1
|
Themann A, Rodriguez M, Calvo DE, Vargas P, Iñiguez SD. Prozac exposure during adolescence increases pain sensitivity in adulthood. J Psychiatr Res 2025; 186:200-204. [PMID: 40250326 DOI: 10.1016/j.jpsychires.2025.04.027] [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: 01/31/2025] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Psychotropic medication prescription rates, particularly of the selective serotonin reuptake inhibitor fluoxetine (FLX; Prozac), are on the rise in the female adolescent population. Particularly, because FLX is dispensed for the treatment of numerous mood-related illnesses, premenstrual dysphoric disorder, as well as for pain management. Although FLX is deemed efficacious shortly post/during chronic treatment, the potential for unexpected long-term side effects has not been thoroughly assessed. For this reason, in this brief report, we examined whether exposure to FLX, during adolescence, influences thermal nociception in adulthood. To do this, postnatal day (PD)-35 female C57BL/6 mice were exposed to FLX (250 mg/L in drinking water) for 15 consecutive days (PD35-49). Once mice reached adulthood (PD70) they were evaluated on thermal nociception sensitivity adopting the hot plate test. We found that adult mice with FLX history displayed reductions in body weight (g) as well as reduced time (s) to display hindpaw-licking behavior, when compared to controls. These outcomes suggest that juvenile FLX exposure induces thermal hyperalgesia in adulthood, thus questioning the safety of antidepressant exposure during early-life stages of development in the female population.
Collapse
Affiliation(s)
- Anapaula Themann
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Minerva Rodriguez
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Daniel E Calvo
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Paulina Vargas
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Sergio D Iñiguez
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, 79968, USA.
| |
Collapse
|
2
|
Ghai U, Chachra P, Mendon S, Janakiraman B, Fanibunda SE, Sarkar A, Gohil D, Jayaprasad AB, Kukkemane K, Singh V, Kolthur-Seetharam U, Vaidya VA. Postnatal and juvenile fluoxetine treatment evokes sex-specific, opposing effects on mood-related behavior, gene expression, mitochondrial function, and dendritic architecture in the rat medial prefrontal cortex. Biol Psychiatry 2025:S0006-3223(25)01188-6. [PMID: 40350070 DOI: 10.1016/j.biopsych.2025.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 04/19/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Serotonin shapes emotional neurocircuit development, and serotonergic neurotransmission is implicated in both the pathophysiology and treatment of neuropsychiatric disorders. The selective serotonin reuptake inhibitor, fluoxetine (Flx) is a common first-line treatment for childhood and adolescent mood disorders given a favourable risk-benefit profile. Using a rodent model we addressed specific long-term behavioral, molecular, bioenergetic and cytoarchitectural consequences of postnatal (PNFlx) and juvenile (JFlx) fluoxetine treatment. METHODS Rat pups received PNFlx (postnatal day 2: P2-P21) or JFlx (P28-48) treatment with the impact on anxiety- and despair-like behavior examined in adulthood, along with assessing global gene expression, mitochondrial function, and dendritic cytoarchitecture in the medial prefrontal cortex (mPFC). RESULTS PNFlx and JFlx evoked long-lasting, opposing changes in anxiety- and despair-like behavior in male, but not female, rats. The PNFlx- and JFlx-evoked increase and decrease in anxiety- and despair-like behavior respectively, were accompanied by distinctive, minimally overlapping, transcriptional changes in the mPFC in adulthood. Furthermore, we noted starkly differing outcomes of PNFlx and JFlx on mitochondrial function and dendritic cytoarchitecture in the mPFC. The PNFlx evoked despair-like behavior was reversed by adult-onset treatment with nicotinamide, a NAD+precursor that enhances mitochondrial bioenergetics. CONCLUSIONS Collectively, our findings highlight distinct developmental epochs wherein fluoxetine exposure can program long-term, sex-specific, opposing outcomes on mood-related behavior, accompanied by persistent changes in gene expression, mitochondrial function and neuronal cytoarchitecture in the mPFC in adulthood. This motivates future studies to examine a potential role for altered bioenergetics in shaping the differential impact of early fluoxetine treatment on emotionality.
