of Sussex, Brighton, UK
- Heinrich Heine Universität, Düsseldorf, Germany
- Sussex Centre for Consciousness Science, University of Sussex, Brighton, UK
| | | | - Sarah N Garfinkel
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Hugo D Critchley
- Sussex Centre for Consciousness Science, University of Sussex, Brighton, UK
- Brighton and Sussex Medical School, Brighton, UK
- Sussex Partnership NHS Foundation Trust, Brighton, UK
| | | |
Collapse
|
10
|
de Noronha SISR, de Moraes LAG, Hassell JE, Stamper CE, Arnold MR, Heinze JD, Foxx CL, Lieb MM, Cler KE, Karns BL, Jaekel S, Loupy KM, Silva FCS, Chianca-Jr DA, Lowry CA, de Menezes RC. High-fat diet, microbiome-gut-brain axis signaling, and anxiety-like behavior in male rats. Biol Res 2024; 57:23. [PMID: 38705984 PMCID: PMC11071217 DOI: 10.1186/s40659-024-00505-1] [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: 10/26/2023] [Accepted: 04/23/2024] [Indexed: 05/07/2024] Open
Abstract
Obesity, associated with the intake of a high-fat diet (HFD), and anxiety are common among those living in modern urban societies. Recent studies suggest a role of microbiome-gut-brain axis signaling, including a role for brain serotonergic systems in the relationship between HFD and anxiety. Evidence suggests the gut microbiome and the serotonergic brain system together may play an important role in this response. Here we conducted a nine-week HFD protocol in male rats, followed by an analysis of the gut microbiome diversity and community composition, brainstem serotonergic gene expression (tph2, htr1a, and slc6a4), and anxiety-related defensive behavioral responses. We show that HFD intake decreased alpha diversity and altered the community composition of the gut microbiome in association with obesity, increased brainstem tph2, htr1a and slc6a4 mRNA expression, including in the caudal part of the dorsomedial dorsal raphe nucleus (cDRD), a subregion previously associated with stress- and anxiety-related behavioral responses, and, finally, increased anxiety-related defensive behavioral responses. The HFD increased the Firmicutes/Bacteroidetes ratio relative to control diet, as well as higher relative abundances of Blautia, and decreases in Prevotella. We found that tph2, htr1a and slc6a4 mRNA expression were increased in subregions of the dorsal raphe nucleus in the HFD, relative to control diet. Specific bacterial taxa were associated with increased serotonergic gene expression in the cDRD. Thus, we propose that HFD-induced obesity is associated with altered microbiome-gut-serotonergic brain axis signaling, leading to increased anxiety-related defensive behavioral responses in rats.
Collapse
Affiliation(s)
- Sylvana I S Rendeiro de Noronha
- Department of Biological Sciences, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG, 35400-000, Brazil
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Lauro Angelo Gonçalves de Moraes
- Department of Biological Sciences, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG, 35400-000, Brazil
- Computing Department, Federal University of Ouro Preto, Ouro Preto, MG, 35400-000, Brazil
| | - James E Hassell
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Christopher E Stamper
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Mathew R Arnold
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Jared D Heinze
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Christine L Foxx
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Margaret M Lieb
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Kristin E Cler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Bree L Karns
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Sophia Jaekel
- 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
| | - Fernanda C S Silva
- Department of Biological Sciences, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG, 35400-000, Brazil
| | - Deoclécio Alves Chianca-Jr
- Department of Biological Sciences, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG, 35400-000, Brazil
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Rodrigo Cunha de Menezes
- Department of Biological Science Laboratory of Cardiovascular Physiology, Campus Morro do Cruzeiro s/n, Ouro Preto, 35400-000, MG, Brazil.
| |
Collapse
|
11
|
Gullino LS, Fuller C, Dunn P, Collins HM, El Mestikawy S, Sharp T. Evidence for a Role of 5-HT-glutamate Co-releasing Neurons in Acute Stress Mechanisms. ACS Chem Neurosci 2024; 15:1185-1196. [PMID: 38377469 PMCID: PMC10958520 DOI: 10.1021/acschemneuro.3c00758] [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: 11/22/2023] [Revised: 01/18/2024] [Accepted: 01/26/2024] [Indexed: 02/22/2024] Open
Abstract
A major subpopulation of midbrain 5-hydroxytryptamine (5-HT) neurons expresses the vesicular glutamate transporter 3 (VGLUT3) and co-releases 5-HT and glutamate, but the function of this co-release is unclear. Given the strong links between 5-HT and uncontrollable stress, we used a combination of c-Fos immunohistochemistry and conditional gene knockout mice to test the hypothesis that glutamate co-releasing 5-HT neurons are activated by stress and involved in stress coping. Acute, uncontrollable swim stress increased c-Fos immunoreactivity in neurons co-expressing VGLUT3 and the 5-HT marker tryptophan hydroxylase 2 (TPH2) in the dorsal raphe nucleus (DRN). This effect was localized in the ventral DRN subregion and prevented by the antidepressant fluoxetine. In contrast, a more controllable stressor, acute social defeat, had no effect on c-Fos immunoreactivity in VGLUT3-TPH2 co-expressing neurons in the DRN. To test whether activation of glutamate co-releasing 5-HT neurons was causally linked to stress coping, mice with a specific deletion of VGLUT3 in 5-HT neurons were exposed to acute swim stress. Compared to wildtype controls, the mutant mice showed increased climbing behavior, a measure of active coping. Wildtype mice also showed increased climbing when administered fluoxetine, revealing an interesting parallel between the behavioral effects of genetic loss of VGLUT3 in 5-HT neurons and 5-HT reuptake inhibition. We conclude that 5-HT-glutamate co-releasing neurons are recruited by exposure to uncontrollable stress. Furthermore, natural variation in the balance of 5-HT and glutamate co-released at the 5-HT synapse may impact stress susceptibility.
Collapse
Affiliation(s)
- L. Sophie Gullino
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| | - Cara Fuller
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| | - Poppy Dunn
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| | - Helen M. Collins
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| | - Salah El Mestikawy
- Douglas
Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC H4H
1R3, Canada
- Sorbonne
Université, INSERM, CNRS, Neuroscience Paris Seine –
Institut de Biologie Paris Seine (NPS – IBPS), 75005 Paris, France
| | - Trevor Sharp
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| |
Collapse
|
12
|
Li HQ, Jiang W, Ling L, Pratelli M, Chen C, Gupta V, Godavarthi SK, Spitzer NC. Generalized fear after acute stress is caused by change in neuronal cotransmitter identity. Science 2024; 383:1252-1259. [PMID: 38484078 PMCID: PMC11830151 DOI: 10.1126/science.adj5996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/22/2024] [Indexed: 03/19/2024]
Abstract
Overgeneralization of fear to harmless situations is a core feature of anxiety disorders resulting from acute stress, yet the mechanisms by which fear becomes generalized are poorly understood. In this study, we show that generalized fear in mice results from a transmitter switch from glutamate to γ-aminobutyric acid (GABA) in serotonergic neurons of the lateral wings of the dorsal raphe. Similar change in transmitter identity was found in the postmortem brains of individuals with posttraumatic stress disorder (PTSD). Overriding the transmitter switch in mice prevented the acquisition of generalized fear. Corticosterone release and activation of glucocorticoid receptors mediated the switch, and prompt antidepressant treatment blocked the cotransmitter switch and generalized fear. Our results provide important insight into the mechanisms involved in fear generalization.
Collapse
Affiliation(s)
- Hui-quan Li
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Wuji Jiang
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Li Ling
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Marta Pratelli
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Cong Chen
- Department of Cellular and Molecular Medicine, University of California San Diego; La Jolla, California 92093
| | - Vaidehi Gupta
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Swetha K. Godavarthi
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Nicholas C. Spitzer
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| |
Collapse
|
13
|
Bernabe CS, Caliman IF, de Abreu ARR, Molosh AI, Truitt WA, Shekhar A, Johnson PL. Identification of a novel perifornical-hypothalamic-area-projecting serotonergic system that inhibits innate panic and conditioned fear responses. Transl Psychiatry 2024; 14:60. [PMID: 38272876 PMCID: PMC10811332 DOI: 10.1038/s41398-024-02769-3] [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: 05/11/2023] [Revised: 01/06/2024] [Accepted: 01/10/2024] [Indexed: 01/27/2024] Open
Abstract
The serotonin (5-HT) system is heavily implicated in the regulation of anxiety and trauma-related disorders such as panic disorder and post-traumatic stress disorder, respectively. However, the neural mechanisms of how serotonergic neurotransmission regulates innate panic and fear brain networks are poorly understood. Our earlier studies have identified that orexin (OX)/glutamate neurons within the perifornical hypothalamic area (PFA) play a critical role in adaptive and pathological panic and fear. While site-specific and electrophysiological studies have shown that intracranial injection and bath application of 5-HT inhibits PFA neurons via 5-HT1a receptors, they largely ignore circuit-specific neurotransmission and its physiological properties that occur in vivo. Here, we investigate the role of raphe nuclei 5-HT inputs into the PFA in panic and fear behaviors. We initially confirmed that photostimulation of glutamatergic neurons in the PFA of rats produces robust cardioexcitation and flight/aversive behaviors resembling panic-like responses. Using the retrograde tracer cholera toxin B, we determined that the PFA receives discrete innervation of serotonergic neurons clustered in the lateral wings of the dorsal (lwDRN) and in the median (MRN) raphe nuclei. Selective lesions of these serotonergic projections with saporin toxin resulted in similar panic-like responses during the suffocation-related CO2 challenge and increased freezing to fear-conditioning paradigm. Conversely, selective stimulation of serotonergic fibers in the PFA attenuated both flight/escape behaviors and cardioexcitation responses elicited by the CO2 challenge and induced conditioned place preference. The data here support the hypothesis that PFA projecting 5-HT neurons in the lwDRN/MRN represents a panic/fear-off circuit and may also play a role in reward behavior.
Collapse
Affiliation(s)
- Cristian S Bernabe
- Department of Anatomy, Cellular Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Izabela F Caliman
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Aline R R de Abreu
- Departamento de Alimentos, Escola de Nutrição da Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Andrei I Molosh
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - William A Truitt
- Department of Anatomy, Cellular Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Anantha Shekhar
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Philip L Johnson
- Department of Biology, University of South Dakota, Vermillion, SD, USA
| |
Collapse
|
14
|
Payet JM, Stevens L, Russo AM, Jaehne EJ, van den Buuse M, Kent S, Lowry CA, Baratta MV, Hale MW. The Role of Dorsal Raphe Nucleus Serotonergic Systems in Emotional Learning and Memory in Male BALB/c Mice. Neuroscience 2023; 534:1-15. [PMID: 37852412 DOI: 10.1016/j.neuroscience.2023.10.003] [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: 08/14/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/20/2023]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the first-line pharmacological treatment for a variety of anxiety-, trauma- and stressor-related disorders. Although they are efficacious, therapeutic improvements require several weeks of treatment and are often associated with an initial exacerbation of symptoms. The dorsal raphe nucleus (DR) has been proposed as an important target for the modulation of emotional responses and the therapeutic effects of SSRIs. Using a fear-conditioning paradigm we aimed to understand how SSRIs affect emotional learning and memory, and their effects on serotonergic circuitry. Adult male BALB/c mice were treated with vehicle (n = 16) or the SSRI fluoxetine (18 mg/kg/d) acutely (n = 16), or chronically (21d, n = 16), prior to fear conditioning. Treatment was stopped, and half of the mice (n = 8/treatment group) were exposed to cued fear memory recall 72 h later. Activation of DR serotonergic neurons during fear conditioning (Experiment 1) or fear memory recall (Experiment 2), was measured using dual-label immunohistochemistry for Tph2 and c-Fos. Acute and chronic fluoxetine treatment reduced associative fear learning without affecting memory recall and had opposite effects on anxiety-like behaviour. Acute fluoxetine decreased serotonergic activity in the DR, while chronic treatment led to serotonergic activity that was indistinguishable from that of control levels in DRD and DRV subpopulations. Chronic fluoxetine facilitated fear extinction, which was associated with rostral DRD inhibition. These findings provide further evidence that SSRIs can alter aspects of learning and memory processes and are consistent with a role for discrete populations of DR serotonergic neurons in regulating fear- and anxiety-related behaviours.
