1
|
Hung LY, Alves ND, Del Colle A, Talati A, Najjar SA, Bouchard V, Gillet V, Tong Y, Huang Z, Browning KN, Hua J, Liu Y, Woodruff JO, Juarez D, Medina M, Posner J, Tonello R, Yalcinkaya N, Israelyan N, Ringel R, Yang L, Leong KW, Yang M, Sze JY, Savidge T, Gingrich J, Shulman RJ, Gershon MD, Ouellet A, Takser L, Ansorge MS, Margolis KG. Intestinal Epithelial Serotonin as a Novel Target for Treating Disorders of Gut-Brain Interaction and Mood. Gastroenterology 2025; 168:754-768. [PMID: 39672518 DOI: 10.1053/j.gastro.2024.11.012] [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: 08/07/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND & AIMS Mood disorders and disorders of gut-brain interaction (DGBI) are highly prevalent, commonly comorbid, and lack fully effective therapies. Although selective serotonin reuptake inhibitors (SSRIs) are first-line pharmacological treatments for these disorders, they may impart adverse effects, including anxiety, anhedonia, dysmotility, and, in children exposed in utero, an increased risk of cognitive, mood, and gastrointestinal disorders. SSRIs act systemically to block the serotonin reuptake transporter and enhance serotonergic signaling in the brain, intestinal epithelium, and enteric neurons. Yet, the compartments that mediate the therapeutic and adverse effects of SSRIs are unknown, as is whether gestational SSRI exposure directly contributes to human DGBI development. METHODS We used transgenic, surgical, and pharmacological approaches to study the effects of intestinal epithelial serotonin reuptake transporter or serotonin on mood and gastrointestinal function, as well as relevant communication pathways. We also conducted a prospective birth cohort study to assess effects of gestational SSRI exposure on DGBI development. RESULTS Serotonin reuptake transporter ablation targeted to the intestinal epithelium promoted anxiolytic and antidepressive-like effects without causing adverse effects on the gastrointestinal tract or brain; conversely, epithelial serotonin synthesis inhibition increased anxiety and depression-like behaviors. Afferent vagal pathways were found to be conduits by which intestinal epithelial serotonin affects behavior. In utero SSRI exposure is a significant and specific risk factor for development of the DGBI, functional constipation, in the first year of life, irrespective of maternal depressive symptoms. CONCLUSION These findings provide fundamental insights into how the gastrointestinal tract modulates emotional behaviors, reveal a novel gut-targeted therapeutic approach for mood modulation, and suggest a new link in humans between in utero SSRI exposure and DGBI development.
Collapse
Affiliation(s)
- Lin Y Hung
- NYU Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
| | - Nuno D Alves
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; New York State Psychiatric Institute, New York, New York
| | - Andrew Del Colle
- NYU Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
| | - Ardesheer Talati
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; New York State Psychiatric Institute, New York, New York
| | - Sarah A Najjar
- NYU Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
| | - Virginie Bouchard
- Department of Pediatrics, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Virginie Gillet
- Department of Pediatrics, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Yan Tong
- NYU Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
| | - Zixing Huang
- NYU Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
| | - Kirsteen N Browning
- Department of Neural and Behavioral Sciences, Pennsylvania State College of Medicine, Hershey, Pennsylvania
| | - Jialiang Hua
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; New York State Psychiatric Institute, New York, New York
| | - Ying Liu
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; New York State Psychiatric Institute, New York, New York
| | - James O Woodruff
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; New York State Psychiatric Institute, New York, New York
| | - Daniel Juarez
- NYU Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
| | - Melissa Medina
- NYU Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
| | - Jonathan Posner
- Department of Psychiatry, Duke University School of Medicine; Durham, North Carolina
| | - Raquel Tonello
- NYU Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
| | - Nazli Yalcinkaya
- Department of Pathology and Department of Immunology, Baylor College of Medicine and Texas Children's Microbiome Center, Texas Children's Hospital, Houston, Texas
| | - Narek Israelyan
- NYU Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
| | - Roey Ringel
- NYU Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
| | - Letao Yang
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, New York; Department of Systems Biology, Columbia University Medical Center, New York, New York
| | - Mu Yang
- Neurobehavior Core, Institute of Genomic Medicine, Columbia University Irving Medical Center, New York, New York
| | - Ji Ying Sze
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York
| | - Tor Savidge
- Department of Pathology and Department of Immunology, Baylor College of Medicine and Texas Children's Microbiome Center, Texas Children's Hospital, Houston, Texas
| | - Jay Gingrich
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; New York State Psychiatric Institute, New York, New York
| | - Robert J Shulman
- Department of Pediatrics and USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas
| | - Michael D Gershon
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Annie Ouellet
- Department of Obstetrics, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Larissa Takser
- Department of Pediatrics, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Mark S Ansorge
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; New York State Psychiatric Institute, New York, New York.
| | - Kara Gross Margolis
- NYU Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York; Department of Cell Biology, NYU Grossman School of Medicine, New York, New York; Department of Pediatrics, NYU Grossman School of Medicine, New York, New York.
| |
Collapse
|
2
|
Cîmpeanu RC, Caragea EM, Mustață LM, Forțofoiu D, Dragne IG, Alexa RE, Balta A, Ceasovschih A, Șorodoc L, Săndulescu LD. The Involvement of Serotonin in the Obesity Pathway-A Last Decade Systematic Review of the Literature. Int J Mol Sci 2025; 26:3081. [PMID: 40243845 PMCID: PMC11988484 DOI: 10.3390/ijms26073081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/19/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
Obesity represents a complex, multifactorial syndrome that represents a high burden for public health systems worldwide. Serotonin is an important factor in feeding behavior and weight regulation and their interplay implies multiple mechanisms that could explain the correlation with obesity, so understanding these interconnections is essential for developing targeted therapeutic strategies. A systematic review of the literature was conducted using PubMed and Scopus databases, using articles published between 1 January 2015 and 1 December 2024, based on predefined inclusion and exclusion criteria. After the selection process, 22 studies were selected for detailed analysis, focusing on the role of serotonin in obesity. Serotonin significantly influences appetite control and energy homeostasis through multiples pathways, including insulin resistance, high-fat diets, gut microbiota, low-grade inflammation, interferences with tryptophan metabolism, psychiatric modifications, genetic alterations of serotonin receptors, serotonin implications in eating behavior, and neurohormonal regulation of appetite. This review highlights the multidimensional characteristics of the serotonin-obesity association, along with its significance in metabolic and psychiatric pathologies. In order to develop more efficient methods for managing obesity, future studies should concentrate on serotonergic regulation and complex management strategies involving the neurohormonal axis.
Collapse
Affiliation(s)
- Radu-Cristian Cîmpeanu
- Doctoral School of University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Emilia-Mariana Caragea
- Doctoral School of University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Lorena-Maria Mustață
- Doctoral School of University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Dragoș Forțofoiu
- Doctoral School of University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ioana-Gabriela Dragne
- Doctoral School of University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Raluca-Elena Alexa
- Doctoral School of “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Anastasia Balta
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Alexandr Ceasovschih
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Laurențiu Șorodoc
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | | |
Collapse
|
3
|
Muñoz-Castañeda R, Palaniswamy R, Palmer J, Drewes R, Elowsky C, Hirokawa KE, Cain N, Venkataraju KU, Dong HW, Harris JA, Wu Z, Osten P. A Comprehensive Atlas of Cell Type Density Patterns and Their Role in Brain Organization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.02.615922. [PMID: 40166303 PMCID: PMC11956909 DOI: 10.1101/2024.10.02.615922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Cell-type composition across brain regions is a critical structural factor shaping both local and long-range brain circuits. Here, we employed single-cell resolution imaging of the mouse brain, combined with computational analyses, to map the distribution of 30 cell classes and types defined by gene marker expression in Cre recombinase-based genetic mouse models. This approach generated a comprehensive atlas of cell type-specific densities across the male and female brain, revealing (1) surprisingly broad sex differences in cells tagged by developmental cell-type markers, (2) shared cell type composition signatures among functionally related brain structures, and (3) close associations not only between specific cell types but also discrete cell type densities and anatomical regions and subregions. In summary, despite the relatively broad cell type classification enabled by the Cre mouse models, our findings highlight intricate relationships between brain cell type distribution and anatomical organization, associating distinct local cell densities with region-specific brain functions.
Collapse
Affiliation(s)
- Rodrigo Muñoz-Castañeda
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- Appel Alzheimer’s Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | | | - Jason Palmer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Rhonda Drewes
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Corey Elowsky
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | | | | | | | - Hong-Wei Dong
- UCLA Brain Research & Artificial Intelligence Nexus, Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Zhuhao Wu
- Appel Alzheimer’s Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Pavel Osten
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
4
|
Zhang H, Hasegawa Y, Suzuki M, Zhang T, Leitner DR, Jackson RP, Waldor MK. Mouse enteric neurons control intestinal plasmacytoid dendritic cell function via serotonin-HTR7 signaling. Nat Commun 2024; 15:9237. [PMID: 39455564 PMCID: PMC11511829 DOI: 10.1038/s41467-024-53545-2] [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: 03/19/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Serotonergic neurons in the central nervous system control behavior and mood, but knowledge of the roles of serotonergic circuits in the regulation of immune homeostasis is limited. Here, we employ mouse genetics to investigate the functions of enteric serotonergic neurons in the control of immune responses and find that these circuits regulate IgA induction and boost host defense against oral, but not systemic Salmonella Typhimurium infection. Enteric serotonergic neurons promote gut-homing, retention and activation of intestinal plasmacytoid dendritic cells (pDC). Mechanistically, this neuro-immune crosstalk is achieved through a serotonin-5-HT receptor 7 (HTR7) signaling axis that ultimately facilitates the pDC-mediated differentiation of IgA+ B cells from IgD+ precursors in the gut. Single-cell RNA-seq data further reveal novel patterns of bidirectional communication between specific subsets of enteric neurons and lamina propria DC. Our findings thus reveal a close interplay between enteric serotonergic neurons and gut immune homeostasis that enhances mucosal defense.
Collapse
Affiliation(s)
- Hailong Zhang
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Yuko Hasegawa
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Masataka Suzuki
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Ting Zhang
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Deborah R Leitner
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Ruaidhrí P Jackson
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
| | - Matthew K Waldor
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA.
- Howard Hughes Medical Institute, Boston, MA, 02115, USA.
| |
Collapse
|
5
|
Pomrenze MB, Vaillancourt S, Salgado JS, Raymond KB, Llorach P, Touponse GC, Cardozo Pinto DF, Rastegar Z, Casey AB, Eshel N, Malenka RC, Heifets BD. 5-HT 2C receptors in the nucleus accumbens constrain the rewarding effects of MDMA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.20.619256. [PMID: 39484424 PMCID: PMC11527024 DOI: 10.1101/2024.10.20.619256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
MDMA is a promising adjunct to psychotherapy and has well-known abuse liability, although less than other amphetamine analogs. While the reinforcing dopamine (DA)-releasing properties of MDMA are on par with methamphetamine (METH), MDMA is a far more potent serotonin (5-HT) releaser, via the 5-HT transporter (SERT). MDMA-mediated 5-HT release in a major reward center, the nucleus accumbens (NAc), drives prosocial behaviors via 5-HT1BR activation. We hypothesized that this prosocial mechanism contributes to the reduced reinforcing properties of MDMA compared to METH and used a platform of assays to predict the balance of prosocial and abuse-linked effects of (R)-MDMA, a novel entactogen in clinical development. NAc DA release, measured by GRAB-DA photometry in vivo, increased in proportion to MDMA (7.5 and 15 mg/kg, i.p.) and METH (2 mg/kg i.p.)-conditioned place preference (CPP). Using conditional knockouts (cKOs) for DAT and SERT, microdialysis, and photometry, we found that MDMA-released 5-HT limited MDMA-released DA through actions in the NAc, rather than at ventral tegmental area DAergic cell bodies. SERT cKO reduced the MDMA dose required for CPP three-fold. This enhanced MDMA-CPP and increased DA release were replicated by intra-NAc infusion of either a 5-HT reuptake inhibitor (escitalopram) to prevent MDMA interaction with SERT, or a 5-HT2CR antagonist (SB242084), but not by the 5-HT1BR antagonist NAS-181. These data support separate mechanisms for the low abuse potential versus prosocial effect of MDMA. Using this platform of assays, (R)-MDMA is predicted to have prosocial effects and low abuse potential.