Collapse
Affiliation(s)
- Utkarsha Ghai
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Parul Chachra
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Suchith Mendon
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Balaganesh Janakiraman
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Sashaina E Fanibunda
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India; Kasturba Health Society - Medical Research Centre, Mumbai 400056, India
| | - Ambalika Sarkar
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Dievya Gohil
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | | | - Kowshik Kukkemane
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Vivek Singh
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Ullas Kolthur-Seetharam
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Vidita A Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India.
| |
Collapse
|
3
|
Zanni G, van Dijk MT, Cagliostro MC, Sepulveda P, Pini N, Rose AL, Kesin AL, Lugo-Candelas C, Goncalves PD, MacKay AS, Iigaya K, Kulkarni P, Ferris CF, Weissman MM, Talati A, Ansorge MS, Gingrich JA. Perinatal SSRI exposure impacts innate fear circuit activation and behavior in mice and humans. Nat Commun 2025; 16:4002. [PMID: 40328752 PMCID: PMC12055977 DOI: 10.1038/s41467-025-58785-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/28/2025] [Indexed: 05/08/2025] Open
Abstract
Before assuming its role in the mature brain, serotonin modulates early brain development across phylogenetically diverse species. In mice and humans, early-life SSRI exposure alters the offspring's brain structure and is associated with anxiety and depression-related behaviors beginning in puberty. However, the impact of early-life SSRI exposure on brain circuit function is unknown. To address this question, we examined how developmental SSRI exposure changes fear-related brain activation and behavior in mice and humans. SSRI-exposed mice showed increased defense responses to a predator odor, and stronger fMRI amygdala and extended fear-circuit activation. Likewise, adolescents exposed to SSRIs in utero exhibited higher anxiety and depression symptoms than unexposed adolescents and also had greater activation of the amygdala and other limbic structures when processing fearful faces. These findings demonstrate that increases in anxiety and fear-related behaviors as well as brain circuit activation following developmental SSRI exposure are conserved between mice and humans. These findings have potential implications for the clinical use of SSRIs during human pregnancy and for designing interventions that protect fetal brain development.
Collapse
Affiliation(s)
- Giulia Zanni
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | - Milenna T van Dijk
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Division of Translational Epidemiology, New York State Psychiatric Institute, New York, NY, USA
| | - Martha Caffrey Cagliostro
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA
| | - Pradyumna Sepulveda
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Nicolò Pini
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | - Ariel L Rose
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | - Alexander L Kesin
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | - Claudia Lugo-Candelas
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Child and Adolescent Psychiatry, New York State Psychiatric Institute, New York, NY, USA
| | - Priscila Dib Goncalves
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Alexandra S MacKay
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | - Kiyohito Iigaya
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA
- Columbia Data Science Institute, Columbia University, New York, NY, USA
- Center for Theoretical Neuroscience, Zuckerman Institute for Mind Brain, and Behavior, Columbia University, New York, NY, USA
| | - Praveen Kulkarni
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA
| | - Craig F Ferris
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA
| | - Myrna M Weissman
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Division of Translational Epidemiology, New York State Psychiatric Institute, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Ardesheer Talati
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Division of Translational Epidemiology, New York State Psychiatric Institute, New York, NY, USA
| | - Mark S Ansorge
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA.
| | - Jay A Gingrich
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
4
|
Choi JM, Lee EJ. Association Among Depressive Symptoms, Physical Symptoms, Serotonin Levels, and Gambling Disorder: A Case-Control Study. J Psychosoc Nurs Ment Health Serv 2025:1-10. [PMID: 40258212 DOI: 10.3928/02793695-20250415-03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
PURPOSE The current study compares depressive symptoms, physical symptoms, and serotonin levels between individuals with gambling disorder (GD) and healthy controls (HC), investigates their relationship, and identifies factors that predict the development of GD. METHOD A case-control study with purposive convenience sampling was conducted using data from GD (n = 76) and HC (n = 56) groups. RESULTS Increased depressive symptoms were associated with greater physical symptoms (r = 0.70, p < 0.001) and problem gambling severity (r = 0.37, p = 0.001). Decreased serotonin levels suggest a potential depressive disorder; however, not all individuals had lower serotonin levels, indicating emotionally vulnerable subgroups. Increased serotonin levels correlated with restlessness, which was associated with problem gambling (r = 0.31, p = 0.006). Decreased problem gambling severity was linked to increased alcohol use (r = -0.29, p = 0.012). CONCLUSION Results indicate a correlation among depressive symptoms, physical symptoms, and serotonin imbalance in individuals with GD. [Journal of Psychosocial Nursing and Mental Health Services, xx(x), xx-xx.].