Collapse
Affiliation(s)
- Jennyfer M Payet
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Laura Stevens
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Adrian M Russo
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Emily J Jaehne
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia; Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Maarten van den Buuse
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Stephen Kent
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Christopher A Lowry
- Department of Integrative Physiology and Centre for Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Michael V Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Matthew W Hale
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia.
| |
Collapse
|
15
|
Miyanishi H, Suga S, Sumi K, Takakuwa M, Izuo N, Asano T, Muramatsu SI, Nitta A. The Role of GABA in the Dorsal Striatum-Raphe Nucleus Circuit Regulating Stress Vulnerability in Male Mice with High Levels of Shati/Nat8l. eNeuro 2023; 10:ENEURO.0162-23.2023. [PMID: 37813564 PMCID: PMC10598637 DOI: 10.1523/eneuro.0162-23.2023] [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: 05/16/2023] [Revised: 09/26/2023] [Accepted: 10/01/2023] [Indexed: 10/17/2023] Open
Abstract
Depression is a frequent and serious illness, and stress is considered the main risk factor for its onset. First-line antidepressants increase serotonin (5-hydroxytryptamine; 5-HT) levels in the brain. We previously reported that an N-acetyltransferase, Shati/Nat8l, is upregulated in the dorsal striatum (dSTR) of stress-susceptible mice exposed to repeated social defeat stress (RSDS) and that dSTR Shati/Nat8l overexpression in mice (dSTR-Shati OE) induces stress vulnerability and local reduction in 5-HT content. Male mice were used in this study, and we found that dSTR 5-HT content decreased in stress-susceptible but not in resilient mice. Moreover, vulnerability to stress in dSTR-Shati OE mice was suppressed by the activation of serotonergic neurons projecting from the dorsal raphe nucleus (dRN) to the dSTR, followed by upregulation of 5-HT content in the dSTR using designer receptors exclusively activated by designer drugs (DREADD). We evaluated the role of GABA in modulating the serotonergic system in the dRN. Stress-susceptible after RSDS and dSTR-Shati OE mice exhibited an increase in dRN GABA content. Furthermore, dRN GABA content was correlated with stress sensitivity. We found that the blockade of GABA signaling in the dRN suppressed stress susceptibility in dSTR-Shati OE mice. In conclusion, we propose that dSTR 5-HT and dRN GABA, controlled by striatal Shati/Nat8l via the dSTR-dRN neuronal circuitry, critically regulate stress sensitivity. Our study provides insights into the neural processes that underlie stress and suggests that dSTR Shati/Nat8l could be a novel therapeutic target for drugs against depression, allowing direct control of the dRN serotonergic system.
Collapse
Affiliation(s)
- Hajime Miyanishi
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Shiori Suga
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Kazuyuki Sumi
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Miho Takakuwa
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Naotaka Izuo
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Takashi Asano
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Shin-Ichi Muramatsu
- Division of Neurological Gene Therapy, Center for Open Innovation, Jichi Medical University, Shimotsuke 329-0498, Japan
- Center for Gene & Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo 108-0071, Japan
| | - Atsumi Nitta
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
16
|
Ikeda N, Kawasaki M, Baba K, Nishimura H, Fujitani T, Suzuki H, Matsuura T, Ohnishi H, Shimizu M, Sanada K, Nishimura K, Yoshimura M, Maruyama T, Conway-Campbell BL, Onaka T, Teranishi H, Hanada R, Ueta Y, Sakai A. Chemogenetic Activation of Oxytocin Neurons Improves Pain in a Reserpine-induced Fibromyalgia Rat Model. Neuroscience 2023; 528:37-53. [PMID: 37532013 DOI: 10.1016/j.neuroscience.2023.07.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Abstract
Fibromyalgia (FM) is a syndrome characterized by chronic pain with depression as a frequent comorbidity. However, efficient management of the pain and depressive symptoms of FM is lacking. Given that endogenous oxytocin (OXT) contributes to the regulation of pain and depressive disorders, herein, we investigated the role of OXT in an experimental reserpine-induced FM model. In FM model, OXT-monomeric red fluorescent protein 1 (OXT-mRFP1) transgenic rats exhibited increased depressive behavior and sensitivity in a mechanical nociceptive test, suggesting reduced pain tolerance. Additionally, the development of the FM-like phenotype in OXT-mRFP1 FM model rats was accompanied by a significant reduction in OXT mRNA expression in the magnocellular neurons of the paraventricular nucleus. OXT-mRFP1 FM model rats also had significantly fewer tryptophan hydroxylase (TPH)- and tyrosine hydroxylase (TH)-immunoreactive (ir) neurons as well as reduced serotonin and norepinephrine levels in the dorsal raphe and locus coeruleus. To investigate the effects of stimulating the endogenous OXT pathway, rats expressing OXT-human muscarinic acetylcholine receptor (hM3Dq)-mCherry designer receptors exclusively activated by designer drugs (DREADDs) were also assessed in the FM model. Treatment of these rats with clozapine-N-oxide (CNO), an hM3Dq-activating drug, significantly improved characteristic FM model-induced pathophysiological pain, but did not alter depressive-like behavior. The chemogenetically induced effects were reversed by pre-treatment with an OXT receptor antagonist, confirming the specificity of action via the OXT pathway. These results indicate that endogenous OXT may have analgesic effects in FM, and could be a potential target for effective pain management strategies for this disorder.
Collapse
Affiliation(s)
- Naofumi Ikeda
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Makoto Kawasaki
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.
| | - Kazuhiko Baba
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Haruki Nishimura
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Teruaki Fujitani
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hitoshi Suzuki
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Takanori Matsuura
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hideo Ohnishi
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Makiko Shimizu
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kenya Sanada
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kazuaki Nishimura
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Mitsuhiro Yoshimura
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Takashi Maruyama
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | | | - Tatsushi Onaka
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Japan
| | - Hitoshi Teranishi
- Department of Neurophysiology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Reiko Hanada
- Department of Neurophysiology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Akinori Sakai
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
17
|
Xie L, Wu H, Chen Q, Xu F, Li H, Xu Q, Jiao C, Sun L, Ullah R, Chen X. Divergent modulation of pain and anxiety by GABAergic neurons in the ventrolateral periaqueductal gray and dorsal raphe. Neuropsychopharmacology 2023; 48:1509-1519. [PMID: 36526697 PMCID: PMC10425368 DOI: 10.1038/s41386-022-01520-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/07/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022]
Abstract
The ventrolateral periaqueductal gray (vlPAG) collaborates with the dorsal raphe (DR) in pain regulation and emotional response. However, the roles of vlPAG and DR γ-aminobutyric acid (GABA)-ergic neurons in regulating nociception and anxiety are contradictory and poorly understood. Here, we observed that pharmacogenetic co-activation of vlPAG and DR GABAergic (vlPAG-DRGABA+) neurons enhanced sensitivity to mechanical stimulation and promoted anxiety-like behavior in naïve mice. Simultaneous inhibition of vlPAG-DRGABA+ neurons showed adaptive anti-nociception and anti-anxiety effects on mice with inflammatory pain. Notably, vlPAGGABA+ and DRGABA+ neurons exhibited opposing effects on the sensitivity to mechanical stimulation in both naïve state and inflammatory pain. In contrast to the role of vlPAGGABA+ neurons in pain processing, chemogenetic inhibition and chronic ablation of DRGABA+ neurons remarkably promoted nociception while selectively activating DRGABA+ neurons ameliorated inflammatory pain. Additionally, utilizing optogenetic technology, we observed that the pronociceptive effect arising from DRGABA+ neuronal inhibition was reversed by the systemic administration of morphine. Our results collectively provide new insights into the modulation of pain and anxiety by specific midbrain GABAergic subpopulations, which may provide a basis for cell type-targeted or subregion-targeted therapies for pain management.
Collapse
Affiliation(s)
- Linghua Xie
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Wu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qing Chen
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Xu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hua Li
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Xu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cuicui Jiao
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lihong Sun
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Rahim Ullah
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Xinzhong Chen
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
18
|
Bauer MB, Currie KPM. Serotonin and the serotonin transporter in the adrenal gland. VITAMINS AND HORMONES 2023; 124:39-78. [PMID: 38408804 PMCID: PMC11217909 DOI: 10.1016/bs.vh.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
The adrenal glands are key components of the mammalian endocrine system, helping maintain physiological homeostasis and the coordinated response to stress. Each adrenal gland has two morphologically and functionally distinct regions, the outer cortex and inner medulla. The cortex is organized into three concentric zones which secrete steroid hormones, including aldosterone and cortisol. Neural crest-derived chromaffin cells in the medulla are innervated by preganglionic sympathetic neurons and secrete catecholamines (epinephrine, norepinephrine) and neuropeptides into the bloodstream, thereby functioning as the neuroendocrine arm of the sympathetic nervous system. In this article we review serotonin (5-HT) and the serotonin transporter (SERT; SLC6A4) in the adrenal gland. In the adrenal cortex, 5-HT, primarily sourced from resident mast cells, acts as a paracrine signal to stimulate aldosterone and cortisol secretion through 5-HT4/5-HT7 receptors. Medullary chromaffin cells contain a small amount of 5-HT due to SERT-mediated uptake and express 5-HT1A receptors which inhibit secretion. The atypical mechanism of the 5-HT1A receptors and interaction with SERT fine tune this autocrine pathway to control stress-evoked catecholamine secretion. Receptor-independent signaling by SERT/intracellular 5-HT modulates the amount and kinetics of transmitter release from single vesicle fusion events. SERT might also influence stress-evoked upregulation of tyrosine hydroxylase transcription. Transient signaling via 5-HT3 receptors during embryonic development can limit the number of chromaffin cells found in the mature adrenal gland. Together, this emerging evidence suggests that the adrenal medulla is a peripheral hub for serotonergic control of the sympathoadrenal stress response.
Collapse
Affiliation(s)
- Mary Beth Bauer
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, South Broadway, Camden, NJ, United States
| | - Kevin P M Currie
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, South Broadway, Camden, NJ, United States.
| |
Collapse
|
19
|
Favoretto CA, Pagliusi M, Morais-Silva G. Involvement of brain cell phenotypes in stress-vulnerability and resilience. Front Neurosci 2023; 17:1175514. [PMID: 37476833 PMCID: PMC10354562 DOI: 10.3389/fnins.2023.1175514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/19/2023] [Indexed: 07/22/2023] Open
Abstract
Stress-related disorders' prevalence is epidemically increasing in modern society, leading to a severe impact on individuals' well-being and a great economic burden on public resources. Based on this, it is critical to understand the mechanisms by which stress induces these disorders. The study of stress made great progress in the past decades, from deeper into the hypothalamic-pituitary-adrenal axis to the understanding of the involvement of a single cell subtype on stress outcomes. In fact, many studies have used state-of-the-art tools such as chemogenetic, optogenetic, genetic manipulation, electrophysiology, pharmacology, and immunohistochemistry to investigate the role of specific cell subtypes in the stress response. In this review, we aim to gather studies addressing the involvement of specific brain cell subtypes in stress-related responses, exploring possible mechanisms associated with stress vulnerability versus resilience in preclinical models. We particularly focus on the involvement of the astrocytes, microglia, medium spiny neurons, parvalbumin neurons, pyramidal neurons, serotonergic neurons, and interneurons of different brain areas in stress-induced outcomes, resilience, and vulnerability to stress. We believe that this review can shed light on how diverse molecular mechanisms, involving specific receptors, neurotrophic factors, epigenetic enzymes, and miRNAs, among others, within these brain cell subtypes, are associated with the expression of a stress-susceptible or resilient phenotype, advancing the understanding/knowledge on the specific machinery implicate in those events.