Collapse
Affiliation(s)
- Matthew B. Pomrenze
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Sam Vaillancourt
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Juliana S. Salgado
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Kendall B. Raymond
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Pierre Llorach
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Gavin C. Touponse
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Daniel F. Cardozo Pinto
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Zahra Rastegar
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Austen B. Casey
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Neir Eshel
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Robert C. Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Boris D. Heifets
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
6
|
Yuan L, Chu Z, Chen X, He M, Lu Y, Xu X, Shen Z. Structural Neuroimaging and Molecular Signatures of Drug-Naive Depression With Melancholic Features. Depress Anxiety 2024; 2024:9680180. [PMID: 40226700 PMCID: PMC11919201 DOI: 10.1155/2024/9680180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 08/20/2024] [Accepted: 09/14/2024] [Indexed: 04/15/2025] Open
Abstract
Objectives: Melancholic depression (MD) is a common subtype of major depressive disorder (MDD). It is difficult to treat because its neurobiological basis is poorly understood. Therefore, to investigate whether MD patients have any structural changes in gray matter (GM) and the molecular foundation of these changes, we combined voxel-based morphometry (VBM) analysis with neurotransmitter system-derived mapping from public data. Methods: 137 drug-naive MDD patients and 75 healthy controls (HCs) were recruited for structural magnetic resonance imaging. The imaging results were analyzed using VBM analysis. MDD patients were then divided into MD and nonmelancholic depression (NMD) subgroups according to their scores on the Montgomery-Asberg Depression Rating Scale (MADRS) and the Hamilton Depression Rating Scale. Next, we analyzed the spatial correlation between the changes in the gray matter volume (GMV) maps and the neurotransmitter receptor/transporter protein density maps provided by the JuSpace toolbox. Results: Compared to HCs, patients with MD had significant GMV reduction in the bilateral hippocampus, bilateral thalamus, right amygdala, and right posterior cingulate cortex (PCC)/precuneus. Compared to patients with NMD, MD patients had significant GMV reduction in the bilateral PCC/precuneus and lateral occipital cortex. Moreover, compared to HCs, changes in GMV introduced by MD were spatially associated with the serotonin transporter, cannabinoid receptor, and μ-opioid receptor. Compared to NMD patients, changes in GMV introduced by MD were spatially associated with the vesicular acetylcholine transporter. Conclusion: The present study discovered abnormal GMV alterations in patients with subtypes of depression. We also found a series of neurotransmitter receptors that may be associated with the alterations. The findings of the current study may provide a more comprehensive understanding of the molecular mechanisms underlying the structural abnormalities in subtypes of depression and potentially offer new insights into developing new therapeutic strategies.
Collapse
Affiliation(s)
- Lijin Yuan
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Zhaosong Chu
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Xianyu Chen
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Mengxin He
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Yi Lu
- Department of Medical Imaging, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Xiufeng Xu
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
- Yunnan Clinical Research Center for Mental Disorders, Kunming 650032, China
| | - Zonglin Shen
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
- Yunnan Clinical Research Center for Mental Disorders, Kunming 650032, China
| |
Collapse
|
7
|
Homberg JR, Brivio P, Greven CU, Calabrese F. Individuals being high in their sensitivity to the environment: Are sensitive period changes in play? Neurosci Biobehav Rev 2024; 159:105605. [PMID: 38417743 DOI: 10.1016/j.neubiorev.2024.105605] [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: 10/11/2023] [Revised: 02/13/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024]
Abstract
All individuals on planet earth are sensitive to the environment, but some more than others. These individual differences in sensitivity to environments are seen across many animal species including humans, and can influence personalities as well as vulnerability and resilience to mental disorders. Yet, little is known about the underlying brain mechanisms. Key genes that contribute to individual differences in environmental sensitivity are the serotonin transporter, dopamine D4 receptor and brain-derived neurotrophic factor genes. By synthesizing neurodevelopmental findings of these genetic factors, and discussing them through the lens of mechanisms related to sensitive periods, which are phases of heightened neuronal plasticity during which a certain network is being finetuned by experiences, we propose that these genetic factors delay but extend postnatal sensitive periods. This may explain why sensitive individuals show behavioral features that are characteristic of a young brain state at the level of sensory information processing, such as reduced filtering or blockade of irrelevant information, resulting in a sensory processing system that 'keeps all options open'.
Collapse
Affiliation(s)
- Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Paola Brivio
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Corina U Greven
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Karakter Child and Adolescent Psychiatry University Center, Nijmegen, the Netherlands; King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry Center, London, United Kingdom
| | - Francesca Calabrese
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
8
|
Sinclair-Wilson A, Lawrence A, Ferezou I, Cartonnet H, Mailhes C, Garel S, Lokmane L. Plasticity of thalamocortical axons is regulated by serotonin levels modulated by preterm birth. Proc Natl Acad Sci U S A 2023; 120:e2301644120. [PMID: 37549297 PMCID: PMC10438379 DOI: 10.1073/pnas.2301644120] [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: 01/31/2023] [Accepted: 07/09/2023] [Indexed: 08/09/2023] Open
Abstract
Sensory inputs are conveyed to distinct primary areas of the neocortex through specific thalamocortical axons (TCA). While TCA have the ability to reorient postnatally to rescue embryonic mistargeting and target proper modality-specific areas, how this remarkable adaptive process is regulated remains largely unknown. Here, using a mutant mouse model with a shifted TCA trajectory during embryogenesis, we demonstrated that TCA rewiring occurs during a short postnatal time window, preceded by a prenatal apoptosis of thalamic neurons-two processes that together lead to the formation of properly innervated albeit reduced primary sensory areas. We furthermore showed that preterm birth, through serotonin modulation, impairs early postnatal TCA plasticity, as well as the subsequent delineation of cortical area boundary. Our study defines a birth and serotonin-sensitive period that enables concerted adaptations of TCA to primary cortical areas with major implications for our understanding of brain wiring in physiological and preterm conditions.
Collapse
Affiliation(s)
- Alexander Sinclair-Wilson
- Team Brain Development and Plasticity, Institut de Biologie de l’ENS, École Normale Supérieure, CNRS, INSERM, PSL Research University, 75005Paris, France
| | - Akindé Lawrence
- Team Brain Development and Plasticity, Institut de Biologie de l’ENS, École Normale Supérieure, CNRS, INSERM, PSL Research University, 75005Paris, France
| | - Isabelle Ferezou
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400Saclay, France
| | - Hugues Cartonnet
- Team Brain Development and Plasticity, Institut de Biologie de l’ENS, École Normale Supérieure, CNRS, INSERM, PSL Research University, 75005Paris, France
| | - Caroline Mailhes
- Acute Transgenesis Facility, Institut de Biologie de l’ENS, École Normale Supérieure, CNRS, INSERM, PSL Research University, 75005Paris, France
| | - Sonia Garel
- Team Brain Development and Plasticity, Institut de Biologie de l’ENS, École Normale Supérieure, CNRS, INSERM, PSL Research University, 75005Paris, France
- Collège de France, PSL Research University, 75005Paris, France
| | - Ludmilla Lokmane
- Team Brain Development and Plasticity, Institut de Biologie de l’ENS, École Normale Supérieure, CNRS, INSERM, PSL Research University, 75005Paris, France
| |
Collapse
|
9
|
De Gregorio R, Subah G, Chan JC, Speranza L, Zhang X, Ramakrishnan A, Shen L, Maze I, Stanton PK, Sze JY. Sex-biased effects on hippocampal circuit development by perinatal SERT expression in CA3 pyramidal neurons. Development 2022; 149:dev200549. [PMID: 36178075 PMCID: PMC10655925 DOI: 10.1242/dev.200549] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 09/08/2022] [Indexed: 11/20/2022]
Abstract
Neurodevelopmental disorders ranging from autism to intellectual disability display sex-biased prevalence and phenotypical presentations. Despite increasing knowledge about temporospatial cortical map development and genetic variants linked to neurodevelopmental disorders, when and how sex-biased neural circuit derailment may arise in diseased brain remain unknown. Here, we identify in mice that serotonin uptake transporter (SERT) in non-serotonergic neurons - hippocampal and prefrontal pyramidal neurons - confers sex-biased effects specifically during neural circuit development. A set of gradient-patterned CA3 pyramidal neurons transiently express SERT to clear extracellular serotonin, coinciding with hippocampal synaptic circuit establishment. Ablating pyramidal neuron SERT (SERTPyramidΔ) alters dendritic spine developmental trajectory in the hippocampus, and precipitates sex-biased impairments in long-term activity-dependent hippocampal synaptic plasticity and cognitive behaviors. Transcriptomic analyses identify sex-biased alterations in gene sets associated with autism, dendritic spine structure, synaptic function and male-specific enrichment of dysregulated genes in glial cells in early postnatal SERTPyramidΔ hippocampus. Our data suggest that SERT function in these pyramidal neurons underscores a temporal- and brain region-specific regulation of normal sex-dimorphic circuit development and a source for sex-biased vulnerability to cognitive and behavioral impairments. This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Roberto De Gregorio
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Galadu Subah
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA
| | - Jennifer C. Chan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Luisa Speranza
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Xiaolei Zhang
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Ian Maze
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Patric K. Stanton
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA
| | - Ji Y. Sze
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
10
|
Pourhamzeh M, Moravej FG, Arabi M, Shahriari E, Mehrabi S, Ward R, Ahadi R, Joghataei MT. The Roles of Serotonin in Neuropsychiatric Disorders. Cell Mol Neurobiol 2022; 42:1671-1692. [PMID: 33651238 PMCID: PMC11421740 DOI: 10.1007/s10571-021-01064-9] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/12/2021] [Indexed: 12/22/2022]
Abstract
The serotonergic system extends throughout the central nervous system (CNS) and the gastrointestinal (GI) tract. In the CNS, serotonin (5-HT, 5-hydroxytryptamine) modulates a broad spectrum of functions, including mood, cognition, anxiety, learning, memory, reward processing, and sleep. These processes are mediated through 5-HT binding to 5-HT receptors (5-HTRs), are classified into seven distinct groups. Deficits in the serotonergic system can result in various pathological conditions, particularly depression, schizophrenia, mood disorders, and autism. In this review, we outlined the complexity of serotonergic modulation of physiologic and pathologic processes. Moreover, we provided experimental and clinical evidence of 5-HT's involvement in neuropsychiatric disorders and discussed the molecular mechanisms that underlie these illnesses and contribute to the new therapies.
Collapse
Affiliation(s)
- Mahsa Pourhamzeh
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Ghasemi Moravej
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrnoosh Arabi
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Radiology and Medical Physics, Faculty of Paramedicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Elahe Shahriari
- Faculty of Medicine, Department of Physiology, Iran University of Medical Sciences, Tehran, Iran
| | - Soraya Mehrabi
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Faculty of Medicine, Department of Physiology, Iran University of Medical Sciences, Tehran, Iran
| | - Richard Ward
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - Reza Ahadi
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Taghi Joghataei
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Wong FK, Selten M, Rosés-Novella C, Sreenivasan V, Pallas-Bazarra N, Serafeimidou-Pouliou E, Hanusz-Godoy A, Oozeer F, Edwards R, Marín O. Serotonergic regulation of bipolar cell survival in the developing cerebral cortex. Cell Rep 2022; 40:111037. [PMID: 35793629 DOI: 10.1016/j.celrep.2022.111037] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 03/09/2022] [Accepted: 06/11/2022] [Indexed: 11/16/2022] Open
Abstract
One key factor underlying the functional balance of cortical networks is the ratio of excitatory and inhibitory neurons. The mechanisms controlling the ultimate number of interneurons are beginning to be elucidated, but to what extent similar principles govern the survival of the large diversity of cortical inhibitory cells remains to be investigated. Here, we investigate the mechanisms regulating developmental cell death in neurogliaform cells, bipolar cells, and basket cells, the three main populations of interneurons originating from the caudal ganglionic eminence and the preoptic region. We found that all three subclasses of interneurons undergo activity-dependent programmed cell death. However, while neurogliaform cells and basket cells require glutamatergic transmission to survive, the final number of bipolar cells is instead modulated by serotonergic signaling. Together, our results demonstrate that input-specific modulation of neuronal activity controls the survival of cortical interneurons during the critical period of programmed cell death.
Collapse
Affiliation(s)
- Fong Kuan Wong
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Martijn Selten
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Claudia Rosés-Novella
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Varun Sreenivasan
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Noemí Pallas-Bazarra
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Eleni Serafeimidou-Pouliou
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Alicia Hanusz-Godoy
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Fazal Oozeer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Robert Edwards
- Department of Physiology and Department of Neurology, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK.
| |
Collapse
|
12
|
What is the mechanism of loudness hyperacusis in autism? Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2021.110759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
13
|
Kingston R, Amin D, Misra S, Gross JM, Kuwajima T. Serotonin transporter-mediated molecular axis regulates regional retinal ganglion cell vulnerability and axon regeneration after nerve injury. PLoS Genet 2021; 17:e1009885. [PMID: 34735454 PMCID: PMC8594818 DOI: 10.1371/journal.pgen.1009885] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/16/2021] [Accepted: 10/17/2021] [Indexed: 11/19/2022] Open
Abstract
Molecular insights into the selective vulnerability of retinal ganglion cells (RGCs) in optic neuropathies and after ocular trauma can lead to the development of novel therapeutic strategies aimed at preserving RGCs. However, little is known about what molecular contexts determine RGC susceptibility. In this study, we show the molecular mechanisms underlying the regional differential vulnerability of RGCs after optic nerve injury. We identified RGCs in the mouse peripheral ventrotemporal (VT) retina as the earliest population of RGCs susceptible to optic nerve injury. Mechanistically, the serotonin transporter (SERT) is upregulated on VT axons after injury. Utilizing SERT-deficient mice, loss of SERT attenuated VT RGC death and led to robust retinal axon regeneration. Integrin β3, a factor mediating SERT-induced functions in other systems, is also upregulated in RGCs and axons after injury, and loss of integrin β3 led to VT RGC protection and axon regeneration. Finally, RNA sequencing analyses revealed that loss of SERT significantly altered molecular signatures in the VT retina after optic nerve injury, including expression of the transmembrane protein, Gpnmb. GPNMB is rapidly downregulated in wild-type, but not SERT- or integrin β3-deficient VT RGCs after injury, and maintaining expression of GPNMB in RGCs via AAV2 viruses even after injury promoted VT RGC survival and axon regeneration. Taken together, our findings demonstrate that the SERT-integrin β3-GPNMB molecular axis mediates selective RGC vulnerability and axon regeneration after optic nerve injury.