Collapse
|
5
|
Talati A, Vande Voort JL, White LJ, Hodge D, Stoppel CJ, Weissman MM, Gingrich JA, Bobo WV. Prenatal Antidepressant Exposure and Risk of Depression and Anxiety Disorders: An Electronic Health Records-Based Cohort Study. J Am Acad Child Adolesc Psychiatry 2025:S0890-8567(25)00179-0. [PMID: 40204059 DOI: 10.1016/j.jaac.2025.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 03/04/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
OBJECTIVE To examine the associations of serotonergic antidepressant exposure during pregnancy with the risk of depression and anxiety disorders in offspring. METHOD The Mayo Clinic Rochester Epidemiology Project medical records-linkage system was used to study offspring born to mothers who were prescribed a selective serotonin reuptake inhibitor or serotonin-norepinephrine reuptake inhibitor (S/NRI users, n = 837) during pregnancy (1997-2010). Cox regression models were used to calculate hazard ratios to examine associations of S/NRIs with diagnosed depression and anxiety, defined based on a review of medical records by 2 board-certified psychiatrists, vs no maternal antidepressant use during pregnancy (nonusers, n = 863) and maternal antidepressant use in the year prior to pregnancy (former users, n = 399) as control groups. RESULTS After all adjustments for covariates, children born to S/NRI users during pregnancy did not differ in onset of depression or anxiety from the children of nonusers (adjusted hazard ratio = 1.00, 95% CI [0.74, 1.85]) or former users (adjusted hazard ratio = 0.94, 95% CI [0.69, 1.27]). These associations were similar when exposure was limited only to selective serotonin reuptake inhibitors. CONCLUSION The results suggest that higher rates of childhood and adolescent depression or anxiety conditioned on maternal S/NRI use in pregnancy are more likely to be driven by maternal depression or underlying propensity for depression rather than direct pharmacological effects of in utero S/NRI exposure.
Collapse
Affiliation(s)
- Ardesheer Talati
- Columbia University Irving Medical Center/Vagelos College of Physicians & Surgeons, New York, New York; New York State Psychiatric Institute, New York, New York.
| | | | | | | | | | - Myrna M Weissman
- Columbia University Irving Medical Center/Vagelos College of Physicians & Surgeons, New York, New York; New York State Psychiatric Institute, New York, New York
| | - Jay A Gingrich
- Columbia University Irving Medical Center/Vagelos College of Physicians & Surgeons, New York, New York; New York State Psychiatric Institute, New York, New York
| | | |
Collapse
|
6
|
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
|
7
|
De Stasi AM, Zorrilla de San Martin J, Soto N, Aguirre A, Olusakin J, Lourenço J, Gaspar P, Bacci A. Alterations of Adult Prefrontal Circuits Induced by Early Postnatal Fluoxetine Treatment Mediated by 5-HT7 Receptors. J Neurosci 2025; 45:e2393232024. [PMID: 39909574 PMCID: PMC11800747 DOI: 10.1523/jneurosci.2393-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 02/07/2025] Open
Abstract
The prefrontal cortex (PFC) plays a key role in high-level cognitive functions and emotional behaviors, and PFC alterations correlate with different brain disorders including major depression and anxiety. In mice, the first two postnatal weeks represent a critical period of high sensitivity to environmental changes. In this temporal window, serotonin (5-HT) levels regulate the wiring of PFC cortical neurons. Early-life insults and postnatal exposure to the selective serotonin reuptake inhibitor fluoxetine (FLX) affect PFC development leading to depressive and anxiety-like phenotypes in adult mice. However, the mechanisms responsible for these dysfunctions remain obscure. We found that early postnatal FLX exposure (PNFLX) results in reduced overall firing and high-frequency bursting of putative pyramidal neurons (PNs) of deep layers of the medial PFC of adult mice of both sexes in vivo. Ex vivo, patch-clamp recordings revealed that PNFLX abolished high-frequency firing in a distinct subpopulation of deep-layer mPFC PNs, which transiently express the serotonin transporter SERT during the first 2 postnatal weeks. SERT+ and SERT- PNs exhibit distinct morphofunctional properties. Genetic deletion of 5-HT7Rs and pharmacological 5-HT7R blockade partially rescued both the PNFLX-induced reduction of PN firing in vivo and the altered firing of SERT+ PNs in vitro. This indicates a pivotal role of this 5-HTR subtype in mediating 5-HT-dependent maturation of PFC circuits that are susceptible to early-life insults. Overall, our results suggest potential novel neurobiological mechanisms, underlying detrimental neurodevelopmental consequences induced by early-life alterations of 5-HT levels.