Collapse
Affiliation(s)
- Cristiane Aparecida Favoretto
- Molecular and Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Marco Pagliusi
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Gessynger Morais-Silva
- Laboratory of Pharmacology, Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| |
Collapse
|
20
|
Adzic M, Lukic I, Mitic M, Glavonic E, Dragicevic N, Ivkovic S. Contribution of the opioid system to depression and to the therapeutic effects of classical antidepressants and ketamine. Life Sci 2023:121803. [PMID: 37245840 DOI: 10.1016/j.lfs.2023.121803] [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: 03/31/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Major depressive disorder (MDD) afflicts approximately 5 % of the world population, and about 30-50 % of patients who receive classical antidepressant medications do not achieve complete remission (treatment resistant depressive patients). Emerging evidence suggests that targeting opioid receptors mu (MOP), kappa (KOP), delta (DOP), and the nociceptin/orphanin FQ receptor (NOP) may yield effective therapeutics for stress-related psychiatric disorders. As depression and pain exhibit significant overlap in their clinical manifestations and molecular mechanisms involved, it is not a surprise that opioids, historically used to alleviate pain, emerged as promising and effective therapeutic options in the treatment of depression. The opioid signaling is dysregulated in depression and numerous preclinical studies and clinical trials strongly suggest that opioid modulation can serve as either an adjuvant or even an alternative to classical monoaminergic antidepressants. Importantly, some classical antidepressants require the opioid receptor modulation to exert their antidepressant effects. Finally, ketamine, a well-known anesthetic whose extremely efficient antidepressant effects were recently discovered, was shown to mediate its antidepressant effects via the endogenous opioid system. Thus, although opioid system modulation is a promising therapeutical venue in the treatment of depression further research is warranted to fully understand the benefits and weaknesses of such approach.
Collapse
Affiliation(s)
- Miroslav Adzic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| | - Iva Lukic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milos Mitic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Emilija Glavonic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Nina Dragicevic
- Department of Pharmacy, Singidunum University, Belgrade, Serbia
| | - Sanja Ivkovic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
21
|
Haynes RL, Trachtenberg F, Darnall R, Haas EA, Goldstein RD, Mena OJ, Krous HF, Kinney HC. Altered 5-HT2A/C receptor binding in the medulla oblongata in the sudden infant death syndrome (SIDS): Part I. Tissue-based evidence for serotonin receptor signaling abnormalities in cardiorespiratory- and arousal-related circuits. J Neuropathol Exp Neurol 2023; 82:467-482. [PMID: 37226597 PMCID: PMC10209647 DOI: 10.1093/jnen/nlad030] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023] Open
Abstract
The sudden infant death syndrome (SIDS), the leading cause of postneonatal infant mortality in the United States, is typically associated with a sleep period. Previously, we showed evidence of serotonergic abnormalities in the medulla (e.g. altered serotonin (5-HT)1A receptor binding), in SIDS cases. In rodents, 5-HT2A/C receptor signaling contributes to arousal and autoresuscitation, protecting brain oxygen status during sleep. Nonetheless, the role of 5-HT2A/C receptors in the pathophysiology of SIDS is unclear. We hypothesize that in SIDS, 5-HT2A/C receptor binding is altered in medullary nuclei that are key for arousal and autoresuscitation. Here, we report altered 5-HT2A/C binding in several key medullary nuclei in SIDS cases (n = 58) compared to controls (n = 12). In some nuclei the reduced 5-HT2A/C and 5-HT1A binding overlapped, suggesting abnormal 5-HT receptor interactions. The data presented here (Part 1) suggest that a subset of SIDS is due in part to abnormal 5-HT2A/C and 5-HT1A signaling across multiple medullary nuclei vital for arousal and autoresuscitation. In Part II to follow, we highlight 8 medullary subnetworks with altered 5-HT receptor binding in SIDS. We propose the existence of an integrative brainstem network that fails to facilitate arousal and/or autoresuscitation in SIDS cases.
Collapse
Affiliation(s)
- Robin L Haynes
- CJ Murphy Laboratory for SIDS Research, Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Robert’s Program on Sudden Unexpected Death in Pediatrics, Division of General Pediatrics, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
| | | | - Ryan Darnall
- CJ Murphy Laboratory for SIDS Research, Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Elisabeth A Haas
- Department of Research, Rady Children’s Hospital, San Diego, California, USA
| | - Richard D Goldstein
- Robert’s Program on Sudden Unexpected Death in Pediatrics, Division of General Pediatrics, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Othon J Mena
- San Diego County Medical Examiner Office, San Diego, California, USA
| | - Henry F Krous
- University of California, San Diego, San Diego, California, USA
- Rady Children’s Hospital, San Diego, California, USA
| | - Hannah C Kinney
- CJ Murphy Laboratory for SIDS Research, Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Robert’s Program on Sudden Unexpected Death in Pediatrics, Division of General Pediatrics, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
22
|
Li HQ, Jiang W, Ling L, Gupta V, Chen C, Pratelli M, Godavarthi SK, Spitzer NC. Generalized fear following acute stress is caused by change in co-transmitter identity of serotonergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.10.540268. [PMID: 37214936 PMCID: PMC10197626 DOI: 10.1101/2023.05.10.540268] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Overgeneralization of fear to harmless situations is a core feature of anxiety disorders resulting from acute stress, yet the mechanisms by which fear becomes generalized are poorly understood. Here we show that generalized fear in mice in response to footshock results from a transmitter switch from glutamate to GABA in serotonergic neurons of the lateral wings of the dorsal raphe. We observe a similar change in transmitter identity in the postmortem brains of PTSD patients. Overriding the transmitter switch in mice using viral tools prevents the acquisition of generalized fear. Corticosterone release and activation of glucocorticoid receptors trigger the switch, and prompt antidepressant treatment blocks the co-transmitter switch and generalized fear. Our results provide new understanding of the plasticity involved in fear generalization.
Collapse
Affiliation(s)
- Hui-Quan Li
- Neurobiology Department and Kavli Institute for Brain and Mind, University of California, San Diego
| | - Wuji Jiang
- Neurobiology Department and Kavli Institute for Brain and Mind, University of California, San Diego
| | - Lily Ling
- Neurobiology Department and Kavli Institute for Brain and Mind, University of California, San Diego
| | - Vaidehi Gupta
- Neurobiology Department and Kavli Institute for Brain and Mind, University of California, San Diego
| | - Cong Chen
- Department of Cellular and Molecular Medicine, University of California, San Diego
| | - Marta Pratelli
- Neurobiology Department and Kavli Institute for Brain and Mind, University of California, San Diego
| | - Swetha K Godavarthi
- Neurobiology Department and Kavli Institute for Brain and Mind, University of California, San Diego
| | - Nicholas C Spitzer
- Neurobiology Department and Kavli Institute for Brain and Mind, University of California, San Diego
| |
Collapse
|
23
|
Lemes JA, Silva MSCF, Gonçalves BSM, Céspedes IC, Viana MB. Deep Brain Stimulation of the dorsal raphe induces anxiolytic and panicolytic-like effects and alters serotonin immunoreactivity. Behav Brain Res 2023; 449:114462. [PMID: 37121276 DOI: 10.1016/j.bbr.2023.114462] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/02/2023]
Abstract
Previously we showed that Deep Brain Stimulation (DBS) of the dorsal region (DRD) and of the lateral wings of the dorsal raphe (lwDR) respectively decreases anxiety and panic-like responses in the elevated T-maze (ETM). This study investigates neurobiological alterations which might respond for these behavioral effects. Male Wistar rats were submitted to high-frequency stimulation (100µA, 100Hz) of the DRD or of the lwDR for 1h, and subsequently tested in the avoidance or escape tasks of the ETM. Since serotonin (5-HT) reuptake inhibitors are first line pharmacological treatment for anxiety disorders, we also tested the effects of chronic fluoxetine administration (10mg/kg, IP, 21 days) on a separate group of rats. An open field was used for locomotor activity assessment. Additionally, we evaluated c-Fos immunoreactivity (Fos-ir) in serotonergic cells of the dorsal raphe (DR). Results showed that DBS of the DRD decreases avoidance reactions, an anxiolytic-like effect, without altering escape or locomotor activity. Both fluoxetine and DBS of the lwDR decreased escape responses in the ETM, a panicolytic-like effect, without altering avoidance measurements or locomotor activity. While DBS of the DRD decreased double immunostaining in the DRD, DBS of the lwDR increased Fos-ir and double immunostaining in the DRD and lwDR. Fluoxetine also increased double immunostaining in the lwDR and in the DRV but decreased it in the DRD. These results suggest that both the anxiolytic and panicolytic-like effects of DBS and fluoxetine are related to 5-HT modulation in different subnuclei of the DR.
Collapse
Affiliation(s)
- J A Lemes
- Departamento de Biociências, Federal University of São Paulo (UNIFESP), Santos, Brazil
| | - M S C F Silva
- Departamento de Biociências, Federal University of São Paulo (UNIFESP), Santos, Brazil
| | - B S M Gonçalves
- Departamento de Biociências, Federal University of São Paulo (UNIFESP), Santos, Brazil
| | - I C Céspedes
- Departamento de Morfologia e Genética, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - M B Viana
- Departamento de Biociências, Federal University of São Paulo (UNIFESP), Santos, Brazil.
| |
Collapse
|
24
|
The Role of α-Synuclein in the Regulation of Serotonin System: Physiological and Pathological Features. Biomedicines 2023; 11:biomedicines11020541. [PMID: 36831077 PMCID: PMC9953742 DOI: 10.3390/biomedicines11020541] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
In patients affected by Parkinson's disease (PD), up to 50% of them experience cognitive changes, and psychiatric disturbances, such as anxiety and depression, often precede the onset of motor symptoms and have a negative impact on their quality of life. Pathologically, PD is characterized by the loss of dopamine (DA) neurons in the substantia nigra pars compacta (SNc) and the presence of intracellular inclusions, called Lewy bodies and Lewy neurites, composed mostly of α-synuclein (α-Syn). Much of PD research has focused on the role of α-Syn aggregates in the degeneration of SNc DA neurons due to the impact of striatal DA deficits on classical motor phenotypes. However, abundant Lewy pathology is also found in other brain regions including the midbrain raphe nuclei, which may contribute to non-motor symptoms. Indeed, dysfunction of the serotonergic (5-HT) system, which regulates mood and emotional pathways, occurs during the premotor phase of PD. However, little is known about the functional consequences of α-Syn inclusions in this neuronal population other than DA neurons. Here, we provide an overview of the current knowledge of α-Syn and its role in regulating the 5-HT function in health and disease. Understanding the relative contributions to α-Syn-linked alterations in the 5-HT system may provide a basis for identifying PD patients at risk for developing depression and could lead to a more targeted therapeutic approach.
Collapse
|
25
|
Ma J, Wang R, Chen Y, Wang Z, Dong Y. 5-HT attenuates chronic stress-induced cognitive impairment in mice through intestinal flora disruption. J Neuroinflammation 2023; 20:23. [PMID: 36737776 PMCID: PMC9896737 DOI: 10.1186/s12974-023-02693-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 01/03/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The microbiota-gut-brain axis plays an important role in the development of depression. The aim of this study was to investigate the effects of 5-HT on cognitive function, learning and memory induced by chronic unforeseeable mild stress stimulation (CUMS) in female mice. CUMS mice and TPH2 KO mice were used in the study. Lactococcus lactis E001-B-8 fungus powder was orally administered to mice with CUMS. METHODS We used the open field test, Morris water maze, tail suspension test and sucrose preference test to examine learning-related behaviours. In addition, AB-PAS staining, immunofluorescence, ELISA, qPCR, Western blotting and microbial sequencing were employed to address our hypotheses. RESULTS The effect of CUMS was more obvious in female mice than in male mice. Compared with female CUMS mice, extracellular serotonin levels in TPH2 KO CUMS mice were significantly reduced, and cognitive dysfunction was aggravated. Increased hippocampal autophagy levels, decreased neurotransmitter levels, reduced oxidative stress damage, increased neuroinflammatory responses and disrupted gut flora were observed. Moreover, L. lactis E001-B-8 significantly improved the cognitive behaviour of mice. CONCLUSIONS These results strongly suggest that L. lactis E001-B-8 but not FLX can alleviate rodent depressive and anxiety-like behaviours in response to CUMS, which is associated with the improvement of 5-HT metabolism and modulation of the gut microbiome composition.