Collapse
Affiliation(s)
- Rody Kingston
- Department of Ophthalmology, The Louis J. Fox Center for Vision Restoration, Pittsburgh, Pennsylvania, United States of America
| | - Dwarkesh Amin
- Department of Ophthalmology, The Louis J. Fox Center for Vision Restoration, Pittsburgh, Pennsylvania, United States of America
| | - Sneha Misra
- Department of Ophthalmology, The Louis J. Fox Center for Vision Restoration, Pittsburgh, Pennsylvania, United States of America
| | - Jeffrey M. Gross
- Department of Ophthalmology, The Louis J. Fox Center for Vision Restoration, Pittsburgh, Pennsylvania, United States of America
- Department of Developmental Biology, The McGowan Institute for Regenerative Medicine, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Takaaki Kuwajima
- Department of Ophthalmology, The Louis J. Fox Center for Vision Restoration, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
14
|
Mao J, Kinkade JA, Bivens NJ, Roberts RM, Rosenfeld CS. Placental Changes in the serotonin transporter (Slc6a4) knockout mouse suggest a role for serotonin in controlling nutrient acquisition. Placenta 2021; 115:158-168. [PMID: 34649169 PMCID: PMC8585720 DOI: 10.1016/j.placenta.2021.09.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/21/2021] [Accepted: 09/28/2021] [Indexed: 01/01/2023]
Abstract
INTRODUCTION The mouse placenta accumulates and possibly produces serotonin (5-hydroxytryptamine; 5-HT) in parietal trophoblast giant cells (pTGC) located at the interface between the placenta and maternal deciduum. However, the roles of 5-HT in placental function are unclear. This lack of information is unfortunate, given that selective serotonin-reuptake inhibitors are commonly used to combat depression in pregnant women. The high affinity 5-HT transporter SLC6A4 (also known as SERT) is the target of such drugs and likely controls much of 5-HT uptake into pTGC and other placental cells. We hypothesized that ablation of the Slc6a4 gene would result in morphological changes correlated with placental gene expression changes, especially for those involved in nutrient acquisition and metabolism, and thereby, provide insights into 5-HT placental function. METHODS Placentas were collected at embryonic age (E) 12.5 from Slc6a4 knockout (KO) and wild-type (WT) conceptuses. Histological analyses, RNAseq, qPCR, and integrative correlation analyses were performed. RESULTS Slc6a4 KO placentas had a considerable increased pTGC to spongiotrophoblast area ratio relative to WT placentas and significantly elevated expression of genes associated with intestinal functions, including nutrient sensing, uptake, and catabolism, and blood clotting. Integrative correlation analyses revealed upregulation of many of these genes was correlated with pTGC layer expansion. One other key gene was dopa decarboxylase (Ddc), which catalyzes conversion of L-5-hydroxytryptophan to 5-HT. DISCUSSION Our studies possibly suggest a new paradigm relating to how 5-HT operates in the placenta, namely as a factor regulating metabolic functions and blood coagulation. We further suggest that pTGC might be functional analogs of enterochromaffin 5-HT-positive cells of the intestinal mucosa, which regulate similar activities within the gut. Further work, including proteomics and metabolomic studies, are needed to buttress our hypothesis.
Collapse
Affiliation(s)
- Jiude Mao
- Christopher S Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA; Biomedical Sciences, University of Missouri, Columbia, MO, 65211, USA.
| | - Jessica A Kinkade
- Christopher S Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA; Biomedical Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Nathan J Bivens
- Genomics Technology Core, University of Missouri, Columbia, MO, 65211, USA
| | - R Michael Roberts
- Christopher S Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA; Animal Sciences, University of Missouri, Columbia, MO, 65211, USA; Biochemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Cheryl S Rosenfeld
- Biomedical Sciences, University of Missouri, Columbia, MO, 65211, USA; MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO, 65211, USA; Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO, 65211, USA; Genetics Area Program, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|
15
|
Pan W, Pan J, Zhao Y, Zhang H, Tang J. Serotonin Transporter Defect Disturbs Structure and Function of the Auditory Cortex in Mice. Front Neurosci 2021; 15:749923. [PMID: 34690685 PMCID: PMC8527018 DOI: 10.3389/fnins.2021.749923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/07/2021] [Indexed: 11/23/2022] Open
Abstract
Serotonin transporter (SERT) modulates the level of 5-HT and significantly affects the activity of serotonergic neurons in the central nervous system. The manipulation of SERT has lasting neurobiological and behavioral consequences, including developmental dysfunction, depression, and anxiety. Auditory disorders have been widely reported as the adverse events of these mental diseases. It is unclear how SERT impacts neuronal connections/interactions and what mechanism(s) may elicit the disruption of normal neural network functions in auditory cortex. In the present study, we report on the neuronal morphology and function of auditory cortex in SERT knockout (KO) mice. We show that the dendritic length of the fourth layer (L-IV) pyramidal neurons and the second-to-third layer (L-II/III) interneurons were reduced in the auditory cortex of the SERT KO mice. The number and density of dendritic spines of these neurons were significantly less than those of wild-type neurons. Also, the frequency-tonotopic organization of primary auditory cortex was disrupted in SERT KO mice. The auditory neurons of SERT KO mice exhibited border frequency tuning with high-intensity thresholds. These findings indicate that SERT plays a key role in development and functional maintenance of auditory cortical neurons. Auditory function should be examined when SERT is selected as a target in the treatment for psychiatric disorders.
Collapse
Affiliation(s)
- Wenlu Pan
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Functional Nucleic Acid Basic and Clinical Research Center, Department of Physiology, School of Basic Medical Sciences, Changsha Medical College, Changsha, China
| | - Jing Pan
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Hearing Research Center, Southern Medical University, Guangzhou, China
| | - Yan Zhao
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hongzheng Zhang
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Hearing Research Center, Southern Medical University, Guangzhou, China
| | - Jie Tang
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Hearing Research Center, Southern Medical University, Guangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, China
| |
Collapse
|
16
|
De Gregorio R, Chen X, Petit EI, Dobrenis K, Sze JY. Disruption of Transient SERT Expression in Thalamic Glutamatergic Neurons Alters Trajectory of Postnatal Interneuron Development in the Mouse Cortex. Cereb Cortex 2021; 30:1623-1636. [PMID: 31504267 DOI: 10.1093/cercor/bhz191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 06/29/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022] Open
Abstract
In mice, terminal differentiation of subpopulations of interneurons occurs in late postnatal stages, paralleling the emergence of the adult cortical architecture. Here, we investigated the effects of altered initial cortical architecture on later interneuron development. We identified that a class of somatostatin (SOM)-expressing GABAergic interneurons undergoes terminal differentiation between 2nd and 3rd postnatal week in the mouse somatosensory barrel cortex and upregulates Reelin expression during neurite outgrowth. Our previous work demonstrated that transient expression (E15-P10) of serotonin uptake transporter (SERT) in thalamocortical projection neurons regulates barrel elaboration during cortical map establishment. We show here that in thalamic neuron SERT knockout mice, these SOM-expressing interneurons develop at the right time, reach correct positions and express correct neurochemical markers, but only 70% of the neurons remain in the adult barrel cortex. Moreover, those neurons that remain display altered dendritic patterning. Our data indicate that a precise architecture at the cortical destination is not essential for specifying late-developing interneuron identities, their cortical deposition, and spatial organization, but dictates their number and dendritic structure ultimately integrated into the cortex. Our study illuminates how disruption of temporal-specific SERT function and related key regulators during cortical map establishment can alter interneuron development trajectory that persists to adult central nervous system.
Collapse
Affiliation(s)
- Roberto De Gregorio
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Xiaoning Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Emilie I Petit
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Kostantin Dobrenis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Ji Ying Sze
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| |
Collapse
|
17
|
Stilley SE, Blakely RD. Rare Opportunities for Insights Into Serotonergic Contributions to Brain and Bowel Disorders: Studies of the SERT Ala56 Mouse. Front Cell Neurosci 2021; 15:677563. [PMID: 34149362 PMCID: PMC8210832 DOI: 10.3389/fncel.2021.677563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Altered structure, expression, and regulation of the presynaptic serotonin (5-HT) transporter (SERT) have been associated with multiple neurobehavioral disorders, including mood disorders, obsessive-compulsive disorder (OCD), and autism spectrum disorder (ASD). Opportunities to investigate mechanistic links supporting these associations were spurred with the identification of multiple, rare human SERT coding variants in a study that established a male-specific linkage of ASD to a linkage marker on chromosome 17 which encompassed the location of the SERT gene (SLC6A4). We have explored the most common of these variants, SERT Ala56, in vitro and in vivo. Results support a tonic elevation of 5-HT transport activity in transfected cells and human lymphoblasts by the variant in vitro that leads to an increased 5-HT clearance rate in vivo when studied in the SERT Ala56 mouse model, along with altered sensitivity to SERT regulatory signaling pathways. Importantly, hyperserotonemia, or an elevated whole blood 5-HT, level, was found in SERT Ala56 mice, reproducing a well-replicated trait observed in a significant fraction of ASD subjects. Additionally, we found multiple biochemical, physiological, and behavioral alterations in the SERT Ala56 mice that can be analogized to those observed in ASD and its medical comorbidities. The similarity of the functional impact of the SERT Ala56 variant to the consequences of p38α MAPK activation, ascribed to the induction of a biased conformation of the transporter toward an outward-facing conformation, has resulted in successful efforts to restore normal behavioral and bowel function via pharmacological and genetic p38α MAPK targeting. Moreover, the ability of the inflammatory cytokine IL-1β to enhance SERT activity via a p38α MAPK-dependent pathway suggests that the SERT Ala56 conformation mimics that of a chronic inflammatory state, supporting findings in ASD of elevated inflammatory cytokine levels. In this report, we review studies of the SERT Ala56 variant, discussing opportunities for continued insight into how chronically altered synaptic 5-HT homeostasis can drive reversible, functional perturbations in 5-HT sensitive pathways in the brain and periphery, and how targeting the SERT regulome, particularly through activating pathways such as those involving IL-1β/p38α MAPK, may be of benefit for neurobehavioral disorders, including ASD.
Collapse
Affiliation(s)
- Samantha E. Stilley
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Randy D. Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
- Brain Institute, Florida Atlantic University, Jupiter, FL, United States
| |
Collapse
|
18
|
Lorsung E, Karthikeyan R, Cao R. Biological Timing and Neurodevelopmental Disorders: A Role for Circadian Dysfunction in Autism Spectrum Disorders. Front Neurosci 2021; 15:642745. [PMID: 33776640 PMCID: PMC7994532 DOI: 10.3389/fnins.2021.642745] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/03/2021] [Indexed: 01/07/2023] Open
Abstract
Autism spectrum disorders (ASDs) are a spectrum of neurodevelopmental disorders characterized by impaired social interaction and communication, as well as stereotyped and repetitive behaviors. ASDs affect nearly 2% of the United States child population and the worldwide prevalence has dramatically increased in recent years. The etiology is not clear but ASD is thought to be caused by a combination of intrinsic and extrinsic factors. Circadian rhythms are the ∼24 h rhythms driven by the endogenous biological clock, and they are found in a variety of physiological processes. Growing evidence from basic and clinical studies suggest that the dysfunction of the circadian timing system may be associated with ASD and its pathogenesis. Here we review the findings that link circadian dysfunctions to ASD in both experimental and clinical studies. We first introduce the organization of the circadian system and ASD. Next, we review physiological indicators of circadian rhythms that are found disrupted in ASD individuals, including sleep-wake cycles, melatonin, cortisol, and serotonin. Finally, we review evidence in epidemiology, human genetics, and biochemistry that indicates underlying associations between circadian regulation and the pathogenesis of ASD. In conclusion, we propose that understanding the functional importance of the circadian clock in normal and aberrant neurodevelopmental processes may provide a novel perspective to tackle ASD, and clinical treatments for ASD individuals should comprise an integrative approach considering the dynamics of daily rhythms in physical, mental, and social processes.
Collapse
Affiliation(s)
- Ethan Lorsung
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, United States
| | - Ramanujam Karthikeyan
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, United States
| | - Ruifeng Cao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, United States
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
19
|
Van der Knaap N, Wiedermann D, Schubert D, Hoehn M, Homberg JR. Perinatal SSRI exposure affects brain functional activity associated with whisker stimulation in adolescent and adult rats. Sci Rep 2021; 11:1680. [PMID: 33462357 PMCID: PMC7814075 DOI: 10.1038/s41598-021-81327-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 01/05/2021] [Indexed: 01/29/2023] Open
Abstract
Selective serotonin reuptake inhibitors (SSRI), such as fluoxetine, are used as first-line antidepressant medication during pregnancy. Since SSRIs cross the placenta the unborn child is exposed to the maternal SSRI medication, resulting in, amongst others, increased risk for autism in offspring. This likely results from developmental changes in brain function. Studies employing rats lacking the serotonin transporter have shown that elevations in serotonin levels particularly affect the development of the whisker related part of the primary somatosensory (barrel) cortex. Therefore, we hypothesized that serotonin level disturbances during development alter brain activity related to whisker stimulation. We treated female dams with fluoxetine or vehicle from gestational day 11 onwards for 21 days. We investigated offspring's brain activity during whisker stimulation using functional magnetic resonance imaging (fMRI) at adolescence and adulthood. Our results indicate that adolescent offspring displayed increased activity in hippocampal subareas and the mammillary body in the thalamus. Adult offspring exhibited increased functional activation of areas associated with (higher) sensory processing and memory such as the hippocampus, perirhinal and entorhinal cortex, retrospinal granular cortex, piriform cortex and secondary visual cortex. Our data imply that perinatal SSRI exposure leads to complex alterations in brain networks involved in sensory perception and processing.