Collapse
Affiliation(s)
| | | | - Nina Soto
- Sorbonne Université, ICM-Paris Brain Institute, CNRS, INSERM, Paris 75013, France
| | - Andrea Aguirre
- Sorbonne Université, ICM-Paris Brain Institute, CNRS, INSERM, Paris 75013, France
| | - Jimmy Olusakin
- INSERM UMRS-839 Institut du Fer à Moulin, Paris 75005, France
| | - Joana Lourenço
- Sorbonne Université, ICM-Paris Brain Institute, CNRS, INSERM, Paris 75013, France
| | - Patricia Gaspar
- Sorbonne Université, ICM-Paris Brain Institute, CNRS, INSERM, Paris 75013, France
| | - Alberto Bacci
- Sorbonne Université, ICM-Paris Brain Institute, CNRS, INSERM, Paris 75013, France
| |
Collapse
|
8
|
Lee MJ, Chen YL, Wu SI, Huang CW, Dewey ME, Chen VCH. Association between maternal antidepressant use during pregnancy and the risk of autism spectrum disorder and attention deficit hyperactivity disorder in offspring. Eur Child Adolesc Psychiatry 2024; 33:4273-4283. [PMID: 38762849 DOI: 10.1007/s00787-024-02460-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/29/2024] [Indexed: 05/20/2024]
Abstract
Prenatal antidepressant exposure has been reported to be associated with adverse neurodevelopmental outcomes, yet studies considering confounding factors in Asian populations are lacking. This study utilized a nationwide data base in Taiwan, enrolling all liveborn children registered in the National Health Insurance system between 2004 and 2016. Subjects were divided into two groups: antidepressant-exposed (n = 55,707)) and antidepressant-unexposed group (n = 2,245,689). The effect of antidepressant exposure during different trimesters on autism spectrum disorder (ASD) and attention-deficit/hyperactivity disorder (ADHD) was examined. Sibling controls and parallel comparisons by paternal exposure status were treated as negative controls. Additional sensitivity analyses were conducted to examine the effects of antidepressant exposure before and after pregnancy. Prenatal antidepressant exposure was associated with increased risks of ASD and ADHD in population-wide and adjusted analysis. However when comparing antidepressant-exposed children with their unexposed siblings, no differences were found for ASD (Hazard ratio [HR]: 1.04, 95% confidence interval [CI] 0.76-1.42 in first trimester; HR: 0.96, 95% CI 0.62-1.50 in second trimester; HR: 0.69, 95% CI 0.32-1.48 in third trimester) and ADHD (HR: 0.98, 95%CI 0.84-1.15 in first trimester; HR: 0.91, 95% CI 0.73-1.14 in second trimester; HR: 0.79, 95% CI 0.54-1.16 in third trimester). Increased risks for ASD and ADHD were also noted in paternal control, before and after pregnancy analyses. These results imply that the association between prenatal antidepressant exposure and ASD and ADHD is not contributed to by an intrauterine medication effect but more likely to be accounted for by maternal depression, genetic, and potential environmental factors.
Collapse
Affiliation(s)
- Min-Jing Lee
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Psychiatry, Chiayi Chang Gung Memorial Hospital, Chiayi County, Puzi City, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Lung Chen
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung, Taiwan
- Department of Psychology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Shu-I Wu
- Department of Psychiatry, Mackay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Chien-Wei Huang
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Michael E Dewey
- Health Service and Population Research Department, King's College London, London, UK
| | - Vincent Chin-Hung Chen
- Department of Psychiatry, Chiayi Chang Gung Memorial Hospital, Chiayi County, Puzi City, Taiwan.