Collapse
Affiliation(s)
- Junxing Ma
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Yaoxing Chen
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Zixu Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Yulan Dong
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
26
|
Lages YV, Balthazar L, Krahe TE, Landeira-Fernandez J. Pharmacological and Physiological Correlates of the Bidirectional Fear Phenotype of the Carioca Rats and Other Bidirectionally Selected Lines. Curr Neuropharmacol 2023; 21:1864-1883. [PMID: 36237160 PMCID: PMC10514533 DOI: 10.2174/1570159x20666221012121534] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 08/08/2022] [Accepted: 09/02/2022] [Indexed: 11/22/2022] Open
Abstract
The Carioca rat lines originated from the selective bidirectional breeding of mates displaying extreme defense responses to contextual conditioned fear. After three generations, two distinct populations could be distinguished: the Carioca High- and Low-conditioned Freezing rats, CHF, and CLF, respectively. Later studies identified strong anxiety-like behaviors in the CHF line, while indications of impulsivity and hyperactivity were prominent in the CLF animals. The present review details the physiological and pharmacological-related findings obtained from these lines. The results discussed here point towards a dysfunctional fear circuitry in CHF rats, including alterations in key brain structures and the serotoninergic system. Moreover, data from these animals highlight important alterations in the stress-processing machinery and its associated systems, such as energy metabolism and antioxidative defense. Finally, evidence of an alteration in the dopaminergic pathway in CLF rats is also debated. Thus, accumulating data gathered over the years, place the Carioca lines as significant animal models for the study of psychiatric disorders, especially fear-related ones like anxiety.
Collapse
Affiliation(s)
- Yury V. Lages
- Department of Psychology, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Laura Balthazar
- Department of Psychology, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Physiological Sciences, Laboratory of Neurophysiology, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thomas. E. Krahe
- Department of Psychology, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - J. Landeira-Fernandez
- Department of Psychology, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
27
|
Süß ST, Olbricht LM, Herlitze S, Spoida K. Constitutive 5-HT2C receptor knock-out facilitates fear extinction through altered activity of a dorsal raphe-bed nucleus of the stria terminalis pathway. Transl Psychiatry 2022; 12:487. [PMID: 36402746 PMCID: PMC9675804 DOI: 10.1038/s41398-022-02252-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/24/2022] [Accepted: 11/07/2022] [Indexed: 11/21/2022] Open
Abstract
Serotonin 2C receptors (5-HT2CRs) are widely distributed throughout the brain and are strongly implicated in the pathophysiology of anxiety disorders such as post-traumatic stress disorder (PTSD). Although in recent years, a considerable amount of evidence supports 5-HT2CRs facilitating effect on anxiety behavior, the involvement in learned fear responses and fear extinction is rather unexplored. Here, we used a 5-HT2CR knock-out mouse line (2CKO) to gain new insights into the involvement of 5-HT2CRs in the neuronal fear circuitry. Using a cued fear conditioning paradigm, our results revealed that global loss of 5-HT2CRs exclusively accelerates fear extinction, without affecting fear acquisition and fear expression. To investigate the neuronal substrates underlying the extinction enhancing effect, we mapped the immediate-early gene product cFos, a marker for neuronal activity, in the dorsal raphe nucleus (DRN), amygdala and bed nucleus of the stria terminalis (BNST). Surprisingly, besides extinction-associated changes, our results revealed alterations in neuronal activity even under basal home cage conditions in specific subregions of the DRN and the BNST in 2CKO mice. Neuronal activity in the dorsal BNST was shifted in an extinction-supporting direction due to 5-HT2CR knock-out. Finally, the assessment of DRN-BNST connectivity using antero- and retrograde tracing techniques uncovered a discrete serotonergic pathway projecting from the most caudal subregion of the DRN (DRC) to the anterodorsal portion of the BNST (BNSTad). This serotonergic DRC-BNSTad pathway showed increased neuronal activity in 2CKO mice. Thus, our results provide new insights for the fear extinction network by revealing a specific serotonergic DRC-BNSTad pathway underlying a 5-HT2CR-sensitive mechanism with high significance in the treatment of PTSD.
Collapse
Affiliation(s)
- Sandra T Süß
- Department of General Zoology and Neurobiology, ND7/31, Ruhr-University Bochum, Universitätsstr. 150, D-44780, Bochum, Germany.
| | - Linda M Olbricht
- Department of General Zoology and Neurobiology, ND7/31, Ruhr-University Bochum, Universitätsstr. 150, D-44780, Bochum, Germany
| | - Stefan Herlitze
- Department of General Zoology and Neurobiology, ND7/31, Ruhr-University Bochum, Universitätsstr. 150, D-44780, Bochum, Germany
| | - Katharina Spoida
- Department of General Zoology and Neurobiology, ND7/31, Ruhr-University Bochum, Universitätsstr. 150, D-44780, Bochum, Germany.
| |
Collapse
|
28
|
Leschik J, Gentile A, Cicek C, Péron S, Tevosian M, Beer A, Radyushkin K, Bludau A, Ebner K, Neumann I, Singewald N, Berninger B, Lessmann V, Lutz B. Brain-derived neurotrophic factor expression in serotonergic neurons improves stress resilience and promotes adult hippocampal neurogenesis. Prog Neurobiol 2022; 217:102333. [PMID: 35872219 DOI: 10.1016/j.pneurobio.2022.102333] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 06/24/2022] [Accepted: 07/19/2022] [Indexed: 11/19/2022]
Abstract
The neurotrophin brain-derived neurotrophic factor (BDNF) stimulates adult neurogenesis, but also influences structural plasticity and function of serotonergic neurons. Both, BDNF/TrkB signaling and the serotonergic system modulate behavioral responses to stress and can lead to pathological states when dysregulated. The two systems have been shown to mediate the therapeutic effect of antidepressant drugs and to regulate hippocampal neurogenesis. To elucidate the interplay of both systems at cellular and behavioral levels, we generated a transgenic mouse line that overexpresses BDNF in serotonergic neurons in an inducible manner. Besides displaying enhanced hippocampus-dependent contextual learning, transgenic mice were less affected by chronic social defeat stress (CSDS) compared to wild-type animals. In parallel, we observed enhanced serotonergic axonal sprouting in the dentate gyrus and increased neural stem/progenitor cell proliferation, which was uniformly distributed along the dorsoventral axis of the hippocampus. In the forced swim test, BDNF-overexpressing mice behaved similarly as wild-type mice treated with the antidepressant fluoxetine. Our data suggest that BDNF released from serotonergic projections exerts this effect partly by enhancing adult neurogenesis. Furthermore, independently of the genotype, enhanced neurogenesis positively correlated with the social interaction time after the CSDS, a measure for stress resilience.
Collapse
Affiliation(s)
- Julia Leschik
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany.
| | - Antonietta Gentile
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany; Department of Systems Medicine, Tor Vergata University, Rome 00183, Italy
| | - Cigdem Cicek
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany; Faculty of Medicine, Department of Medical Biochemistry, Hacettepe University, 06100 Ankara, Turkey; Faculty of Medicine, Department of Medical Biochemistry, Yuksek Ihtisas University, 06520 Ankara, Turkey
| | - Sophie Péron
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany; Institute of Psychiatry, Psychology & Neuroscience, Centre for Developmental Neurobiology, King's College London, London SE11UL, United Kingdom
| | - Margaryta Tevosian
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany; Leibniz Institute for Resilience Research (LIR), Mainz 55122, Germany
| | - Annika Beer
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany; Leibniz Institute for Resilience Research (LIR), Mainz 55122, Germany
| | | | - Anna Bludau
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg 93053, Germany
| | - Karl Ebner
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck, Leopold Franzens University Innsbruck, Innsbruck 6020, Austria
| | - Inga Neumann
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg 93053, Germany
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck, Leopold Franzens University Innsbruck, Innsbruck 6020, Austria
| | - Benedikt Berninger
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany; Institute of Psychiatry, Psychology & Neuroscience, Centre for Developmental Neurobiology, King's College London, London SE11UL, United Kingdom; Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE11UL, United Kingdom
| | - Volkmar Lessmann
- Institute of Physiology, Medical Faculty, Otto-von-Guericke-University, Magdeburg 39120, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39120, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany; Leibniz Institute for Resilience Research (LIR), Mainz 55122, Germany
| |
Collapse
|
29
|
Uzungil V, Tran H, Aitken C, Wilson C, Opazo CM, Li S, Payet JM, Mawal CH, Bush AI, Hale MW, Hannan AJ, Renoir T. Novel Antidepressant-Like Properties of the Iron Chelator Deferiprone in a Mouse Model of Depression. Neurotherapeutics 2022; 19:1662-1685. [PMID: 35861925 PMCID: PMC9606181 DOI: 10.1007/s13311-022-01257-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 10/17/2022] Open
Abstract
Depressed individuals who carry the short allele for the serotonin-transporter-linked promotor region of the gene are more vulnerable to stress and have reduced response to first-line antidepressants such as selective serotonin reuptake inhibitors. Since depression severity has been reported to correlate with brain iron levels, the present study aimed to characterise the potential antidepressant properties of the iron chelator deferiprone. Using the serotonin transporter knock-out (5-HTT KO) mouse model, we assessed the behavioural effects of acute deferiprone on the Porsolt swim test (PST) and novelty-suppressed feeding test (NSFT). Brain and blood iron levels were also measured following acute deferiprone. To determine the relevant brain regions activated by deferiprone, we then measured c-Fos expression and applied network-based analyses. We found that deferiprone reduced immobility time in the PST in 5-HTT KO mice and reduced latency to feed in the NSFT in both genotypes, suggesting potential antidepressant-like effects. There was no effect on brain or blood iron levels following deferiprone treatment, potentially indicating an acute iron-independent mechanism. Deferiprone reversed the increase in c-Fos expression induced by swim stress in 5-HTT KO mice in the lateral amygdala. Functional network analyses suggest that hub regions of activity in mice treated with deferiprone include the caudate putamen and prefrontal cortex. The PST-induced increase in network modularity in wild-type mice was not observed in 5-HTT KO mice. Altogether, our data show that the antidepressant-like effects of deferiprone could be acting via an iron-independent mechanism and that these therapeutic effects are underpinned by changes in neuronal activity in the lateral amygdala.
Collapse
Affiliation(s)
- Volkan Uzungil
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Harvey Tran
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Connor Aitken
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Carey Wilson
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Carlos M Opazo
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Shanshan Li
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Jennyfer M Payet
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Celeste H Mawal
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Matthew W Hale
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Anthony J Hannan
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Thibault Renoir
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia.
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia.
| |
Collapse
|
30
|
Paquelet GE, Carrion K, Lacefield CO, Zhou P, Hen R, Miller BR. Single-cell activity and network properties of dorsal raphe nucleus serotonin neurons during emotionally salient behaviors. Neuron 2022; 110:2664-2679.e8. [PMID: 35700737 PMCID: PMC9575686 DOI: 10.1016/j.neuron.2022.05.015] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 11/23/2021] [Accepted: 05/13/2022] [Indexed: 12/20/2022]
Abstract
The serotonin system modulates a wide variety of emotional behaviors and states, including reward processing, anxiety, and social interaction. To reveal the underlying patterns of neural activity, we visualized serotonergic neurons in the dorsal raphe nucleus (DRN5-HT) of mice using miniaturized microscopy during diverse emotional behaviors. We discovered ensembles of cells with highly correlated activity and found that DRN5-HT neurons are preferentially recruited by emotionally salient stimuli as opposed to neutral stimuli. Individual DRN5-HT neurons responded to diverse combinations of salient stimuli, with some preference for valence and sensory modality. Anatomically defined subpopulations projecting to either a reward-related structure (the ventral tegmental area) or an anxiety-related structure (the bed nucleus of the stria terminalis) contained all response types but were enriched in reward- and anxiety-responsive cells, respectively. Our results suggest that the DRN serotonin system responds to emotional salience using ensembles with mixed selectivity and biases in downstream connectivity.