Collapse
Affiliation(s)
- Noortje Van der Knaap
- Donders Institute for Brain, Cognition and Behaviour, Radboud University and Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Dirk Wiedermann
- In-Vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Dirk Schubert
- Donders Institute for Brain, Cognition and Behaviour, Radboud University and Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Mathias Hoehn
- In-Vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Judith R Homberg
- Donders Institute for Brain, Cognition and Behaviour, Radboud University and Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands.
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Kapittelweg 29, 6525 EN, Nijmegen, The Netherlands.
| |
Collapse
|
20
|
Vahid-Ansari F, Albert PR. Rewiring of the Serotonin System in Major Depression. Front Psychiatry 2021; 12:802581. [PMID: 34975594 PMCID: PMC8716791 DOI: 10.3389/fpsyt.2021.802581] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/17/2021] [Indexed: 12/14/2022] Open
Abstract
Serotonin is a key neurotransmitter that is implicated in a wide variety of behavioral and cognitive phenotypes. Originating in the raphe nuclei, 5-HT neurons project widely to innervate many brain regions implicated in the functions. During the development of the brain, as serotonin axons project and innervate brain regions, there is evidence that 5-HT plays key roles in wiring the developing brain, both by modulating 5-HT innervation and by influencing synaptic organization within corticolimbic structures. These actions are mediated by 14 different 5-HT receptors, with region- and cell-specific patterns of expression. More recently, the role of the 5-HT system in synaptic re-organization during adulthood has been suggested. The 5-HT neurons have the unusual capacity to regrow and reinnervate brain regions following insults such as brain injury, chronic stress, or altered development that result in disconnection of the 5-HT system and often cause depression, anxiety, and cognitive impairment. Chronic treatment with antidepressants that amplify 5-HT action, such as selective serotonin reuptake inhibitors (SSRIs), appears to accelerate the rewiring of the 5-HT system by mechanisms that may be critical to the behavioral and cognitive improvements induced in these models. In this review, we survey the possible 5-HT receptor mechanisms that could mediate 5-HT rewiring and assess the evidence that 5-HT-mediated brain rewiring is impacting recovery from mental illness. By amplifying 5-HT-induced rewiring processes using SSRIs and selective 5-HT agonists, more rapid and effective treatments for injury-induced mental illness or cognitive impairment may be achieved.
Collapse
Affiliation(s)
- Faranak Vahid-Ansari
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Paul R Albert
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| |
Collapse
|
21
|
Lipiec MA, Bem J, Koziński K, Chakraborty C, Urban-Ciećko J, Zajkowski T, Dąbrowski M, Szewczyk ŁM, Toval A, Ferran JL, Nagalski A, Wiśniewska MB. TCF7L2 regulates postmitotic differentiation programmes and excitability patterns in the thalamus. Development 2020; 147:dev.190181. [PMID: 32675279 PMCID: PMC7473649 DOI: 10.1242/dev.190181] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022]
Abstract
Neuronal phenotypes are controlled by terminal selector transcription factors in invertebrates, but only a few examples of such regulators have been provided in vertebrates. We hypothesised that TCF7L2 regulates different stages of postmitotic differentiation in the thalamus, and functions as a thalamic terminal selector. To investigate this hypothesis, we used complete and conditional knockouts of Tcf7l2 in mice. The connectivity and clustering of neurons were disrupted in the thalamo-habenular region in Tcf7l2-/- embryos. The expression of subregional thalamic and habenular transcription factors was lost and region-specific cell migration and axon guidance genes were downregulated. In mice with a postnatal Tcf7l2 knockout, the induction of genes that confer thalamic terminal electrophysiological features was impaired. Many of these genes proved to be direct targets of TCF7L2. The role of TCF7L2 in terminal selection was functionally confirmed by impaired firing modes in thalamic neurons in the mutant mice. These data corroborate the existence of master regulators in the vertebrate brain that control stage-specific genetic programmes and regional subroutines, maintain regional transcriptional network during embryonic development, and induce terminal selection postnatally.
Collapse
Affiliation(s)
- Marcin Andrzej Lipiec
- Centre of New Technologies, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland.,Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Joanna Bem
- Centre of New Technologies, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland
| | - Kamil Koziński
- Centre of New Technologies, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland
| | - Chaitali Chakraborty
- Centre of New Technologies, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland
| | | | - Tomasz Zajkowski
- Centre of New Technologies, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland
| | - Michał Dąbrowski
- Nencki Institute of Experimental Biology, Pasteur 3, 02-093 Warsaw, Poland
| | | | - Angel Toval
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia and IMIB-Arrixaca Institute, Campus de la Salud, 30120 El Palmar, Murcia, Spain
| | - José Luis Ferran
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia and IMIB-Arrixaca Institute, Campus de la Salud, 30120 El Palmar, Murcia, Spain
| | - Andrzej Nagalski
- Centre of New Technologies, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland
| | | |
Collapse
|
22
|
Hanswijk SI, Spoelder M, Shan L, Verheij MMM, Muilwijk OG, Li W, Liu C, Kolk SM, Homberg JR. Gestational Factors throughout Fetal Neurodevelopment: The Serotonin Link. Int J Mol Sci 2020; 21:E5850. [PMID: 32824000 PMCID: PMC7461571 DOI: 10.3390/ijms21165850] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/24/2020] [Accepted: 08/11/2020] [Indexed: 12/21/2022] Open
Abstract
Serotonin (5-HT) is a critical player in brain development and neuropsychiatric disorders. Fetal 5-HT levels can be influenced by several gestational factors, such as maternal genotype, diet, stress, medication, and immune activation. In this review, addressing both human and animal studies, we discuss how these gestational factors affect placental and fetal brain 5-HT levels, leading to changes in brain structure and function and behavior. We conclude that gestational factors are able to interact and thereby amplify or counteract each other's impact on the fetal 5-HT-ergic system. We, therefore, argue that beyond the understanding of how single gestational factors affect 5-HT-ergic brain development and behavior in offspring, it is critical to elucidate the consequences of interacting factors. Moreover, we describe how each gestational factor is able to alter the 5-HT-ergic influence on the thalamocortical- and prefrontal-limbic circuitry and the hypothalamo-pituitary-adrenocortical-axis. These alterations have been associated with risks to develop attention deficit hyperactivity disorder, autism spectrum disorders, depression, and/or anxiety. Consequently, the manipulation of gestational factors may be used to combat pregnancy-related risks for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sabrina I. Hanswijk
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Marcia Spoelder
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Ling Shan
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands;
| | - Michel M. M. Verheij
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Otto G. Muilwijk
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Weizhuo Li
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China; (W.L.); (C.L.)
| | - Chunqing Liu
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China; (W.L.); (C.L.)
| | - Sharon M. Kolk
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behavior, Radboud University, 6525 AJ Nijmegen, The Netherlands;
| | - Judith R. Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| |
Collapse
|
23
|
Abstract
Neurons that synthesize and release 5-hydroxytryptamine (5-HT; serotonin) express a core set of genes that establish and maintain this neurotransmitter phenotype and distinguish these neurons from other brain cells. Beyond a shared 5-HTergic phenotype, these neurons display divergent cellular properties in relation to anatomy, morphology, hodology, electrophysiology and gene expression, including differential expression of molecules supporting co-transmission of additional neurotransmitters. This diversity suggests that functionally heterogeneous subtypes of 5-HT neurons exist, but linking subsets of these neurons to particular functions has been technically challenging. We discuss recent data from molecular genetic, genomic and functional methods that, when coupled with classical findings, yield a reframing of the 5-HT neuronal system as a conglomeration of diverse subsystems with potential to inspire novel, more targeted therapies for clinically distinct 5-HT-related disorders.
Collapse
|
24
|
Zindler F, Beedgen F, Brandt D, Steiner M, Stengel D, Baumann L, Braunbeck T. Analysis of tail coiling activity of zebrafish (Danio rerio) embryos allows for the differentiation of neurotoxicants with different modes of action. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 186:109754. [PMID: 31606639 DOI: 10.1016/j.ecoenv.2019.109754] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 06/10/2023]
Abstract
In (eco)toxicology, there is a critical need for efficient methods to evaluate the neurotoxic potential of environmental chemicals. Recent studies proposed analysis of early coiling activity in zebrafish embryos as a powerful tool for the identification of neurotoxic compounds. In order to demonstrate that the analysis of early tail movements of zebrafish embryos allows for the discrimination of neurotoxicants acting via different mechanisms, the present study investigated the effects of four different neurotoxicants on the embryogenesis (fish embryo toxicity test) and early tail coiling movements of zebrafish embryos. Cadmium predominantly increased the frequency of tail coiling at the late pharyngula stage. Dichlorvos delayed embryonic development and caused convulsive tail movements resulting in prolonged duration of tail coils. Embryos exposed to teratogenic concentrations of fluoxetine and citalopram displayed absence of spontaneous tail movements at 24 h post-fertilization. In contrast, a non-teratogenic test concentration of citalopram decreased coiling frequency at multiple time points. Results demonstrated that the analysis of tail coiling movements of zebrafish embryos has the potential to discriminate neurotoxic compounds with different primary modes of action. In addition, chemical-induced effects on coiling activity were shown to potentially overlap with effects on embryogenesis. Further studies are needed to clarify the interplay of unspecific developmental toxicity of neurotoxic chemicals and effects resulting from specific neurotoxic mechanisms.
Collapse
Affiliation(s)
- Florian Zindler
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, Heidelberg, D-69120, Germany.
| | - Franziska Beedgen
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, Heidelberg, D-69120, Germany
| | - Diana Brandt
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, Heidelberg, D-69120, Germany
| | - Madeleine Steiner
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, Heidelberg, D-69120, Germany
| | - Daniel Stengel
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, Heidelberg, D-69120, Germany
| | - Lisa Baumann
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, Heidelberg, D-69120, Germany
| | - Thomas Braunbeck
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, Heidelberg, D-69120, Germany
| |
Collapse
|
25
|
Heifets BD, Salgado JS, Taylor MD, Hoerbelt P, Cardozo Pinto DF, Steinberg EE, Walsh JJ, Sze JY, Malenka RC. Distinct neural mechanisms for the prosocial and rewarding properties of MDMA. Sci Transl Med 2019; 11:eaaw6435. [PMID: 31826983 PMCID: PMC7123941 DOI: 10.1126/scitranslmed.aaw6435] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 08/01/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022]
Abstract
The extensively abused recreational drug (±)3,4-methylenedioxymethamphetamine (MDMA) has shown promise as an adjunct to psychotherapy for treatment-resistant psychiatric disease. It is unknown, however, whether the mechanisms underlying its prosocial therapeutic effects and abuse potential are distinct. We modeled both the prosocial and nonsocial drug reward of MDMA in mice and investigated the mechanism of these processes using brain region-specific pharmacology, transgenic manipulations, electrophysiology, and in vivo calcium imaging. We demonstrate in mice that MDMA acting at the serotonin transporter within the nucleus accumbens is necessary and sufficient for MDMA's prosocial effect. MDMA's acute rewarding properties, in contrast, require dopaminergic signaling. MDMA's prosocial effect requires 5-HT1b receptor activation and is mimicked by d-fenfluramine, a selective serotonin-releasing compound. By dissociating the mechanisms of MDMA's prosocial effects from its addictive properties, we provide evidence for a conserved neuronal pathway, which can be leveraged to develop novel therapeutics with limited abuse liability.
Collapse
Affiliation(s)
- Boris D Heifets
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Juliana S Salgado
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Madison D Taylor
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Paul Hoerbelt
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Daniel F Cardozo Pinto
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Elizabeth E Steinberg
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Jessica J Walsh
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Ji Y Sze
- Department of Molecular Pharmacology and Rose F. Kennedy Intellectual and Developmental Disabilities Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Robert C Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
26
|
Mercurio S, Serra L, Nicolis SK. More than just Stem Cells: Functional Roles of the Transcription Factor Sox2 in Differentiated Glia and Neurons. Int J Mol Sci 2019; 20:E4540. [PMID: 31540269 PMCID: PMC6769708 DOI: 10.3390/ijms20184540] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/02/2019] [Accepted: 09/06/2019] [Indexed: 02/06/2023] Open
Abstract
The Sox2 transcription factor, encoded by a gene conserved in animal evolution, has become widely known because of its functional relevance for stem cells. In the developing nervous system, Sox2 is active in neural stem cells, and important for their self-renewal; differentiation to neurons and glia normally involves Sox2 downregulation. Recent evidence, however, identified specific types of fully differentiated neurons and glia that retain high Sox2 expression, and critically require Sox2 function, as revealed by functional studies in mouse and in other animals. Sox2 was found to control fundamental aspects of the biology of these cells, such as the development of correct neuronal connectivity. Sox2 downstream target genes identified within these cell types provide molecular mechanisms for cell-type-specific Sox2 neuronal and glial functions. SOX2 mutations in humans lead to a spectrum of nervous system defects, involving vision, movement control, and cognition; the identification of neurons and glia requiring Sox2 function, and the investigation of Sox2 roles and molecular targets within them, represents a novel perspective for the understanding of the pathogenesis of these defects.