- School of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
9
|
Agrawal P, Singh P, Singh KP. Vilazodone exposure during pregnancy: Effects on embryo-fetal development, pregnancy outcomes and fetal neurotoxicity by BDNF/Bax-Bcl2/5-HT mediated mechanisms. Neurotoxicology 2024; 105:280-292. [PMID: 39532268 DOI: 10.1016/j.neuro.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/16/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
The high prevalence of major depressive disorder (MDD) among women of childbearing age necessitates careful consideration of antidepressant use during pregnancy. Although newer antidepressants, such as Vilazodone (VLZ), are preferred for their enhanced therapeutic profiles; however, their safety during pregnancy and long-term effects on offspring brains remain inadequately addressed. Therefore, this study aimed to investigate the reproductive and developmental neurotoxicity of VLZ given at equivalent therapeutic doses during gestation in a rat model. Pregnant Wistar dams were orally administered either with 1 mg/day or 2 mg/day of VLZ from gestation day (GD) 6-21. The dams were sacrificed at GD 21, and the placentas and fetuses were collected. Fetal brains were then subjected to neurohistopathological, neurochemical, and biochemical analysis. Prenatal exposure to VLZ at 2 mg/day resulted in significant maternal, reproductive, and embryo-fetal toxicity, characterized by reduced food intake, diminished weight gain in pregnant dams, and smaller litter sizes, along with decreased fetal and placental weights. These effects were associated with developmental neurotoxicity, which manifested as decreased fetal brain size and weight, a substantial reduction in neocortical layer thickness, brain-derived neurotrophic factor (BDNF) expression, serotonin, dopamine, and norepinephrine neurotransmitter levels (5-HT, DA, and NE), and increased apoptotic activity (Bax and Bcl-2 ratio) and acetylcholinesterase levels in the developing brain. Our findings indicate that prenatal VLZ exposure interfere with crucial brain development processes involving the BDNF/Bax-Bcl2/5-HT signalling pathways, leading to long-lasting neurodevelopmental impairments. This study is the first to document the adverse effects of VLZ on fetal brain development, highlighting the need for further research to assess the safety of VLZ use during pregnancy.
Collapse
Affiliation(s)
- Priyanka Agrawal
- Neurobiology Lab., Department of Zoology, University of Allahabad, Prayagraj, UP 211002, India.
| | - Pallavi Singh
- Neurobiology Lab., Department of Zoology, University of Allahabad, Prayagraj, UP 211002, India.
| | - K P Singh
- Neurobiology Lab., Department of Zoology, University of Allahabad, Prayagraj, UP 211002, India.
| |
Collapse
|
10
|
Orav E, Kokinovic B, Teppola H, Siimon M, Lauri SE, Hartung H. Arginine vasopressin activates serotonergic neurons in the dorsal raphe nucleus during neonatal development in vitro and in vivo. Neuropharmacology 2024; 258:110068. [PMID: 38996832 DOI: 10.1016/j.neuropharm.2024.110068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024]
Abstract
Birth stress is a risk factor for psychiatric disorders and associated with exaggerated release of the stress hormone arginine vasopressin (AVP) into circulation and in the brain. In perinatal hippocampus, AVP activates GABAergic interneurons which leads to suppression of spontaneous network events and suggests a protective function of AVP on cortical networks during birth. However, the role of AVP in developing subcortical networks is not known. Here we tested the effect of AVP on the dorsal raphe nucleus (DRN) 5-hydroxytryptamine (5-HT, serotonin) system in male and female neonatal rats, since early 5-HT homeostasis is critical for the development of cortical brain regions and emotional behaviors. We show that AVP is strongly excitatory in neonatal DRN: it increases excitatory synaptic inputs of 5-HT neurons via V1A receptors in vitro and promotes their action potential firing through a combination of its effect on glutamatergic synaptic transmission and a direct effect on the excitability of these neurons. Furthermore, we identified two major firing patterns of neonatal 5-HT neurons in vivo, tonic regular firing and low frequency oscillations of regular spike trains and confirmed that these neurons are also activated by AVP in vivo. Finally, we show that the sparse vasopressinergic innervation in neonatal DRN originates exclusively from cell groups in medial amygdala and bed nucleus of stria terminalis. Hyperactivation of the neonatal 5-HT system by AVP during birth stress may impact its own functional development and affect the maturation of cortical target regions, which may increase the risk for psychiatric conditions later on.