Collapse
Affiliation(s)
- Grace E Paquelet
- Department of Neuroscience, Columbia University, New York, NY 10032, USA
| | - Kassandra Carrion
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Clay O Lacefield
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA
| | - Pengcheng Zhou
- Department of Neuroscience, Columbia University, New York, NY 10032, USA; Department of Statistics, Columbia University, New York, NY 10027, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027
| | - René Hen
- Department of Neuroscience, Columbia University, New York, NY 10032, USA; Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA
| | - Bradley R Miller
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
31
|
Rahaman SM, Chowdhury S, Mukai Y, Ono D, Yamaguchi H, Yamanaka A. Functional Interaction Between GABAergic Neurons in the Ventral Tegmental Area and Serotonergic Neurons in the Dorsal Raphe Nucleus. Front Neurosci 2022; 16:877054. [PMID: 35663550 PMCID: PMC9160575 DOI: 10.3389/fnins.2022.877054] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/21/2022] [Indexed: 12/02/2022] Open
Abstract
GABAergic neurons in the ventral tegmental area (VTA) have brain-wide projections and are involved in multiple behavioral and physiological functions. Here, we revealed the responsiveness of Gad67+ neurons in VTA (VTAGad67+) to various neurotransmitters involved in the regulation of sleep/wakefulness by slice patch clamp recording. Among the substances tested, a cholinergic agonist activated, but serotonin, dopamine and histamine inhibited these neurons. Dense VTAGad67+ neuronal projections were observed in brain areas regulating sleep/wakefulness, including the central amygdala (CeA), dorsal raphe nucleus (DRN), and locus coeruleus (LC). Using a combination of electrophysiology and optogenetic studies, we showed that VTAGad67+ neurons inhibited all neurons recorded in the DRN, but did not inhibit randomly recorded neurons in the CeA and LC. Further examination revealed that the serotonergic neurons in the DRN (DRN5–HT) were monosynaptically innervated and inhibited by VTAGad67+ neurons. All recorded DRN5–HT neurons received inhibitory input from VTAGad67+ neurons, while only one quarter of them received inhibitory input from local GABAergic neurons. Gad67+ neurons in the DRN (DRNGad67+) also received monosynaptic inhibitory input from VTAGad67+ neurons. Taken together, we found that VTAGad67+ neurons were integrated in many inputs, and their output inhibits DRN5–HT neurons, which may regulate physiological functions including sleep/wakefulness.
Collapse
Affiliation(s)
- Sheikh Mizanur Rahaman
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Srikanta Chowdhury
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Yasutaka Mukai
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Yamaguchi
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
- *Correspondence: Akihiro Yamanaka,
| |
Collapse
|
32
|
de Kort AR, Joosten EAJ, Patijn J, Tibboel D, van den Hoogen NJ. The development of descending serotonergic modulation of the spinal nociceptive network: a life span perspective. Pediatr Res 2022; 91:1361-1369. [PMID: 34257402 DOI: 10.1038/s41390-021-01638-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/10/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023]
Abstract
The nociceptive network, responsible for transmission of nociceptive signals that generate the pain experience, is not fully developed at birth. Descending serotonergic modulation of spinal nociception, an important part of the pain network, undergoes substantial postnatal maturation and is suggested to be involved in the altered pain response observed in human newborns. This review summarizes preclinical data of the development of descending serotonergic modulation of the spinal nociceptive network across the life span, providing a comprehensive background to understand human newborn pain experience and treatment. Sprouting of descending serotonergic axons, originating from the rostroventral medulla, as well as changes in receptor function and expression take place in the first postnatal weeks of rodents, corresponding to human neonates in early infancy. Descending serotonergic modulation switches from facilitation in early life to bimodal control in adulthood, masking an already functional 5-HT inhibitory system at early ages. Specifically the 5-HT3 and 5-HT7 receptors seem distinctly important for pain facilitation at neonatal and early infancy, while the 5-HT1a, 5-HT1b, and 5-HT2 receptors mediate inhibitory effects at all ages. Analgesic therapy that considers the neurodevelopmental phase is likely to result in a more targeted treatment of neonatal pain and may improve both short- and long-term effects. IMPACT: The descending serotonergic system undergoes anatomical changes from birth to early infancy, as its sprouts and descending projections increase and the dorsal horn innervation pattern changes. Descending serotonergic modulation from the rostral ventral medulla switches from facilitation in early life via the 5-HT3 and 5-HT7 receptors to bimodal control in adulthood. A functional inhibitory serotonergic system mainly via 5-HT1a, 5-HT1b, and 5-HT2a receptors at the spinal level exists already at the neonatal phase but is masked by descending facilitation.
Collapse
Affiliation(s)
- Anne R de Kort
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre+, Maastricht, the Netherlands. .,Department of Translational Neuroscience, School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Elbert A J Joosten
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre+, Maastricht, the Netherlands.,Department of Translational Neuroscience, School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Jacob Patijn
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre+, Maastricht, the Netherlands.,Department of Translational Neuroscience, School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Dick Tibboel
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Nynke J van den Hoogen
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre+, Maastricht, the Netherlands.,Department of Translational Neuroscience, School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Department of Comparative Biology and Experimental Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
33
|
Batistela MF, Vilela-Costa HH, Frias AT, Hernandes PM, Lovick TA, Zangrossi H. Enhanced responsiveness to hypoxic panicogenic challenge in female rats in late diestrus is suppressed by short-term, low-dose fluoxetine: Involvement of the dorsal raphe nucleus and the dorsal periaqueductal gray. J Psychopharmacol 2021; 35:1523-1535. [PMID: 34872406 DOI: 10.1177/02698811211058986] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Acute hypoxia, which is panicogenic in humans, also evokes panic-like behavior in male rats. Panic disorder is more common in women and susceptibility increases during the premenstrual phase of the cycle. AIMS We here investigated for the first time the impact of hypoxia on the expression of panic-like escape behavior by female rats and its relationship with the estrous cycle. We also evaluated functional activation of the midbrain panic circuitry in response to this panicogenic stimulus and whether short-term, low-dose fluoxetine treatment inhibits the hyper-responsiveness of females in late diestrus. METHODS Male and female Sprague Dawley rats were exposed to 7% O2. Females in late diestrus were also tested after short-term treatment with fluoxetine (1.75 or 10 mg/kg, i.p.). Brains were harvested and processed for c-Fos and tryptophan hydroxylase immunoreactivity in the periaqueductal gray matter (PAG) and dorsal raphe nucleus (DR). RESULTS Acute hypoxia evoked escape in both sexes. Overall, females were more responsive than males and this is clearer in late diestrus phase. In both sexes, hypoxia induced functional activation (c-Fos expression) in non-serotonergic cells in the lateral wings of the DR and dorsomedial PAG, which was greater in late diestrus than proestrus (lowest behavioral response to hypoxia). Increased responding in late diestrus (behavioral and cellular levels) was prevented by 1.75, but not 10 mg/kg fluoxetine. DISCUSSION The response of female rats to acute hypoxia models panic behavior in women. Low-dose fluoxetine administered in the premenstrual phase deserves further attention for management of panic disorders in women.
Collapse
Affiliation(s)
- Matheus F Batistela
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Heloísa H Vilela-Costa
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Alana T Frias
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Paloma M Hernandes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Thelma A Lovick
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Helio Zangrossi
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
34
|
Moriya R, Kanamaru M, Okuma N, Yoshikawa A, Tanaka KF, Hokari S, Ohshima Y, Yamanaka A, Honma M, Onimaru H, Kikuchi T, Izumizaki M. Optogenetic activation of DRN 5-HT neurons induced active wakefulness, not quiet wakefulness. Brain Res Bull 2021; 177:129-142. [PMID: 34563634 DOI: 10.1016/j.brainresbull.2021.09.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 11/16/2022]
Abstract
There has been a long-standing controversy regarding the physiological role of serotonin (5-HT) neurons in the dorsal raphe nucleus (DRN) in sleep/wake architecture. Some studies have reported that 5-HT acts as a sleep-promoting agent, but several studies have suggested that DRN 5-HT neurons function predominantly to promote wakefulness and inhibit rapid eye movement (REM) sleep. Furthermore, recent studies have reported that there is a clear neurobiological difference between a waking state that includes alertness and active exploration (i.e., active wakefulness) and a waking state that is devoid of locomotion (i.e., quiet wakefulness). These states have also been shown to differ clinically in terms of memory consolidation. However, the effects of 5-HT neurons on the regulation of these two different waking states have not been fully elucidated. In the present study, we attempted to examine the physiological role of DRN 5-HT neurons in various sleep/wake states using optogenetic methods that allowed manipulation of cell-type specific neuronal activation with high temporal and anatomical precision. We crossed TPH2-tTA and TetO-ChR2(C128S) mice to obtain mice with channelrhodopsin-2 (ChR2) [C128S]-expressing central 5-HT neurons, and we activated DRN-5HT neurons or medullary 5-HT neurons. Optogenetic activation of DRN 5-HT neurons caused rapid transition from non-REM sleep to active wakefulness, not quiet wakefulness, whereas activation of medullary 5-HT neurons did not appear to affect sleep/wake states or locomotor activity. Our results may shed light on the physiological role of DRN 5-HT neurons in sleep/wake architecture and encourage further investigations of the cortical functional connectivity involved in sleep/wake state regulation.
Collapse
Affiliation(s)
- Rika Moriya
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 757-1 Asahimachi-dori, Chuo-ku, Niigata, Niigata 951-8520, Japan
| | - Mitsuko Kanamaru
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Naoki Okuma
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Akira Yoshikawa
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Kenji F Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo 160-8582, Japan
| | - Satoshi Hokari
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 757-1 Asahimachi-dori, Chuo-ku, Niigata, Niigata 951-8520, Japan
| | - Yasuyoshi Ohshima
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 757-1 Asahimachi-dori, Chuo-ku, Niigata, Niigata 951-8520, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Motoyasu Honma
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Hiroshi Onimaru
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Toshiaki Kikuchi
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 757-1 Asahimachi-dori, Chuo-ku, Niigata, Niigata 951-8520, Japan
| | - Masahiko Izumizaki
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
| |
Collapse
|
35
|
Alexander C, Vasefi M. Cannabidiol and the corticoraphe circuit in post-traumatic stress disorder. IBRO Neurosci Rep 2021; 11:88-102. [PMID: 34485973 PMCID: PMC8408530 DOI: 10.1016/j.ibneur.2021.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/04/2021] [Accepted: 08/18/2021] [Indexed: 01/06/2023] Open
Abstract
Post-Traumatic Stress Disorder (PTSD), characterized by re-experiencing, avoidance, negative affect, and impaired memory processing, may develop after traumatic events. PTSD is complicated by impaired plasticity and medial prefrontal cortex (mPFC) activity, hyperactivity of the amygdala, and impaired fear extinction. Cannabidiol (CBD) is a promising candidate for treatment due to its multimodal action that enhances plasticity and calms hyperexcitability. CBD’s mechanism in the mPFC of PTSD patients has been explored extensively, but literature on the mechanism in the dorsal raphe nucleus (DRN) is lacking. Following the PRISMA guidelines, we examined current literature regarding CBD in PTSD and overlapping symptomologies to propose a mechanism by which CBD treats PTSD via corticoraphe circuit. Acute CBD inhibits excess 5-HT release from DRN to amygdala and releases anandamide (AEA) onto amygdala inputs. By first reducing amygdala and DRN hyperactivity, CBD begins to ameliorate activity disparity between mPFC and amygdala. Chronic CBD recruits the mPFC, creating harmonious corticoraphe signaling. DRN releases enough 5-HT to ameliorate mPFC hypoactivity, while the mPFC continuously excites DRN 5-HT neurons via glutamate. Meanwhile, AEA regulates corticoraphe activity to stabilize signaling. AEA prevents DRN GABAergic interneurons from inhibiting 5-HT release so the DRN can assist the mPFC in overcoming its hypoactivity. DRN-mediated restoration of mPFC activity underlies CBD’s mechanism on fear extinction and learning of stress coping. CBD reduces PTSD symptoms via the DRN and corticoraphe circuit. Acute effects of CBD reduce DRN-amygdala excitatory signaling to lessen the activity disparity between amygdala and mPFC. Chronic CBD officially resolves mPFC hypoactivity by facilitating 5-HT release from DRN to mPFC. CBD-facilitated endocannabinoid signaling stabilizes DRN activity and restores mPFC inhibitory control. Chronically administered CBD acts via the corticoraphe circuit to favor fear extinction over fear memory reconsolidation.