Collapse
Affiliation(s)
- Sara Mercurio
- Department of Biotechnology and Biosciences, University Milano-Bicocca, 20126 Milano, Italy.
| | - Linda Serra
- Department of Biotechnology and Biosciences, University Milano-Bicocca, 20126 Milano, Italy
- CNRS, Inserm, iBV, Université Côte d'Azur, 06108 Nice, France
| | - Silvia K Nicolis
- Department of Biotechnology and Biosciences, University Milano-Bicocca, 20126 Milano, Italy.
| |
Collapse
|
27
|
Mercurio S, Serra L, Motta A, Gesuita L, Sanchez-Arrones L, Inverardi F, Foglio B, Barone C, Kaimakis P, Martynoga B, Ottolenghi S, Studer M, Guillemot F, Frassoni C, Bovolenta P, Nicolis SK. Sox2 Acts in Thalamic Neurons to Control the Development of Retina-Thalamus-Cortex Connectivity. iScience 2019; 15:257-273. [PMID: 31082736 PMCID: PMC6517317 DOI: 10.1016/j.isci.2019.04.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/05/2019] [Accepted: 04/23/2019] [Indexed: 12/19/2022] Open
Abstract
Visual system development involves the formation of neuronal projections connecting the retina to the thalamic dorso-lateral geniculate nucleus (dLGN) and the thalamus to the visual cerebral cortex. Patients carrying mutations in the SOX2 transcription factor gene present severe visual defects, thought to be linked to SOX2 functions in the retina. We show that Sox2 is strongly expressed in mouse postmitotic thalamic projection neurons. Cre-mediated deletion of Sox2 in these neurons causes reduction of the dLGN, abnormal distribution of retino-thalamic and thalamo-cortical projections, and secondary defects in cortical patterning. Reduced expression, in mutants, of Sox2 target genes encoding ephrin-A5 and the serotonin transport molecules SERT and vMAT2 (important for establishment of thalamic connectivity) likely provides a molecular contribution to these defects. These findings unveil thalamic SOX2 function as a novel regulator of visual system development and a plausible additional cause of brain-linked genetic blindness in humans. Sox2 is expressed in postmitotic neurons of the visual thalamic nucleus (dLGN) Sox2 ablation in the dLGN perturbs retino-thalamic and thalamo-cortical projections The visual cortex is not correctly patterned in Sox2 thalamic mutants Downregulation of EphrinA5 and SERT expression may mediate these defects
Collapse
Affiliation(s)
- Sara Mercurio
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza 2, 20126 Milano, Italy
| | - Linda Serra
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza 2, 20126 Milano, Italy; Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | - Alessia Motta
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza 2, 20126 Milano, Italy
| | - Lorenzo Gesuita
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza 2, 20126 Milano, Italy
| | - Luisa Sanchez-Arrones
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid and CIBER de Enfermedades Raras (CIBERER), ISCIII Madrid, Madrid, Spain
| | - Francesca Inverardi
- Clinical and Experimental Epileptology Unit, Fondazione I.R.C.C.S. Istituto Neurologico "Carlo Besta", c/o AMADEOLAB, via Amadeo 42, 20133 Milano, Italy
| | - Benedetta Foglio
- Clinical and Experimental Epileptology Unit, Fondazione I.R.C.C.S. Istituto Neurologico "Carlo Besta", c/o AMADEOLAB, via Amadeo 42, 20133 Milano, Italy
| | - Cristiana Barone
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza 2, 20126 Milano, Italy
| | - Polynikis Kaimakis
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid and CIBER de Enfermedades Raras (CIBERER), ISCIII Madrid, Madrid, Spain
| | - Ben Martynoga
- The Francis Crick Institute, Midland Road, London NW 1AT, UK
| | - Sergio Ottolenghi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza 2, 20126 Milano, Italy
| | | | | | - Carolina Frassoni
- Clinical and Experimental Epileptology Unit, Fondazione I.R.C.C.S. Istituto Neurologico "Carlo Besta", c/o AMADEOLAB, via Amadeo 42, 20133 Milano, Italy
| | - Paola Bovolenta
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid and CIBER de Enfermedades Raras (CIBERER), ISCIII Madrid, Madrid, Spain
| | - Silvia K Nicolis
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza 2, 20126 Milano, Italy.
| |
Collapse
|
28
|
The development of synaptic transmission is time-locked to early social behaviors in rats. Nat Commun 2019; 10:1195. [PMID: 30867422 PMCID: PMC6416358 DOI: 10.1038/s41467-019-09156-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 02/24/2019] [Indexed: 11/19/2022] Open
Abstract
The development of functional synapses is a sequential process preserved across many brain areas. Here, we show that glutamatergic postsynaptic currents anticipated GABAergic currents in Layer II/III of the rat neocortex, in contrast to the pattern described for other brain areas. The frequencies of both glutamatergic and GABAergic currents increased abruptly at the beginning of the second postnatal week, supported by a serotonin upsurge. Integrative behaviors arose on postnatal day (P)9, while most motor and sensory behaviors, which are fundamental for pup survival, were already in place at approximately P7. A reduction in serotonin reuptake accelerated the development of functional synapses and integrative huddling behavior, while sparing motor and sensory function development. A decrease in synaptic transmission in Layer II/III induced by a chemogenetic approach only inhibited huddling. Thus, precise developmental sequences mediate early, socially directed behaviors for which neurotransmission and its modulation in supragranular cortical layers play key roles. The development of functional synapses is a key milestone in neurodevelopment. Here, the authors show how serotonin signalling coordinates development of glutamatergic and GABAergic currents and triggers the emergence of integrative behavior (huddling) in rat pups.
Collapse
|
29
|
Garbarino VR, Gilman TL, Daws LC, Gould GG. Extreme enhancement or depletion of serotonin transporter function and serotonin availability in autism spectrum disorder. Pharmacol Res 2019; 140:85-99. [PMID: 30009933 PMCID: PMC6345621 DOI: 10.1016/j.phrs.2018.07.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/22/2018] [Accepted: 07/12/2018] [Indexed: 12/15/2022]
Abstract
A variety of human and animal studies support the hypothesis that serotonin (5-hydroxytryptamine or 5-HT) system dysfunction is a contributing factor to the development of autism in some patients. However, many questions remain about how developmental manipulation of various components that influence 5-HT signaling (5-HT synthesis, transport, metabolism) persistently impair social behaviors. This review will summarize key aspects of central 5-HT function important for normal brain development, and review evidence implicating perinatal disruptions in 5-HT signaling in the pathophysiology of autism spectrum disorder. We discuss the importance, and relative dearth, of studies that explore the possible correlation to autism in the interactions between important intrinsic and extrinsic factors that may disrupt 5-HT homeostasis during development. In particular, we focus on exposure to 5-HT transport altering mechanisms such as selective serotonin-reuptake inhibitors or genetic polymorphisms in primary or auxiliary transporters of 5-HT, and how they relate to neurological stores of serotonin and its precursors. A deeper understanding of the many mechanisms by which 5-HT signaling can be disrupted, alone and in concert, may contribute to an improved understanding of the etiologies and heterogeneous nature of this disorder. We postulate that extreme bidirectional perturbations of these factors during development likely compound or synergize to facilitate enduring neurochemical changes resulting in insufficient or excessive 5-HT signaling, that could underlie the persistent behavioral characteristics of autism spectrum disorder.
Collapse
Affiliation(s)
- Valentina R Garbarino
- Department of Cellular and Integrative Physiology, United States; The Sam and Ann Barshop Institute for Longevity and Aging Studies, United States.
| | - T Lee Gilman
- Department of Cellular and Integrative Physiology, United States; Addiction Research, Treatment & Training Center of Excellence, United States.
| | - Lynette C Daws
- Department of Cellular and Integrative Physiology, United States; Addiction Research, Treatment & Training Center of Excellence, United States; Department of Pharmacology, United States.
| | - Georgianna G Gould
- Department of Cellular and Integrative Physiology, United States; Center for Biomedical Neuroscience, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
30
|
Soiza-Reilly M, Meye FJ, Olusakin J, Telley L, Petit E, Chen X, Mameli M, Jabaudon D, Sze JY, Gaspar P. SSRIs target prefrontal to raphe circuits during development modulating synaptic connectivity and emotional behavior. Mol Psychiatry 2019; 24:726-745. [PMID: 30279456 PMCID: PMC6445781 DOI: 10.1038/s41380-018-0260-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/08/2018] [Accepted: 09/06/2018] [Indexed: 12/24/2022]
Abstract
Antidepressants that block the serotonin transporter, (Slc6a4/SERT), selective serotonin reuptake inhibitors (SSRIs) improve mood in adults but have paradoxical long-term effects when administered during perinatal periods, increasing the risk to develop anxiety and depression. The basis for this developmental effect is not known. Here, we show that during an early postnatal period in mice (P0-P10), Slc6a4/SERT is transiently expressed in a subset of layer 5-6 pyramidal neurons of the prefrontal cortex (PFC). PFC-SERT+ neurons establish glutamatergic synapses with subcortical targets, including the serotonin (5-HT) and GABA neurons of the dorsal raphe nucleus (DRN). PFC-to-DRN circuits develop postnatally, coinciding with the period of PFC Slc6a4/SERT expression. Complete or cortex-specific ablation of SERT increases the number of functional PFC glutamate synapses on both 5-HT and GABA neurons in the DRN. This PFC-to-DRN hyperinnervation is replicated by early-life exposure to the SSRI, fluoxetine (from P2 to P14), that also causes anxiety/depressive-like symptoms. We show that pharmacogenetic manipulation of PFC-SERT+ neuron activity bidirectionally modulates these symptoms, suggesting that PFC hypofunctionality has a causal role in these altered responses to stress. Overall, our data identify specific PFC descending circuits that are targets of antidepressant drugs during development. We demonstrate that developmental expression of SERT in this subset of PFC neurons controls synaptic maturation of PFC-to-DRN circuits, and that remodeling of these circuits in early life modulates behavioral responses to stress in adulthood.
Collapse
Affiliation(s)
- M. Soiza-Reilly
- 0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, Paris, France ,0000000121866389grid.7429.8Inserm, UMR-S 839, Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, Paris, France
| | - F. J. Meye
- 0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, Paris, France ,0000000121866389grid.7429.8Inserm, UMR-S 839, Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, Paris, France
| | - J. Olusakin
- 0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, Paris, France ,0000000121866389grid.7429.8Inserm, UMR-S 839, Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, Paris, France
| | - L. Telley
- 0000 0001 2322 4988grid.8591.5Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - E. Petit
- 0000000121791997grid.251993.5Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York USA
| | - X. Chen
- 0000000121791997grid.251993.5Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York USA
| | - M. Mameli
- 0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, Paris, France ,0000000121866389grid.7429.8Inserm, UMR-S 839, Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, Paris, France
| | - D. Jabaudon
- 0000 0001 2322 4988grid.8591.5Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - J.-Y. Sze
- 0000000121791997grid.251993.5Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York USA
| | - P. Gaspar
- 0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, Paris, France ,0000000121866389grid.7429.8Inserm, UMR-S 839, Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, Paris, France
| |
Collapse
|
31
|
Abstract
The gastrointestinal tract contains its own set of intrinsic neuroglial circuits - the enteric nervous system (ENS) - which detects and responds to diverse signals from the environment. Here, we address recent advances in the understanding of ENS development, including how neural-crest-derived progenitors migrate into and colonize the bowel, the formation of ganglionated plexuses and the molecular mechanisms of enteric neuronal and glial diversification. Modern lineage tracing and transcription-profiling technologies have produced observations that simultaneously challenge and affirm long-held beliefs about ENS development. We review many genetic and environmental factors that can alter ENS development and exert long-lasting effects on gastrointestinal function, and discuss how developmental defects in the ENS might account for some of the large burden of digestive disease.
Collapse
Affiliation(s)
- Meenakshi Rao
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Michael D Gershon
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
32
|
Janušonis S, Detering N. A stochastic approach to serotonergic fibers in mental disorders. Biochimie 2018; 161:15-22. [PMID: 30056260 DOI: 10.1016/j.biochi.2018.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 07/24/2018] [Indexed: 01/09/2023]
Abstract
Virtually all brain circuits are physically embedded in a three-dimensional matrix of fibers that release 5-hydroxytryptamine (5-HT, serotonin). The density of this matrix varies across brain regions and cortical laminae, and it is altered in some mental disorders, including Major Depressive Disorder and Autism Spectrum Disorder. We investigate how the regional structure of the serotonergic matrix depends on the stochastic behavior of individual serotonergic fibers and introduce a new framework for the quantitative analysis of this behavior. In particular, we show that a step-wise random walk, based on the von Mises-Fisher probability distribution, can provide a realistic and mathematically concise description of these fibers. We also consider other stochastic models, including the fractional Brownian motion. The proposed approach seeks to advance the current understanding of the ascending reticular activating system (ARAS) and may also support future theory-guided therapeutic approaches.
Collapse
Affiliation(s)
- Skirmantas Janušonis
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, CA, 93106-9660, USA.
| | - Nils Detering
- Department of Statistics and Applied Probability, University of California, Santa Barbara, CA, 93106-3110, USA
| |
Collapse
|
33
|
Brindley RL, Bauer MB, Walker LA, Quinlan MA, Carneiro AMD, Sze JY, Blakely RD, Currie KPM. Adrenal serotonin derives from accumulation by the antidepressant-sensitive serotonin transporter. Pharmacol Res 2018; 140:56-66. [PMID: 29894763 DOI: 10.1016/j.phrs.2018.06.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 06/06/2018] [Accepted: 06/07/2018] [Indexed: 12/30/2022]
Abstract
Adrenal chromaffin cells comprise the neuroendocrine arm of the sympathetic nervous system and secrete catecholamines to coordinate the appropriate stress response. Deletion of the serotonin (5-HT) transporter (SERT) gene in mice (SERT-/- mice) or pharmacological block of SERT function in rodents and humans augments this sympathoadrenal stress response (epinephrine secretion). The prevailing assumption is that loss of CNS SERT alters central drive to the peripheral sympathetic nervous system. Adrenal chromaffin cells also prominently express SERT where it might coordinate accumulation of 5-HT for reuse in the autocrine control of stress-evoked catecholamine secretion. To help test this hypothesis, we have generated a novel mouse model with selective excision of SERT in the peripheral sympathetic nervous system (SERTΔTH), generated by crossing floxed SERT mice with tyrosine hydroxylase Cre driver mice. SERT expression, assessed by western blot, was abolished in the adrenal gland but not perturbed in the CNS of SERTΔTH mice. SERT-mediated [3H] 5-HT uptake was unaltered in midbrain, hindbrain, and spinal cord synaptosomes, confirming transporter function was intact in the CNS. Endogenous midbrain and whole blood 5-HT homeostasis was unperturbed in SERTΔTH mice, contrasting with the depleted 5-HT content in SERT-/- mice. Selective SERT excision reduced adrenal gland 5-HT content by ≈ 50% in SERTΔTH mice but had no effect on adrenal catecholamine content. This novel model confirms that SERT expressed in adrenal chromaffin cells is essential for maintaining wild-type levels of 5-HT and provides a powerful tool to help dissect the role of SERT in the sympathetic stress response.
Collapse
Affiliation(s)
- Rebecca L Brindley
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA; Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Mary Beth Bauer
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville TN, USA
| | - L Anne Walker
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Meagan A Quinlan
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville TN, USA; Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, USA
| | - Ana M D Carneiro
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Ji-Ying Sze
- Department of Molecular Pharmacology and Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, USA; Florida Atlantic University Brain Institute, Jupiter, FL, USA
| | - Kevin P M Currie
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA; Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville TN, USA.
| |
Collapse
|
34
|
Garcia LP, Witteveen JS, Middelman A, van Hulten JA, Martens GJM, Homberg JR, Kolk SM. Perturbed Developmental Serotonin Signaling Affects Prefrontal Catecholaminergic Innervation and Cortical Integrity. Mol Neurobiol 2018; 56:1405-1420. [PMID: 29948943 PMCID: PMC6400880 DOI: 10.1007/s12035-018-1105-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 05/03/2018] [Indexed: 11/26/2022]
Abstract
Proper development of the medial prefrontal cortex (mPFC), crucial for correct cognitive functioning, requires projections from, among others, the serotonergic (5-HT) and catecholaminergic systems, but it is unclear how these systems influence each other during development. Here, we describe the parallel development of the 5-HT and catecholaminergic prefrontal projection systems in rat and demonstrate a close engagement of both systems in the proximity of Cajal-Retzius cells. We further show that in the absence of the 5-HT transporter (5-HTT), not only the developing 5-HT but also the catecholaminergic system, including their projections towards the mPFC, are affected. In addition, the layer identity of the mPFC neurons and reelin-positive interneuron number and integration are altered in the absence of the 5-HTT. Together, our data demonstrate a functional interplay between the developing mPFC 5-HT and catecholaminergic systems, and call for a holistic approach in studying neurotransmitter systems-specific developmental consequences for adult behavior, to eventually allow the design of better treatment strategies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Lidiane P Garcia
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Molecular Animal Physiology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Josefine S Witteveen
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Molecular Animal Physiology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Anthonieke Middelman
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Cognitive Neuroscience, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Josephus A van Hulten
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Molecular Animal Physiology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Gerard J M Martens
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Molecular Animal Physiology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Judith R Homberg
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Cognitive Neuroscience, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Sharon M Kolk
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Molecular Animal Physiology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
35
|
Ellegood J, Yee Y, Kerr TM, Muller CL, Blakely RD, Henkelman RM, Veenstra-VanderWeele J, Lerch JP. Analysis of neuroanatomical differences in mice with genetically modified serotonin transporters assessed by structural magnetic resonance imaging. Mol Autism 2018; 9:24. [PMID: 29651330 PMCID: PMC5894125 DOI: 10.1186/s13229-018-0210-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/21/2018] [Indexed: 02/03/2023] Open
Abstract
Background The serotonin (5-HT) system has long been implicated in autism spectrum disorder (ASD) as indicated by elevated whole blood and platelet 5-HT, altered platelet and brain receptor and transporter binding, and genetic linkage and association findings. Based upon work in genetically modified mice, 5-HT is known to influence several aspects of brain development, but systematic neuroimaging studies have not previously been reported. In particular, the 5-HT transporter (serotonin transporter, SERT; 5-HTT) gene, Slc6a4, has been extensively studied. Methods Using a 7-T MRI and deformation-based morphometry, we assessed neuroanatomical differences in an Slc6a4 knockout mouse on a C57BL/6 genetic background, along with an Slc6a4 Ala56 knockin mouse on two different genetic backgrounds (129S and C57BL/6). Results Individually (same sex, same background, same genotype), the only differences found were in the female Slc6a4 knockout mouse; all the others had no significant differences. However, an analysis of variance across the whole study sample revealed a significant effect of Slc6a4 on the amygdala, thalamus, dorsal raphe nucleus, and lateral and frontal cortices. Conclusions This work shows that an increase or decrease in SERT function has a significant effect on the neuroanatomy in 5-HT relevant regions, particularly the raphe nuclei. Notably, the Slc6a4 Ala56 knockin alone appears to have an insignificant, but suggestive, effect compared to the KO, which is consistent with Slc6a4 function. Despite the small number of 5-HT neurons and their localization to the brainstem, it is clear that 5-HT plays an important role in neuroanatomical organization. Electronic supplementary material The online version of this article (10.1186/s13229-018-0210-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jacob Ellegood
- 1Mouse Imaging Centre (MICe), Hospital for Sick Children, 25 Orde Street, Toronto, Ontario M5T 3H7 Canada
| | - Yohan Yee
- 1Mouse Imaging Centre (MICe), Hospital for Sick Children, 25 Orde Street, Toronto, Ontario M5T 3H7 Canada.,4Department of Medical Biophysics, University of Toronto, Toronto, ON M5S Canada
| | - Travis M Kerr
- 3Department of Psychiatry, Vanderbilt University, Nashville, TN 37235 USA
| | | | - Randy D Blakely
- 2Department of Pharmacology, Vanderbilt University, Nashville, TN 37235 USA.,3Department of Psychiatry, Vanderbilt University, Nashville, TN 37235 USA.,5Department of Biomedical Science and Brain Institute, Florida Atlantic University, Jupiter, FL 33431 USA
| | - R Mark Henkelman
- 1Mouse Imaging Centre (MICe), Hospital for Sick Children, 25 Orde Street, Toronto, Ontario M5T 3H7 Canada.,4Department of Medical Biophysics, University of Toronto, Toronto, ON M5S Canada
| | - Jeremy Veenstra-VanderWeele
- 2Department of Pharmacology, Vanderbilt University, Nashville, TN 37235 USA.,6Department of Psychiatry, Columbia University, New York, NY 10027 USA
| | - Jason P Lerch
- 1Mouse Imaging Centre (MICe), Hospital for Sick Children, 25 Orde Street, Toronto, Ontario M5T 3H7 Canada.,4Department of Medical Biophysics, University of Toronto, Toronto, ON M5S Canada
| |
Collapse
|
36
|
Wang B, Iyengar R, Li-Harms X, Joo JH, Wright C, Lavado A, Horner L, Yang M, Guan JL, Frase S, Green DR, Cao X, Kundu M. The autophagy-inducing kinases, ULK1 and ULK2, regulate axon guidance in the developing mouse forebrain via a noncanonical pathway. Autophagy 2017; 14:796-811. [PMID: 29099309 DOI: 10.1080/15548627.2017.1386820] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Mammalian ULK1 (unc-51 like kinase 1) and ULK2, Caenorhabditis elegans UNC-51, and Drosophila melanogaster Atg1 are serine/threonine kinases that regulate flux through the autophagy pathway in response to various types of cellular stress. C. elegans UNC-51 and D. melanogaster Atg1 also promote axonal growth and defasciculation; disruption of these genes results in defective axon guidance in invertebrates. Although disrupting ULK1/2 function impairs normal neurite outgrowth in vitro, the role of ULK1 and ULK2 in the developing brain remains poorly characterized. Here, we show that ULK1 and ULK2 are required for proper projection of axons in the forebrain. Mice lacking Ulk1 and Ulk2 in their central nervous systems showed defects in axonal pathfinding and defasciculation affecting the corpus callosum, anterior commissure, corticothalamic axons and thalamocortical axons. These defects impaired the midline crossing of callosal axons and caused hypoplasia of the anterior commissure and disorganization of the somatosensory cortex. The axon guidance defects observed in ulk1/2 double-knockout mice and central nervous system-specific (Nes-Cre) Ulk1/2-conditional double-knockout mice were not recapitulated in mice lacking other autophagy genes (i.e., Atg7 or Rb1cc1 [RB1-inducible coiled-coil 1]). The brains of Ulk1/2-deficient mice did not show stem cell defects previously attributed to defective autophagy in ambra1 (autophagy/Beclin 1 regulator 1)- and Rb1cc1-deficient mice or accumulation of SQSTM1 (sequestosome 1)+ or ubiquitin+ deposits. Together, these data demonstrate that ULK1 and ULK2 regulate axon guidance during mammalian brain development via a noncanonical (i.e., autophagy-independent) pathway.
Collapse
Affiliation(s)
- Bo Wang
- a Department of Pathology , St. Jude Children's Research Hospital , Memphis , TN , USA.,f Integrated Biomedical Sciences Program , the University of Tennessee Health Science Center , Memphis , TN , USA
| | - Rekha Iyengar
- a Department of Pathology , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Xiujie Li-Harms
- a Department of Pathology , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Joung Hyuck Joo
- a Department of Pathology , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Christopher Wright
- a Department of Pathology , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Alfonso Lavado
- b Department of Developmental Neurobiology , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Linda Horner
- d Cell and Tissue Imaging Center , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Mao Yang
- c Department of Immunology , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Jun-Lin Guan
- e Department of Cancer Biology , University of Cincinnati College of Medicine , Cincinnati , OH , USA
| | - Sharon Frase
- d Cell and Tissue Imaging Center , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Douglas R Green
- c Department of Immunology , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Xinwei Cao
- b Department of Developmental Neurobiology , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Mondira Kundu
- a Department of Pathology , St. Jude Children's Research Hospital , Memphis , TN , USA
| |
Collapse
|
37
|
Carter DA. Molecular phenotyping of transient postnatal tyrosine hydroxylase neurons in the rat bed nucleus of the stria terminalis. J Chem Neuroanat 2017; 82:29-38. [DOI: 10.1016/j.jchemneu.2017.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/08/2017] [Accepted: 04/10/2017] [Indexed: 01/18/2023]
|
38
|
Song NN, Huang Y, Yu X, Lang B, Ding YQ, Zhang L. Divergent Roles of Central Serotonin in Adult Hippocampal Neurogenesis. Front Cell Neurosci 2017; 11:185. [PMID: 28713247 PMCID: PMC5492328 DOI: 10.3389/fncel.2017.00185] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/16/2017] [Indexed: 11/13/2022] Open
Abstract
The central serotonin (5-HT) system is the main target of selective serotonin reuptake inhibitors (SSRIs), the first-line antidepressants widely used in current general practice. One of the prominent features of chronic SSRI treatment in rodents is the enhanced adult neurogenesis in the hippocampus, which has been proposed to contribute to antidepressant effects. Therefore, tremendous effort has been made to decipher how central 5-HT regulates adult hippocampal neurogenesis. In this paper, we review how changes in the central serotonergic system alter adult hippocampal neurogenesis. We focus on data obtained from three categories of genetically engineered mouse models: (1) mice with altered central 5-HT levels from embryonic stages, (2) mice with deletion of 5-HT receptors from embryonic stages, and (3) mice with altered central 5-HT system exclusively in adulthood. These recent findings provide unique insights to interpret the multifaceted roles of central 5-HT on adult hippocampal neurogenesis and its associated effects on depression.
Collapse
Affiliation(s)
- Ning-Ning Song
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Ying Huang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Xin Yu
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Bing Lang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China.,Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Key Laboratory of Psychiatry and Mental Health of Hunan Province, Central South UniversityChangsha, China
| | - Yu-Qiang Ding
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Lei Zhang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| |
Collapse
|
39
|
Millard SJ, Weston-Green K, Newell KA. The effects of maternal antidepressant use on offspring behaviour and brain development: Implications for risk of neurodevelopmental disorders. Neurosci Biobehav Rev 2017. [PMID: 28629713 DOI: 10.1016/j.neubiorev.2017.06.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Approximately 10% of pregnant women are prescribed antidepressant drugs (ADDs), with selective serotonin reuptake inhibitors (SSRIs) the most widely prescribed. SSRIs bind to the serotonin transporter (SERT), blocking the reabsorption of serotonin by the presynaptic neuron and increasing serotonin levels in the synaptic cleft. The serotonergic system regulates a range of brain development processes including neuronal proliferation, migration, differentiation and synaptogenesis. Given the presence of SERT in early brain development, coupled with the ability of SSRIs to cross the placenta and also enter breast milk, concerns have been raised regarding the effects of SSRI exposure on the developing foetus and newborns. In this review, we evaluate preclinical and clinical studies that have examined the effects of maternal SSRI exposure and the risk for altered neurodevelopment and associated behaviours in offspring. While the current body of evidence suggests that maternal SSRI treatment may cause perturbations to the neurobiology, behaviour and ultimately risk for neurodevelopmental disorders in exposed offspring, conflicting findings do exist and the evidence is not conclusive. However, given the increasing incidence of depression and number of women prescribed ADDs during pregnancy, further investigation into this area is warranted.