Collapse
Key Words
- 2-AG, 2-arachidonoylglycerol
- 5-HT, Serotonin
- 5-HT1AR, 5-HT Receptor Type 1A
- 5-HT2AR, 5-HT Receptor Type 2 A
- AEA, Anandamide
- CB1R, Cannabinoid Receptor Type 1
- CB2R, Cannabinoid Receptor Type 2
- CBD, Cannabidiol
- COVID-19, SARS-CoV-2
- Cannabidiol
- DRN, Dorsal Raphe Nucleus
- ERK1/2, Extracellular Signal-Related Kinases Type 1 or Type 2
- FAAH, Fatty Acid Amide Hydrolase
- GABA, Gamma-Aminobutyric Acid
- GPCRs, G-Protein Coupled Receptors
- NMDAR, N-Methyl-D-aspartate Receptors
- PET, Positron Emission Tomography
- PFC, DRN and Raphe
- PFC, Prefrontal Cortex
- PTSD
- PTSD, Post-Traumatic Stress Disorder
- SSNRI, Selective Norepinephrine Reuptake Inhibitor
- SSRI, Selective Serotonin Reuptake Inhibitor
- Serotonin
- TRPV1, Transient Receptor Potential Vanilloid 1 Channels
- Traumatic Stress
- fMRI, Functional Magnetic Resonance Imaging
- mPFC, Medial Prefrontal Cortex
Collapse
Affiliation(s)
- Claire Alexander
- Department of Biology, Lamar University, Beaumont, TX 77710, USA
| | - Maryam Vasefi
- Department of Biology, Lamar University, Beaumont, TX 77710, USA
| |
Collapse
|
36
|
Makrygianni EA, Chrousos GP. From Brain Organoids to Networking Assembloids: Implications for Neuroendocrinology and Stress Medicine. Front Physiol 2021; 12:621970. [PMID: 34177605 PMCID: PMC8222922 DOI: 10.3389/fphys.2021.621970] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 04/19/2021] [Indexed: 12/13/2022] Open
Abstract
Brain organoids are three-dimensional cultures that contain multiple types of cells and cytoarchitectures, and resemble fetal human brain structurally and functionally. These organoids are being used increasingly to model brain development and disorders, however, they only partially recapitulate such processes, because of several limitations, including inability to mimic the distinct cortical layers, lack of functional neuronal circuitry as well as non-neural cells and gyrification, and increased cellular stress. Efforts to create improved brain organoid culture systems have led to region-specific organoids, vascularized organoids, glia-containing organoids, assembloids, sliced organoids and polarized organoids. Assembloids are fused region-specific organoids, which attempt to recapitulate inter-regional and inter-cellular interactions as well as neural circuitry development by combining multiple brain regions and/or cell lineages. As a result, assembloids can be used to model subtle functional aberrations that reflect complex neurodevelopmental, neuropsychiatric and neurodegenerative disorders. Mammalian organisms possess a highly complex neuroendocrine system, the stress system, whose main task is the preservation of systemic homeostasis, when the latter is threatened by adverse forces, the stressors. The main central parts of the stress system are the paraventricular nucleus of the hypothalamus and the locus caeruleus/norepinephrine-autonomic nervous system nuclei in the brainstem; these centers innervate each other and interact reciprocally as well as with various other CNS structures. Chronic dysregulation of the stress system has been implicated in major pathologies, the so-called chronic non-communicable diseases, including neuropsychiatric, neurodegenerative, cardiometabolic and autoimmune disorders, which lead to significant population morbidity and mortality. We speculate that brain organoids and/or assembloids could be used to model the development, regulation and dysregulation of the stress system and to better understand stress-related disorders. Novel brain organoid technologies, combined with high-throughput single-cell omics and gene editing, could, thus, have major implications for precision medicine.
Collapse
Affiliation(s)
- Evanthia A Makrygianni
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Center for Adolescent Medicine and UNESCO Chair on Adolescent Health Care, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, Athens, Greece
| |
Collapse
|
37
|
Lonstein JS, Linning-Duffy K, Tang Y, Moody A, Yan L. Impact of daytime light intensity on the central orexin (hypocretin) system of a diurnal rodent (Arvicanthis niloticus). Eur J Neurosci 2021; 54:4167-4181. [PMID: 33899987 DOI: 10.1111/ejn.15248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 11/29/2022]
Abstract
The neuropeptide orexin/hypocretin is implicated in sleep and arousal, energy expenditure, reward, affective state and cognition. Our previous work using diurnal Nile grass rats (Arvicanthis niloticus) found that orexin mediates the effects of environmental light, particularly daytime light intensity, on affective and cognitive behaviours. The present study further investigated how daytime light intensity affects the central orexin system in male and female grass rats. Subjects were housed for 4 weeks in 12:12 hr dim light:dark (50 lux, dimLD) or in 12:12 hr bright light:dark cycle (1000 lux, brightLD). Day/night fluctuations in some orexin measures were also assessed. Despite similar hypothalamic prepro-orexin mRNA expression across all conditions, there were significantly more orexin-immunoreactive neurons, larger somata, greater optical density or higher orexin A content at night (ZT14) than during the day (ZT2), and/or in animals housed in brightLD compared to dimLD. Grass rats in brightLD also had higher cisternal CSF levels of orexin A. Furthermore, orexin receptor OX1R and OX2R proteins in the medial prefrontal cortex were higher in brightLD than dimLD males, but lower in brightLD than dimLD females. In the CA1 and dorsal raphe nucleus, females had higher OX1R than males without any significant effects of light condition, and OX2R levels were unaffected by sex or light. These results reveal that daytime light intensity alters the central orexin system of both male and female diurnal grass rats, sometimes sex-specifically, and provides insight into the mechanisms underlying how daytime light intensity impacts orexin-regulated functions.
Collapse
Affiliation(s)
- Joseph S Lonstein
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Katrina Linning-Duffy
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Yuping Tang
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Anna Moody
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Lily Yan
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
38
|
Payet JM, Wilson KE, Russo AM, Angiolino A, Kavanagh-Ryan W, Kent S, Lowry CA, Hale MW. Involvement of dorsal raphe nucleus serotonergic systems in social approach-avoidance behaviour and in the response to fluoxetine treatment in peri-adolescent female BALB/c mice. Behav Brain Res 2021; 408:113268. [PMID: 33811952 DOI: 10.1016/j.bbr.2021.113268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/03/2021] [Accepted: 03/26/2021] [Indexed: 12/19/2022]
Abstract
Serotonergic systems are involved in the development and regulation of social behaviour, and drugs that target serotonin neurotransmission, such as selective serotonin reuptake inhibitors (SSRIs), also alter aspects of social approach-avoidance. The midbrain dorsal raphe nucleus (DR), which is a major serotonergic nucleus and main source of serotonergic innervation of the forebrain, has been proposed as an important target for SSRIs, although evidence in females is lacking. In this study, we examined the involvement of the DR serotonergic systems in social behaviour and in response to SSRI treatment, using peri-adolescent female BALB/c mice. Mice were exposed to the SSRI fluoxetine either chronically (18 mg/kg/day, in drinking water, for 12 days) or acutely (18 mg/kg, i.p.), or to vehicle control condition (0.9 % saline, i.p.), prior to being exposed to the three-chambered sociability test. Activation of serotonergic neurons across subregions of the DR were subsequently measured, using dual-label immunohistochemistry for TPH2 and c-Fos. Acute fluoxetine administration increased generalised and social avoidance, while mice exposed to chronic fluoxetine treatment showed levels of social approach behaviour that were comparable to controls. Serotonergic populations across the DR showed reduced activity following acute fluoxetine treatment. Further, activation of serotonergic neurons in the ventral DR correlated with social approach behaviour in vehicle-treated control mice. These data provide some support for the involvement of discrete populations of DR serotonergic neurons in the regulation of social approach-avoidance, although more research is needed to understand the effects and mechanisms of chronic SSRI treatment in females.
Collapse
Affiliation(s)
- Jennyfer M Payet
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Kira-Elise Wilson
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Adrian M Russo
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Anthony Angiolino
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - William Kavanagh-Ryan
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Stephen Kent
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Christopher A Lowry
- Department of Integrative Physiology and Center for Neuroscience, 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
|
39
|
Sepulveda M, Manning EE, Gogos A, Hale M, van den Buuse M. Long-term effects of young-adult methamphetamine on dorsal raphe serotonin systems in mice: Role of brain-derived neurotrophic factor. Brain Res 2021; 1762:147428. [PMID: 33737066 DOI: 10.1016/j.brainres.2021.147428] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/15/2021] [Accepted: 03/08/2021] [Indexed: 01/21/2023]
Abstract
To assess the long-term effects of chronic adolescent methamphetamine (METH) treatment on the serotonin system in the brain, we used serotonin-1A receptor (5-HT1A) and serotonin transporter (SERT) autoradiography, and quantitative tryptophan-hydroxylase 2 (TPH2) immunohistochemistry in the raphe nuclei of mice. Because of the modulatory role of brain-derived neurotrophic factor (BDNF) on the serotonin system and the effects of METH, we included both BDNF heterozygous (HET) mice and wildtype (WT) controls. Male and female mice of both genotypes were treated with an escalating METH dose regimen from the age of 6-9 weeks. At least two weeks later, acute locomotor hyperactivity induced by a 5 mg/kg D-amphetamine challenge was significantly enhanced in METH-pretreated mice, showing long-term sensitisation. METH pretreatment caused a small, but significant decrease of 5-HT1A receptor binding in the dorsal raphe nucleus (DRN) of males independent of genotype, but there were no changes in the median raphe nucleus (MRN) or in SERT binding density. METH treatment reduced the number of TPH2 positive cells in ventral subregions of the rostral and medial DRN independent of genotype. METH treatment selectively reduced DRN cell counts in BDNF HET mice compared to wildtype mice in medial and caudal ventrolateral subregions previously associated with panic-like behaviour. The data increase our understanding of the long-term and selective effects of METH on brain serotonin systems. These findings could be relevant for some of the psychosis-like symptoms associated with long-term METH use.