Collapse
Affiliation(s)
- Samuel J Millard
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales 2522, Australia; Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522, Australia.
| | - Katrina Weston-Green
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales 2522, Australia; Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522, Australia.
| | - Kelly A Newell
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales 2522, Australia; Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522, Australia.
| |
Collapse
|
40
|
Teissier A, Soiza-Reilly M, Gaspar P. Refining the Role of 5-HT in Postnatal Development of Brain Circuits. Front Cell Neurosci 2017; 11:139. [PMID: 28588453 PMCID: PMC5440475 DOI: 10.3389/fncel.2017.00139] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 04/26/2017] [Indexed: 11/30/2022] Open
Abstract
Changing serotonin (5-hydroxytryptamine, 5-HT) brain levels during critical periods in development has long-lasting effects on brain function, particularly on later anxiety/depression-related behaviors in adulthood. A large part of the known developmental effects of 5-HT occur during critical periods of postnatal life, when activity-dependent mechanisms remodel neural circuits. This was first demonstrated for the maturation of sensory brain maps in the barrel cortex and the visual system. More recently this has been extended to the 5-HT raphe circuits themselves and to limbic circuits. Recent studies overviewed here used new genetic models in mice and rats and combined physiological and structural approaches to provide new insights on the cellular and molecular mechanisms controlled by 5-HT during late stages of neural circuit maturation in the raphe projections, the somatosensory cortex and the visual system. Similar mechanisms appear to be also involved in the maturation of limbic circuits such as prefrontal circuits. The latter are of particular relevance to understand the impact of transient 5-HT dysfunction during postnatal life on psychiatric illnesses and emotional disorders in adult life.
Collapse
Affiliation(s)
- Anne Teissier
- Institut du Fer à Moulin, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S839Paris, France.,Université Pierre et Marie CurieParis, France.,Institut du Fer à MoulinParis, France
| | - Mariano Soiza-Reilly
- Institut du Fer à Moulin, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S839Paris, France.,Université Pierre et Marie CurieParis, France.,Institut du Fer à MoulinParis, France
| | - Patricia Gaspar
- Institut du Fer à Moulin, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S839Paris, France.,Université Pierre et Marie CurieParis, France.,Institut du Fer à MoulinParis, France
| |
Collapse
|
41
|
Brindley RL, Bauer MB, Blakely RD, Currie KP. Serotonin and Serotonin Transporters in the Adrenal Medulla: A Potential Hub for Modulation of the Sympathetic Stress Response. ACS Chem Neurosci 2017; 8:943-954. [PMID: 28406285 PMCID: PMC5541362 DOI: 10.1021/acschemneuro.7b00026] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Serotonin (5-HT) is an important neurotransmitter in the central nervous system where it modulates circuits involved in mood, cognition, movement, arousal, and autonomic function. The 5-HT transporter (SERT; SLC6A4) is a key regulator of 5-HT signaling, and genetic variations in SERT are associated with various disorders including depression, anxiety, and autism. This review focuses on the role of SERT in the sympathetic nervous system. Autonomic/sympathetic dysfunction is evident in patients with depression, anxiety, and other diseases linked to serotonergic signaling. Experimentally, loss of SERT function (SERT knockout mice or chronic pharmacological block) has been reported to augment the sympathetic stress response. Alterations to serotonergic signaling in the CNS and thus central drive to the peripheral sympathetic nervous system are presumed to underlie this augmentation. Although less widely recognized, SERT is robustly expressed in chromaffin cells of the adrenal medulla, the neuroendocrine arm of the sympathetic nervous system. Adrenal chromaffin cells do not synthesize 5-HT but accumulate small amounts by SERT-mediated uptake. Recent evidence demonstrated that 5-HT1A receptors inhibit catecholamine secretion from adrenal chromaffin cells via an atypical mechanism that does not involve modulation of cellular excitability or voltage-gated Ca2+ channels. This raises the possibility that the adrenal medulla is a previously unrecognized peripheral hub for serotonergic control of the sympathetic stress response. As a framework for future investigation, a model is proposed in which stress-evoked adrenal catecholamine secretion is fine-tuned by SERT-modulated autocrine 5-HT signaling.
Collapse
Affiliation(s)
- Rebecca L. Brindley
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mary Beth Bauer
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Randy D. Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, and Florida Atlantic University Brain Institute, Jupiter, FL, USA
| | - Kevin P.M. Currie
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
42
|
Siemann JK, Muller CL, Forsberg CG, Blakely RD, Veenstra-VanderWeele J, Wallace MT. An autism-associated serotonin transporter variant disrupts multisensory processing. Transl Psychiatry 2017; 7:e1067. [PMID: 28323282 PMCID: PMC5416665 DOI: 10.1038/tp.2017.17] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/29/2016] [Accepted: 01/09/2017] [Indexed: 01/29/2023] Open
Abstract
Altered sensory processing is observed in many children with autism spectrum disorder (ASD), with growing evidence that these impairments extend to the integration of information across the different senses (that is, multisensory function). The serotonin system has an important role in sensory development and function, and alterations of serotonergic signaling have been suggested to have a role in ASD. A gain-of-function coding variant in the serotonin transporter (SERT) associates with sensory aversion in humans, and when expressed in mice produces traits associated with ASD, including disruptions in social and communicative function and repetitive behaviors. The current study set out to test whether these mice also exhibit changes in multisensory function when compared with wild-type (WT) animals on the same genetic background. Mice were trained to respond to auditory and visual stimuli independently before being tested under visual, auditory and paired audiovisual (multisensory) conditions. WT mice exhibited significant gains in response accuracy under audiovisual conditions. In contrast, although the SERT mutant animals learned the auditory and visual tasks comparably to WT littermates, they failed to show behavioral gains under multisensory conditions. We believe these results provide the first behavioral evidence of multisensory deficits in a genetic mouse model related to ASD and implicate the serotonin system in multisensory processing and in the multisensory changes seen in ASD.
Collapse
Affiliation(s)
- J K Siemann
- Neuroscience Program, Vanderbilt University, Nashville, TN, USA
| | - C L Muller
- Neuroscience Program, Vanderbilt University, Nashville, TN, USA
| | - C G Forsberg
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA
| | - R D Blakely
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA
- Silvio O. Conte Center for Neuroscience Research, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter, FL, USA
- Florida Atlantic University Brain Institute, Florida Atlantic University, Jupiter, FL, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - J Veenstra-VanderWeele
- Silvio O. Conte Center for Neuroscience Research, Vanderbilt University, Nashville, TN, USA
- Department of Psychiatry, Sackler Institute for Developmental Psychobiology, Columbia University, New York, NY, USA
- Center for Autism and The Developing Brain, New York Presbyterian Hospital, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - M T Wallace
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA
- Silvio O. Conte Center for Neuroscience Research, Vanderbilt University, Nashville, TN, USA
- Department of Psychology, Vanderbilt University, Nashville, TN, USA
- Department of Hearing and Speech Sciences, Vanderbilt University, Nashville, TN, USA
- Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
43
|
Miceli S, Nadif Kasri N, Joosten J, Huang C, Kepser L, Proville R, Selten MM, van Eijs F, Azarfar A, Homberg JR, Celikel T, Schubert D. Reduced Inhibition within Layer IV of Sert Knockout Rat Barrel Cortex is Associated with Faster Sensory Integration. Cereb Cortex 2017; 27:933-949. [PMID: 28158484 PMCID: PMC5390402 DOI: 10.1093/cercor/bhx016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 12/07/2016] [Accepted: 01/11/2017] [Indexed: 12/19/2022] Open
Abstract
Neural activity is essential for the maturation of sensory systems. In the rodent primary somatosensory cortex (S1), high extracellular serotonin (5-HT) levels during development impair neural transmission between the thalamus and cortical input layer IV (LIV). Rodent models of impaired 5-HT transporter (SERT) function show disruption in their topological organization of S1 and in the expression of activity-regulated genes essential for inhibitory cortical network formation. It remains unclear how such alterations affect the sensory information processing within cortical LIV. Using serotonin transporter knockout (Sert-/-) rats, we demonstrate that high extracellular serotonin levels are associated with impaired feedforward inhibition (FFI), fewer perisomatic inhibitory synapses, a depolarized GABA reversal potential and reduced expression of KCC2 transporters in juvenile animals. At the neural population level, reduced FFI increases the excitatory drive originating from LIV, facilitating evoked representations in the supragranular layers II/III. The behavioral consequence of these changes in network excitability is faster integration of the sensory information during whisker-based tactile navigation, as Sert-/- rats require fewer whisker contacts with tactile targets and perform object localization with faster reaction times. These results highlight the association of serotonergic homeostasis with formation and excitability of sensory cortical networks, and consequently with sensory perception.
Collapse
Affiliation(s)
- Stéphanie Miceli
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
- Department of Neural Networks, Center of Advanced European Studies and Research (caesar), Max Planck Society, Germany
| | - Nael Nadif Kasri
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Joep Joosten
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Chao Huang
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Lara Kepser
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Rémi Proville
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Martijn M. Selten
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Fenneke van Eijs
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Alireza Azarfar
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Judith R. Homberg
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Tansu Celikel
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Dirk Schubert
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
44
|
Stemme T, Stern M, Bicker G. Serotonin-containing neurons in basal insects: In search of ground patterns among tetraconata. J Comp Neurol 2017; 525:79-115. [PMID: 27203729 DOI: 10.1002/cne.24043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/17/2016] [Accepted: 05/18/2016] [Indexed: 11/08/2022]
Abstract
The ventral nerve cord of Tetraconata contains a comparably low number of serotonin-immunoreactive neurons, facilitating individual identification of cells and their characteristic neurite morphology. This offers the rather unique possibility of establishing homologies at the single cell level. Because phylogenetic relationships within Tetraconata are still discussed controversially, comparisons of individually identifiable neurons can help to unravel these issues. Serotonin immunoreactivity has been investigated in numerous tetraconate taxa, leading to reconstructions of hypothetical ground patterns for major lineages. However, detailed descriptions of basal insects are still missing, but are crucial for meaningful evolutionary considerations. We investigated the morphology of individually identifiable serotonin-immunoreactive neurons in the ventral nerve cord of Zygentoma (Thermobia domestica, Lepisma saccharina, Atelura formicaria) and Archaeognatha (Machilis germanica, Dilta hibernica). To improve immunocytochemical resolution, we also performed preincubation experiments with 5-hydroxy-L-tryptophan and serotonin. Additionally, we checked for immunolabeling of tryptophan hydroxylase, an enzyme associated with the synthesis of serotonin. Besides the generally identified groups of anterolateral, medial, and posterolateral neurons within each ganglion of the ventral nerve cord, we identified several other immunoreactive cells, which seem to have no correspondence in other tetraconates. Furthermore, we show that not all immunoreactive neurons produce serotonin, but have the capability for serotonin uptake. Comparisons with the patterns of serotonin-containing neurons in major tetraconate taxa suggest a close phylogenetic relationship of Remipedia, Cephalocarida, and Hexapoda, supporting the Miracrustacea hypothesis. J. Comp. Neurol., 2016. © 2016 Wiley Periodicals, Inc. J. Comp. Neurol. 525:79-115, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Torben Stemme
- University of Veterinary Medicine Hannover, Division of Cell Biology, D-30173, Hannover, Germany
| | - Michael Stern
- University of Veterinary Medicine Hannover, Division of Cell Biology, D-30173, Hannover, Germany
| | - Gerd Bicker
- University of Veterinary Medicine Hannover, Division of Cell Biology, D-30173, Hannover, Germany
| |
Collapse
|
45
|
Muller CL, Anacker AMJ, Rogers TD, Goeden N, Keller EH, Forsberg CG, Kerr TM, Wender CLA, Anderson GM, Stanwood GD, Blakely RD, Bonnin A, Veenstra-VanderWeele J. Impact of Maternal Serotonin Transporter Genotype on Placental Serotonin, Fetal Forebrain Serotonin, and Neurodevelopment. Neuropsychopharmacology 2017; 42:427-436. [PMID: 27550733 PMCID: PMC5399236 DOI: 10.1038/npp.2016.166] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 07/16/2016] [Accepted: 08/04/2016] [Indexed: 12/29/2022]
Abstract
Biomarker, neuroimaging, and genetic findings implicate the serotonin transporter (SERT) in autism spectrum disorder (ASD). Previously, we found that adult male mice expressing the autism-associated SERT Ala56 variant have altered central serotonin (5-HT) system function, as well as elevated peripheral blood 5-HT levels. Early in gestation, before midbrain 5-HT projections have reached the cortex, peripheral sources supply 5-HT to the forebrain, suggesting that altered maternal or placenta 5-HT system function could impact the developing embryo. We therefore used different combinations of maternal and embryo SERT Ala56 genotypes to examine effects on blood, placenta and embryo serotonin levels and neurodevelopment at embryonic day E14.5, when peripheral sources of 5-HT predominate, and E18.5, when midbrain 5-HT projections have reached the forebrain. Maternal SERT Ala56 genotype was associated with decreased placenta and embryonic forebrain 5-HT levels at E14.5. Low 5-HT in the placenta persisted, but forebrain levels normalized by E18.5. Maternal SERT Ala56 genotype effects on forebrain 5-HT levels were accompanied by a broadening of 5-HT-sensitive thalamocortical axon projections. In contrast, no effect of embryo genotype was seen in concepti from heterozygous dams. Blood 5-HT levels were dynamic across pregnancy and were increased in SERT Ala56 dams at E14.5. Placenta RNA sequencing data at E14.5 indicated substantial impact of maternal SERT Ala56 genotype, with alterations in immune and metabolic-related pathways. Collectively, these findings indicate that maternal SERT function impacts offspring placental 5-HT levels, forebrain 5-HT levels, and neurodevelopment.