Collapse
Affiliation(s)
- Mauricio Sepulveda
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Australia
| | - Elizabeth E Manning
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Australia
| | - Andrea Gogos
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Australia
| | - Matthew Hale
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia; Department of Pharmacology, University of Melbourne, Australia; College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia.
| |
Collapse
|
40
|
Dos Anjos Rosário B, de Fátima Santana de Nazaré M, Lemes JA, de Andrade JS, da Silva RB, Pereira CDS, Ribeiro DA, de Barros Viana M. Repeated crack cocaine administration alters panic-related responses and delta FosB immunoreactivity in panic-modulating brain regions. Exp Brain Res 2021; 239:1179-1191. [PMID: 33569614 DOI: 10.1007/s00221-020-06031-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/30/2020] [Indexed: 01/09/2023]
Abstract
Crack cocaine is the crystal form of cocaine, produced by adding sodium bicarbonate to cocaine base paste. Brazil is the largest consumer of crack cocaine in the world. Users of crack cocaine show important physiological and behavioral alterations, including neuropsychiatric symptoms, such as anxiety-related symptoms. Nevertheless, few pre-clinical studies have been previously performed to understand the neurobiological effects of crack cocaine. The purpose of the present study was to investigate effects of the subchronic treatment (5 days, IP) of rats with crack cocaine in an animal model of anxiety/panic, the elevated T-maze (ETM). The ETM model allows the measurement of two behavioral defensive responses, avoidance and escape, in clinical terms, respectively, associated to generalized anxiety and panic disorder, the two main psychiatric conditions that accompany substance use disorders. Immediately after the ETM model, animals were tested in an open field for locomotor activity assessment. Analysis of delta FosB protein immunoreactivity was used to map areas activated by crack cocaine exposure. Results showed that crack treatment selectively altered escape displayed by rats in the ETM test, inducing either a panicolytic (18 mg/kg IP) or a panicogenic-like effect (25 and 36 mg/kg IP). These effects were followed by the altered functioning of panic-modulating brain regions, i.e., the periaqueductal gray and the dorsal region and lateral wings of the dorsal raphe nucleus. Treatment with 36 mg/kg of crack cocaine also increased locomotor activity. These are the first observations performed with crack cocaine in a rodent model of anxiety/panic and contribute to a better understanding of the behavioral and neurobiological effects of crack cocaine.
Collapse
Affiliation(s)
| | | | - Jéssica Alves Lemes
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos, SP, Brazil
| | - José Simões de Andrade
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos, SP, Brazil
| | - Regina Barbosa da Silva
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos, SP, Brazil
| | | | - Daniel Araki Ribeiro
- 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.
| |
Collapse
|
41
|
Fonseca R, Madeira N, Simoes C. Resilience to fear: The role of individual factors in amygdala response to stressors. Mol Cell Neurosci 2020; 110:103582. [PMID: 33346000 DOI: 10.1016/j.mcn.2020.103582] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 11/13/2020] [Accepted: 12/02/2020] [Indexed: 10/22/2022] Open
Abstract
Resilience to stress is an adaptive process that varies individually. Resilience refers to the adaptation, or the ability to maintain or regain mental health, despite being subject to adverse situation. Resilience is a dynamic concept that reflects a combination of internal individual factors, including age and gender interacting with external factors such as social, cultural and environmental factors. In the last decade, we have witnessed an increase in the prevalence of anxiety disorders, including post-traumatic stress disorder. Given that stress in unavoidable, it is of great interest to understand the neurophysiological mechanisms of resilience, the individual factors that may contribute to susceptibility and promote efficacious approaches to improve resilience. Here, we address this complex question, attempting at defining clear and operational definitions that may allow us to improve our analysis of behavior incorporating individuality. We examine how individual perception of the stressor can alter the outcome of an adverse situation using as an example, the fear-conditioning paradigm and discuss how individual differences in the reward system can contribute to resilience. Given the central role of the endocannabinoid system in regulating fear responses and anxiety, we discuss the evidence that polymorphisms in several molecules of this signaling system contribute to different anxiety phenotypes. The endocannabinoid system is highly interconnected with the serotoninergic and dopaminergic modulatory systems, contributing to individual differences in stress perception and coping mechanisms. We review how the individual variability in these modulatory systems can be used towards a multivariable assessment of stress risk. Incorporating individuality in our research will allow us to define biomarkers of anxiety disorders as well as assess prognosis, towards a personalized clinical approach to mental health.
Collapse
Affiliation(s)
- Rosalina Fonseca
- Cellular and Systems Neurobiology, Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130 1169-056 Lisboa, Portugal.
| | - Natália Madeira
- Cellular and Systems Neurobiology, Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130 1169-056 Lisboa, Portugal
| | - Carla Simoes
- Cellular and Systems Neurobiology, Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130 1169-056 Lisboa, Portugal
| |
Collapse
|
42
|
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: 12] [Impact Index Per Article: 2.4] [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
|
43
|
Ge R, Dai Y. Three-Week Treadmill Exercise Enhances Persistent Inward Currents, Facilitates Dendritic Plasticity, and Upregulates the Excitability of Dorsal Raphe Serotonin Neurons in ePet-EYFP Mice. Front Cell Neurosci 2020; 14:575626. [PMID: 33177992 PMCID: PMC7595958 DOI: 10.3389/fncel.2020.575626] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/04/2020] [Indexed: 12/14/2022] Open
Abstract
Exercise plays a key role in preventing or treating mental or motor disorders caused by dysfunction of the serotonergic system. However, the electrophysiological and ionic channel mechanisms underlying these effects remain unclear. In this study, we investigated the effects of 3-week treadmill exercise on the electrophysiological and channel properties of dorsal raphe nucleus (DRN). Serotonin (5-HT) neurons in ePet-EYFP mice, using whole-cell patch clamp recording. Treadmill exercise was induced in ePet-EYFP mice of P21–24 for 3 weeks, and whole-cell patch clamp recording was performed on EYFP-positive 5-HT neurons from DRN slices of P42–45 mice. Experiment data showed that 5-HT neurons in the DRN were a heterogeneous population with multiple firing patterns (single firing, phasic firing, and tonic firing). Persistent inward currents (PICs) with multiple patterns were expressed in 5-HT neurons and composed of Cav1.3 (Ca-PIC) and sodium (Na-PIC) components. Exercise hyperpolarized the voltage threshold for action potential (AP) by 3.1 ± 1.0 mV (control: n = 14, exercise: n = 18, p = 0.005) and increased the AP amplitude by 6.7 ± 3.0 mV (p = 0.031) and firing frequency by more than 22% especially within a range of current stimulation stronger than 70 pA. A 3-week treadmill exercise was sufficient to hyperpolarize PIC onset by 2.6 ± 1.3 mV (control: −53.4 ± 4.7 mV, n = 28; exercise: −56.0 ± 4.7 mV, n = 25, p = 0.050) and increase the PIC amplitude by 28% (control: 193.6 ± 81.8 pA; exercise: 248.5 ± 105.4 pA, p = 0.038). Furthermore, exercise hyperpolarized Na-PIC onset by 3.8 ± 1.8 mV (control: n = 8, exercise: n = 9, p = 0.049) and increased the Ca-PIC amplitude by 23% (p = 0.013). The exercise-induced enhancement of the PIC amplitude was mainly mediated by Ca-PIC and hyperpolarization of PIC onset by Na-PIC. Moreover, exercise facilitated dendritic plasticity, which was shown as the increased number of branch points by 1.5 ± 0.5 (p = 0.009) and dendritic branches by 2.1 ± 0.6 (n = 20, p = 0.001) and length by 732.0 ± 100.1 μm (p < 0.001) especially within the range of 50–200 μm from the soma. Functional analysis suggested that treadmill exercise enhanced Na-PIC for facilitation of spike initiation and Ca-PIC for regulation of repetitive firing. We concluded that PICs broadly existed in DRN 5-HT neurons and could influence serotonergic neurotransmission in juvenile mice and that 3-week treadmill exercise induced synaptic adaptations, enhanced PICs, and thus upregulated the excitability of the 5-HT neurons.
Collapse
Affiliation(s)
- Renkai Ge
- Shanghai Key Laboratory of Multidimensional Information Processing, School of Communication and Electronic Engineering, East China Normal University, Shanghai, China.,School of Physical Education and Health Care, East China Jiaotong University, Nanchang, China
| | - Yue Dai
- Shanghai Key Laboratory of Multidimensional Information Processing, School of Communication and Electronic Engineering, East China Normal University, Shanghai, China.,Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, School of Physical Education and Health Care, East China Normal University, Shanghai, China
| |
Collapse
|
44
|
Daut RA, Ravenel JR, Watkins LR, Maier SF, Fonken LK. The behavioral and neurochemical effects of an inescapable stressor are time of day dependent. Stress 2020; 23:405-416. [PMID: 31868091 PMCID: PMC7335331 DOI: 10.1080/10253890.2019.1707180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Circadian rhythms are ∼24 h fluctuations in physiology and behavior that are synchronized with the light-dark cycle. The circadian system ensures homeostatic balance by regulating multiple systems that respond to environmental stimuli including stress systems. In rats, acute exposure to a series of uncontrollable tailshocks (inescapable stress, IS) produces an anxiety and depression-like phenotype. Anxiety- and fear-related behavioral changes produced by IS are driven by sensitization of serotonergic (5-hydroxytryptamine, 5-HT) neurons in the dorsal raphe nucleus (DRN). Because the circadian and serotonergic systems are closely linked, here we tested whether the DRN-dependent behavioral and neurochemical effects of IS are time of day dependent. Exposure to IS during the light (inactive) phase elicited the expected changes in mood related behaviors. In contrast, rats that underwent IS during the dark (active) phase were buffered against stress-induced changes in juvenile social exploration and shock-elicited freezing, both DRN-dependent outcomes. Interestingly, behavioral anhedonia, which is not a DRN-dependent behavior, was comparably reduced by stress at both times of day. Neurochemical changes complimented the behavioral results: IS-induced activation of DRN 5-HT neurons was greater during the light phase compared to the dark phase. Additionally, 5-HT1AR and 5-HTT, two genes that regulate 5-HT activity were up-regulated during the middle of the light cycle. These data suggest that DRN-dependent behavioral outcomes of IS are time of day dependent and may be mediated by circadian gating of the DRN response to stress.Lay summaryHere we show that the time of day at which a stressor occurs impacts the behavioral and neurochemical outcomes of the stressor. In particular, animals appear more vulnerable to a stressor that occurs during their rest phase. This work may have important implications for shift-workers and other populations that are more likely to encounter stressors during their rest phase.
Collapse
Affiliation(s)
- Rachel A. Daut
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - J. Russell Ravenel
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Linda R. Watkins
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Steven F. Maier
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Laura K. Fonken
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX 78712, USA
- Corresponding Author: Laura K. Fonken, Division of Pharmacology and Toxicology, University of Texas, Austin, TX 78712; phone: (512) 232-8331;
| |
Collapse
|
45
|
Babicola L, Pietrosanto M, Ielpo D, D'Addario SL, Cabib S, Ventura R, Ferlazzo F, Helmer-Citterich M, Andolina D, Lo Iacono L. RISC RNA sequencing in the Dorsal Raphè reveals microRNAs regulatory activities associated with behavioral and functional adaptations to chronic stress. Brain Res 2020; 1736:146763. [DOI: 10.1016/j.brainres.2020.146763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/03/2020] [Accepted: 03/08/2020] [Indexed: 01/06/2023]
|
46
|
Matthiesen M, Mendes LD, Spiacci A, Fortaleza EA, Corrêa FM, Zangrossi H. Serotonin 2C receptors in the basolateral amygdala mediate the anxiogenic effect caused by serotonergic activation of the dorsal raphe dorsomedial subnucleus. J Psychopharmacol 2020; 34:391-399. [PMID: 31637976 DOI: 10.1177/0269881119882797] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Stimulation of serotonergic neurons within the dorsal raphe dorsomedial subnucleus facilitates inhibitory avoidance acquisition in the elevated T-maze. It has been hypothesized that such anxiogenic effect is due to serotonin release in the basolateral nucleus of the amygdala, where facilitation of serotonin 2C receptor-mediated neurotransmission increases anxiety. Besides the dorsal raphe dorsomedial subnucleus, the dorsal raphe caudal subnucleus is recruited by anxiogenic stimulus/situations. However, the behavioral consequences of pharmacological manipulation of this subnucleus are still unknown. AIMS Investigate whether blockade of serotonin 2C receptors in the basolateral nucleus of the amygdala counteracts the anxiogenic effect caused by the stimulation of dorsal raphe dorsomedial subnucleus serotonergic neurons. Evaluate the effects caused by the excitatory amino acid kainic acid or serotonin 1A receptor-modulating drugs in the dorsal raphe caudal subnucleus. METHODS Male Wistar rats were tested in the elevated T-maze and light-dark transition tests after intra-basolateral nucleus of the amygdala injection of the serotonin 2C receptor antagonist SB-242084 (6-chloro-2,3-dihydro-5-methyl-N-[6-[(2-methyl-3-pyridinyl)oxy]-3-pyridinyl]-1H-indole-1-carboxyamide dihydrochloride) followed by intra-dorsal raphe dorsomedial subnucleus administration of the serotonin 1A receptor antagonist WAY-100635 (N-[2-[4-2-methoxyphenyl)-1-piperazinyl]ethyl]-N-2-pyridinil-cyclohexanecarboxamide maleate). In the dorsal raphe caudal subnucleus, animals were injected with kainic acid, WAY-100635 or the serotonin 1A receptor agonist 8-OH-DPAT ((±)-8-hydroxy-2-(di-n-propylamino) tetralin hydrobromide) and tested in the elevated T-maze. RESULTS SB-242084 in the basolateral nucleus of the amygdala blocked the anxiogenic effect caused by the injection of WAY-100635 in the dorsal raphe dorsomedial subnucleus. Kainic acid in the dorsal raphe caudal subnucleus increased anxiety, but also impaired escape expression in the elevated T-maze. Neither WAY-100635 nor 8-OH-DPAT in the dorsal raphe caudal subnucleus affected rat's behavior in the elevated T-maze. CONCLUSION Serotonin 2C receptors in the basolateral nucleus of the amygdala mediate the anxiogenic effect caused by the stimulation of serotonergic neurons in the dorsal raphe dorsomedial subnucleus. The dorsal raphe caudal subnucleus regulates anxiety- and panic-like behaviors, presumably by a serotonin 1A receptor-independent mechanism.