Collapse
Affiliation(s)
| | - Allison MJ Anacker
- Department of Psychiatry, Columbia University Medical School, New York, NY, USA
| | - Tiffany D Rogers
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Nick Goeden
- Department of Cell and Neurobiology and Zilkha Neurogenetic Institute, University of Southern California School of Medicine, Los Angeles, CA, USA
| | - Elizabeth H Keller
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - C Gunnar Forsberg
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Travis M Kerr
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Carly LA Wender
- Department of Psychiatry, Columbia University Medical School, New York, NY, USA
| | - George M Anderson
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA
| | - Gregg D Stanwood
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA
| | - Randy D Blakely
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Alexandre Bonnin
- Department of Cell and Neurobiology and Zilkha Neurogenetic Institute, University of Southern California School of Medicine, Los Angeles, CA, USA
| | - Jeremy Veenstra-VanderWeele
- Department of Psychiatry, Columbia University Medical School, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Center for Autism and the Developing Brain, New York Presbyterian Hospital, New York, NY, USA
- Sackler Institute for Developmental Psychobiology, Columbia University, New York, NY, USA
| |
Collapse
|
46
|
Park S, Nevin ABC, Cardozo-Pelaez F, Lurie DI. Pb exposure prolongs the time period for postnatal transient uptake of 5-HT by murine LSO neurons. Neurotoxicology 2016; 57:258-269. [PMID: 27771255 DOI: 10.1016/j.neuro.2016.10.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 09/27/2016] [Accepted: 10/17/2016] [Indexed: 01/09/2023]
Abstract
Pb exposure is associated with cognitive deficits including Attention Deficit Hyperactivity Disorder (ADHD) in children and alters auditory temporal processing in humans and animals. Serotonin has been implicated in auditory temporal processing and previous studies from our laboratory have demonstrated that developmental Pb decreases expression of serotonin (5-HT) in the adult murine lateral superior olive (LSO). During development, certain non-serotonergic sensory neurons, including auditory LSO neurons, transiently take up 5-HT through the serotonin reuptake transporter (SERT). The uptake of 5-HT is important for development of sensory systems. This study examines the effect of Pb on the serotonergic system in the LSO of the early postnatal mouse. Mice were exposed to moderate Pb (0.01mM) or high Pb (0.1mM) throughout gestation and postnatal day 4 (P4) and P8. We found that Pb exposure prolongs the normal developmental expression of 5-HT by LSO neurons and this is correlated with expression of SERT on LSO cell bodies. The prolonged expression of 5-HT by postnatal LSO neurons is correlated with decreased synaptic immunolabeling within the LSO. This Pb-associated decrease in synaptic density within the LSO could contribute to the auditory temporal processing deficits and cognitive deficits associated with developmental Pb exposure.
Collapse
Affiliation(s)
- Sunyoung Park
- Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, Department of Biomedical & Pharmaceutical Sciences, College of Health Professions and Biomedical Sciences, The University of Montana, Missoula, MT 59812, United States; Business Planning Department, Kyowa Hakko Kirin Korea Co., Ltd., Seoul, Republic of Korea
| | - Andrew B C Nevin
- Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, Department of Biomedical & Pharmaceutical Sciences, College of Health Professions and Biomedical Sciences, The University of Montana, Missoula, MT 59812, United States
| | - Fernando Cardozo-Pelaez
- Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, Department of Biomedical & Pharmaceutical Sciences, College of Health Professions and Biomedical Sciences, The University of Montana, Missoula, MT 59812, United States
| | - Diana I Lurie
- Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, Department of Biomedical & Pharmaceutical Sciences, College of Health Professions and Biomedical Sciences, The University of Montana, Missoula, MT 59812, United States.
| |
Collapse
|
47
|
Ascl1 Is Required for the Development of Specific Neuronal Subtypes in the Enteric Nervous System. J Neurosci 2016; 36:4339-50. [PMID: 27076429 DOI: 10.1523/jneurosci.0202-16.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 02/25/2016] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED The enteric nervous system (ENS) is organized into neural circuits within the gastrointestinal wall where it controls the peristaltic movements, secretion, and blood flow. Although proper gut function relies on the complex neuronal composition of the ENS, little is known about the transcriptional networks that regulate the diversification into different classes of enteric neurons and glia during development. Here we redefine the role of Ascl1 (Mash1), one of the few regulatory transcription factors described during ENS development. We show that enteric glia and all enteric neuronal subtypes appear to be derived from Ascl1-expressing progenitor cells. In the gut of Ascl1(-/-) mutant mice, neurogenesis is delayed and reduced, and posterior gliogenesis impaired. The ratio of neurons expressing Calbindin, TH, and VIP is selectively decreased while, for instance, 5-HT(+) neurons, which previously were believed to be Ascl1-dependent, are formed in normal numbers. Essentially the same differentiation defects are observed in Ascl1(KINgn2) transgenic mutants, where the proneural activity of Ngn2 replaces Ascl1, demonstrating that Ascl1 is required for the acquisition of specific enteric neuronal subtype features independent of its role in neurogenesis. In this study, we provide novel insights into the expression and function of Ascl1 in the differentiation process of specific neuronal subtypes during ENS development. SIGNIFICANCE STATEMENT The molecular mechanisms underlying the generation of different neuronal subtypes during development of the enteric nervous system are poorly understood despite its pivotal function in gut motility and involvement in gastrointestinal pathology. This report identifies novel roles for the transcription factor Ascl1 in enteric gliogenesis and neurogenesis. Moreover, independent of its proneurogenic activity, Ascl1 is required for the normal expression of specific enteric neuronal subtype characteristics. Distinct enteric neuronal subtypes are formed in a temporally defined order, and we observe that the early-born 5-HT(+) neurons are generated in Ascl1(-/-) mutants, despite the delayed neurogenesis. Enteric nervous system progenitor cells may therefore possess strong intrinsic control over their specification at the initial waves of neurogenesis.
Collapse
|
48
|
Chen X, Petit EI, Dobrenis K, Sze JY. Spatiotemporal SERT expression in cortical map development. Neurochem Int 2016; 98:129-37. [PMID: 27282696 PMCID: PMC4969137 DOI: 10.1016/j.neuint.2016.05.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 05/03/2016] [Accepted: 05/17/2016] [Indexed: 11/21/2022]
Abstract
The cerebral cortex is organized into morphologically distinct areas that provide biological frameworks underlying perception, cognition, and behavior. Profiling mouse and human cortical transcriptomes have revealed temporal-specific differential gene expression modules in distinct neocortical areas during cortical map establishment. However, the biological roles of spatiotemporal gene expression in cortical patterning and how cortical topographic gene expression is regulated are largely unknown. Here, we characterize temporal- and spatial-defined expression of serotonin (5-HT) transporter (SERT) in glutamatergic neurons during sensory map development in mice. SERT is transiently expressed in glutamatergic thalamic neurons projecting to sensory cortices and in pyramidal neurons in the prefrontal cortex (PFC) and hippocampus (HPC) during the period that lays down the basic functional neural circuits. We previously identified that knockout of SERT in the thalamic neurons blocks 5-HT uptake by their thalamocortical axons, resulting in excessive 5-HT signaling that impairs sensory map architecture. In contrast, here we show that selective SERT knockout in the PFC and HPC neurons does not perturb sensory map patterning. These data suggest that transient SERT expression in specific glutamatergic neurons provides area-specific instructions for cortical map patterning. Hence, genetic and pharmacological manipulations of this SERT function could illuminate the fundamental genetic programming of cortex-specific maps and biological roles of temporal-specific cortical topographic gene expression in normal development and mental disorders.
Collapse
Affiliation(s)
- Xiaoning Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Emilie I Petit
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kostantin Dobrenis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ji Ying Sze
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
49
|
Brindley RL, Bauer MB, Blakely RD, Currie KPM. An interplay between the serotonin transporter (SERT) and 5-HT receptors controls stimulus-secretion coupling in sympathoadrenal chromaffin cells. Neuropharmacology 2016; 110:438-448. [PMID: 27544824 DOI: 10.1016/j.neuropharm.2016.08.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 08/04/2016] [Accepted: 08/15/2016] [Indexed: 11/16/2022]
Abstract
Adrenal chromaffin cells (ACCs), the neuroendocrine arm of the sympathetic nervous system, secrete catecholamines to mediate the physiological response to stress. Although ACCs do not synthesize 5-HT, they express the serotonin transporter (SERT). Genetic variations in SERT are linked to several CNS disorders but the role(s) of SERT/5-HT in ACCs has remained unclear. Adrenal glands from wild-type mice contained 5-HT at ≈ 750 fold lower abundance than adrenaline, and in SERT(-/-) mice this was reduced by ≈80% with no change in catecholamines. Carbon fibre amperometry showed that SERT modulated the ability of 5-HT1A receptors to inhibit exocytosis. 5-HT reduced the number of amperometric spikes (vesicular fusion events) evoked by KCl in SERT(-/-) cells and wild-type cells treated with escitalopram, a SERT antagonist. The 5-HT1A receptor antagonist WAY100635 blocked the inhibition by 5-HT which was mimicked by the 5-HT1A agonist 8-OH-DPAT but not the 5-HT1B agonist CP93129. There was no effect on voltage-gated Ca(2+) channels, K(+) channels, or intracellular [Ca(2+)] handling, showing the 5-HT receptors recruit an atypical inhibitory mechanism. Spike charge and kinetics were not altered by 5-HT receptors but were reduced in SERT(-/-) cells compared to wild-type cells. Our data reveal a novel role for SERT and suggest that adrenal chromaffin cells might be a previously unrecognized hub for serotonergic control of the sympathetic stress response.
Collapse
Affiliation(s)
- Rebecca L Brindley
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Mary Beth Bauer
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Randy D Blakely
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kevin P M Currie
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
50
|
Muller CL, Anacker AMJ, Veenstra-VanderWeele J. The serotonin system in autism spectrum disorder: From biomarker to animal models. Neuroscience 2016; 321:24-41. [PMID: 26577932 PMCID: PMC4824539 DOI: 10.1016/j.neuroscience.2015.11.010] [Citation(s) in RCA: 356] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 10/26/2015] [Accepted: 11/04/2015] [Indexed: 02/02/2023]
Abstract
Elevated whole blood serotonin, or hyperserotonemia, was the first biomarker identified in autism spectrum disorder (ASD) and is present in more than 25% of affected children. The serotonin system is a logical candidate for involvement in ASD due to its pleiotropic role across multiple brain systems both dynamically and across development. Tantalizing clues connect this peripheral biomarker with changes in brain and behavior in ASD, but the contribution of the serotonin system to ASD pathophysiology remains incompletely understood. Studies of whole blood serotonin levels in ASD and in a large founder population indicate greater heritability than for the disorder itself and suggest an association with recurrence risk. Emerging data from both neuroimaging and postmortem samples also indicate changes in the brain serotonin system in ASD. Genetic linkage and association studies of both whole blood serotonin levels and of ASD risk point to the chromosomal region containing the serotonin transporter (SERT) gene in males but not in females. In ASD families with evidence of linkage to this region, multiple rare SERT amino acid variants lead to a convergent increase in serotonin uptake in cell models. A knock-in mouse model of one of these variants, SERT Gly56Ala, recapitulates the hyperserotonemia biomarker and shows increased brain serotonin clearance, increased serotonin receptor sensitivity, and altered social, communication, and repetitive behaviors. Data from other rodent models also suggest an important role for the serotonin system in social behavior, in cognitive flexibility, and in sensory development. Recent work indicates that reciprocal interactions between serotonin and other systems, such as oxytocin, may be particularly important for social behavior. Collectively, these data point to the serotonin system as a prime candidate for treatment development in a subgroup of children defined by a robust, heritable biomarker.
Collapse
Affiliation(s)
- C L Muller
- Vanderbilt Brain Institute, Vanderbilt University, 465 21st Avenue South, Nashville, TN 37232, USA.
| | - A M J Anacker
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, Mail Unit 78, New York, NY 10032, USA.
| | - J Veenstra-VanderWeele
- Sackler Institute for Developmental Psychobiology, Department of Psychiatry, Columbia University; Center for Autism and the Developing Brain, New York Presbyterian Hospital; New York State Psychiatric Institute, 1051 Riverside Drive, Mail Unit 78, New York, NY 10032, USA.
| |
Collapse
|