Collapse
Affiliation(s)
- Melina Matthiesen
- Department of Pharmacology, University of São Paulo, São Paulo, Brazil
| | - Leonardo D Mendes
- Department of Pharmacology, University of São Paulo, São Paulo, Brazil
| | - Ailton Spiacci
- Department of Pharmacology, University of São Paulo, São Paulo, Brazil
| | | | | | - Hélio Zangrossi
- Department of Pharmacology, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
47
|
Prakash N, Stark CJ, Keisler MN, Luo L, Der-Avakian A, Dulcis D. Serotonergic Plasticity in the Dorsal Raphe Nucleus Characterizes Susceptibility and Resilience to Anhedonia. J Neurosci 2020; 40:569-584. [PMID: 31792153 PMCID: PMC6961996 DOI: 10.1523/jneurosci.1802-19.2019] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/04/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023] Open
Abstract
Chronic stress induces anhedonia in susceptible but not resilient individuals, a phenomenon observed in humans as well as animal models, but the molecular mechanisms underlying susceptibility and resilience are not well understood. We hypothesized that the serotonergic system, which is implicated in stress, reward, and antidepressant therapy, may play a role. We found that plasticity of the serotonergic system contributes to the differential vulnerability to stress displayed by susceptible and resilient animals. Stress-induced anhedonia was assessed in adult male rats using social defeat and intracranial self-stimulation, while changes in serotonergic phenotype were investigated using immunohistochemistry and in situ hybridization. Susceptible, but not resilient, rats displayed an increased number of neurons expressing the biosynthetic enzyme for serotonin, tryptophan-hydroxylase-2 (TPH2), in the ventral subnucleus of the dorsal raphe nucleus (DRv). Further, a decrease in the number of DRv glutamatergic (VGLUT3+) neurons was observed in all stressed rats. This neurotransmitter plasticity is activity-dependent, as was revealed by chemogenetic manipulation of the central amygdala, a stress-sensitive nucleus that forms a major input to the DR. Activation of amygdalar corticotropin-releasing hormone (CRH)+ neurons abolished the increase in DRv TPH2+ neurons and ameliorated stress-induced anhedonia in susceptible rats. These findings show that activation of amygdalar CRH+ neurons induces resilience, and suppresses the gain of serotonergic phenotype in the DRv that is characteristic of susceptible rats. This molecular signature of vulnerability to stress-induced anhedonia and the active nature of resilience could be targeted to develop new treatments for stress-related disorders like depression.SIGNIFICANCE STATEMENT Depression and other mental disorders can be induced by chronic or traumatic stressors. However, some individuals are resilient and do not develop depression in response to chronic stress. A complete picture of the molecular differences between susceptible and resilient individuals is necessary to understand how plasticity of limbic circuits is associated with the pathophysiology of stress-related disorders. Using a rodent model, our study identifies a novel molecular marker of susceptibility to stress-induced anhedonia, a core symptom of depression, and a means to modulate it. These findings will guide further investigation into cellular and circuit mechanisms of resilience, and the development of new treatments for depression.
Collapse
Affiliation(s)
- Nandkishore Prakash
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093
| | - Christiana J Stark
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093
| | - Maria N Keisler
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093
| | - Lily Luo
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093
| | - Andre Der-Avakian
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093
| | - Davide Dulcis
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
48
|
Donner NC, Mani S, Fitz SD, Kienzle DM, Shekhar A, Lowry CA. Crh receptor priming in the bed nucleus of the stria terminalis (BNST) induces tph2 gene expression in the dorsomedial dorsal raphe nucleus and chronic anxiety. Prog Neuropsychopharmacol Biol Psychiatry 2020; 96:109730. [PMID: 31415826 PMCID: PMC6815726 DOI: 10.1016/j.pnpbp.2019.109730] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 07/31/2019] [Accepted: 08/03/2019] [Indexed: 11/28/2022]
Abstract
The bed nucleus of the stria terminalis (BNST) is a nodal structure in neural circuits controlling anxiety-related defensive behavioral responses. It contains neurons expressing the stress- and anxiety-related neuropeptide corticotropin-releasing hormone (Crh) as well as Crh receptors. Repeated daily subthreshold activation of Crh receptors in the BNST is known to induce a chronic anxiety-like state, but how this affects neurotransmitter-relevant gene expression in target regions of the BNST is still unclear. Since the BNST projects heavily to the dorsal raphe nucleus (DR), the main source of brain serotonin, we here tested the hypothesis that such repeated, anxiety-inducing activation of Crh receptors in the BNST alters the expression of serotonergic genes in the DR, including tph2, the gene encoding the rate-limiting enzyme for brain serotonin synthesis, and slc6a4, the gene encoding the serotonin transporter (SERT). For 5 days, adult male Wistar rats received daily, bilateral, intra-BNST microinjections of vehicle (1% bovine serum albumin in 0.9% saline, n = 11) or behaviorally subthreshold doses of urocortin 1 (Ucn1, n = 11), a potent Crh receptor agonist. Priming with Ucn1 increased tph2 mRNA expression selectively within the anxiety-related dorsal part of the DR (DRD) and decreased social interaction (SI) time, a measure of anxiety-related defensive behavioral responses in rodents. Decreased social interaction was strongly correlated with increased tph2 mRNA expression in the DRD. Together with previous studies, our data are consistent with the hypothesis that Crh-mediated control of the BNST/DRD-serotonergic system plays a key role in the development of chronic anxiety states, possibly also contributing to stress-induced relapses in drug abuse and addiction behavior.
Collapse
Affiliation(s)
- Nina C. Donner
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Sofia Mani
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Stephanie D. Fitz
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Drake M. Kienzle
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Anantha Shekhar
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Christopher A. Lowry
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA,Department of Physical Medicine & Rehabilitation and 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, Aurora, CO 80045, USA
| |
Collapse
|
49
|
Rantala MJ, Luoto S, Krama T, Krams I. Eating Disorders: An Evolutionary Psychoneuroimmunological Approach. Front Psychol 2019; 10:2200. [PMID: 31749720 PMCID: PMC6842941 DOI: 10.3389/fpsyg.2019.02200] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 09/12/2019] [Indexed: 12/18/2022] Open
Abstract
Eating disorders are evolutionarily novel conditions. They lead to some of the highest mortality rates of all psychiatric disorders. Several evolutionary hypotheses have been proposed for eating disorders, but only the intrasexual competition hypothesis is extensively supported by evidence. We present the mismatch hypothesis as a necessary extension to the current theoretical framework of eating disorders. This hypothesis explains the evolutionarily novel adaptive metaproblem that has arisen when mating motives conflict with the large-scale and easy availability of hyper-rewarding but obesogenic foods. This situation is exacerbated particularly in those contemporary environments that are characterized by sedentary lifestyles, ever-present junk foods, caloric surplus and the ubiquity of social comparisons that take place via social media. Our psychoneuroimmunological model connects ultimate-level causation with proximate mechanisms by showing how the adaptive metaproblem between mating motives and food rewards leads to chronic stress and, further, to disordered eating. Chronic stress causes neuroinflammation, which increases susceptibility to OCD-like behaviors that typically co-occur with eating disorders. Chronic stress upregulates the serotonergic system and causes dysphoric mood in anorexia nervosa patients. Dieting, however, reduces serotonin levels and dysphoric mood, leading to a vicious serotonergic-homeostatic stress/starvation cycle whereby cortisol and neuroinflammation increase through stringent dieting. Our psychoneuroimmunological model indicates that between-individual and within-individual variation in eating disorders partially arises from (co)variation in gut microbiota and stress responsivity, which influence neuroinflammation and the serotonergic system. We review the advances that have been made in recent years in understanding how to best treat eating disorders, outlining directions for future clinical research. Current evidence indicates that eating disorder treatments should aim to reduce the chronic stress, neuroinflammation, stress responsivity and gut dysbiosis that fuel the disorders. Connecting ultimate causes with proximate mechanisms and treating biopsychosocial causes rather than manifest symptoms is expected to bring more effective and sophisticated long-term interventions for the millions of people who suffer from eating disorders.
Collapse
Affiliation(s)
| | - Severi Luoto
- English, Drama and Writing Studies, University of Auckland, Auckland, New Zealand
- School of Psychology, University of Auckland, Auckland, New Zealand
| | - Tatjana Krama
- Department of Biotechnology, Daugavpils University, Daugavpils, Latvia
| | - Indrikis Krams
- Department of Biotechnology, Daugavpils University, Daugavpils, Latvia
- Institute of Ecology and Earth Sciences, University of Tartu, Tartu, Estonia
| |
Collapse
|
50
|
Neurobehavioral effects of acute and chronic lead exposure in a desert rodent Meriones shawi: Involvement of serotonin and dopamine. J Chem Neuroanat 2019; 102:101689. [PMID: 31580902 DOI: 10.1016/j.jchemneu.2019.101689] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 11/22/2022]
Abstract
Lead (Pb) is a non physiological metal that has been implicated in toxic processes affecting several organs and biological systems, including the central nervous system. Several studies have focused on changes in lead-associated neurobehavioral and neurochemical alterations that occur due to Pb exposure. The present study evaluates the effects of acute and chronic Pb acetate exposure on serotoninergic and dopaminergic systems within the dorsal raphe nucleus, regarding motor activity and anxiety behaviours. Experiments were carried out on adult male Meriones shawi exposed to acute lead acetate intoxication (25 mg/kg b.w., 3 i.p. injections) or to a chronic lead exposure (0,5%) in drinking water from intrauterine age to adult age. Immunohistochemical staining demonstrated that both acute and chronic lead exposure increased anti-serotonin (anti-5HT) and tyrosine hydroxylase (anti-TH) immuno-reactivities in the dorsal raphe nucleus. In parallel, our results demonstrated that a long term Pb-exposure, but not an acute lead intoxication, induced behavioural alterations including, hyperactivity (open field test), and anxiogenic like-effects. Such neurobehavioral impairments induced by Pb-exposure in Meriones shawi may be related to dopaminergic and serotoninergic injuries identified in the dorsal raphe nucleus.
Collapse
|