Collapse
Affiliation(s)
- Ester Orav
- HiLIFE Neuroscience Center, University of Helsinki, Helsinki, Finland.
| | - Bojana Kokinovic
- HiLIFE Neuroscience Center, University of Helsinki, Helsinki, Finland.
| | - Heidi Teppola
- HiLIFE Neuroscience Center, University of Helsinki, Helsinki, Finland.
| | - Mari Siimon
- HiLIFE Neuroscience Center, University of Helsinki, Helsinki, Finland.
| | - Sari E Lauri
- HiLIFE Neuroscience Center, University of Helsinki, Helsinki, Finland.
| | - Henrike Hartung
- HiLIFE Neuroscience Center, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
11
|
Singh P, Agrawal P, Singh KP. Prenatal exposure to vortioxetine and vilazodone: Impact on depressive- and anxiety-like behavioral manifestations in young rat offspring. Behav Brain Res 2024; 471:115128. [PMID: 38945303 DOI: 10.1016/j.bbr.2024.115128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/14/2024] [Accepted: 06/24/2024] [Indexed: 07/02/2024]
Abstract
Major depressive disorder (MDD) affects millions of people worldwide, with women at a higher risk during the childbearing age. Vortioxetine (VOX) and Vilazodone (VLZ) are newer antidepressants with improved therapeutic profile commonly used, but their safety during pregnancy and long-term effects on offspring are poorly understood due to paucity of literature in preclinical and clinical studies. This study aimed to investigate whether prenatal exposure to VOX and VLZ impacts depressive- and anxiety-like neurobehavioral alterations in offspring, focusing on neurotransmitter-mediated mechanisms. Pregnant Wistar dams received either VOX or VLZ, 1 mg/day and 2 mg/day of the drug orally from gestation day (GD) 6-21. The dams naturally delivered their offspring and reared until they reached postnatal day (PND) 21. Offspring of both sexes were tested for display of depressive-and anxiety-like behaviors from PND 56-70. After PND 70, offspring were sacrificed, and their brains were collected to estimate neurotransmitter levels. As per protocol, controls were maintained simultaneously for each experimental design. Prenatal exposure to VOX or VLZ induced an increased state of depressive- and anxiety-like behaviors in both male and female offspring. Additionally, neurotransmitter (serotonin, dopamine, and nor-epinephrine) levels in the prefrontal cortex region of the brain were substantially reduced in exposed offspring. No sex specific neurobehavioral and neurochemical implications were observed in the present study. Our findings suggest that prenatal exposure to VOX and VLZ disrupts neurochemical balance in the fetal brain, leading to long-lasting neurobehavioral impairments in offspring of both sexes.
Collapse
Affiliation(s)
- Pallavi Singh
- Neurobiology Lab., Department of Zoology, University of Allahabad, Prayagraj 211002, India.
| | - Priyanka Agrawal
- Neurobiology Lab., Department of Zoology, University of Allahabad, Prayagraj 211002, India.
| | - K P Singh
- Neurobiology Lab., Department of Zoology, University of Allahabad, Prayagraj 211002, India.
| |
Collapse
|
12
|
Soto NN, Gaspar P, Bacci A. Not Just a Mood Disorder─Is Depression a Neurodevelopmental, Cognitive Disorder? Focus on Prefronto-Thalamic Circuits. ACS Chem Neurosci 2024; 15:1611-1618. [PMID: 38580316 PMCID: PMC11027097 DOI: 10.1021/acschemneuro.3c00828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 04/07/2024] Open
Abstract
Depression is one of the most burdensome psychiatric disorders, affecting hundreds of millions of people worldwide. The disease is characterized not only by severe emotional and affective impairments, but also by disturbed vegetative and cognitive functions. Although many candidate mechanisms have been proposed to cause the disease, the pathophysiology of cognitive impairments in depression remains unclear. In this article, we aim to assess the link between cognitive alterations in depression and possible developmental changes in neuronal circuit wiring during critical periods of susceptibility. We review the existing literature and propose a role of serotonin signaling during development in shaping the functional states of prefrontal neuronal circuits and prefronto-thalamic loops. We discuss how early life insults affecting the serotonergic system could be important in the alterations of these local and long-range circuits, thus favoring the emergence of neurodevelopmental disorders, such as depression.
Collapse
Affiliation(s)
- Nina Nitzan Soto
- ICM−Paris
Brain Institute, CNRS, INSERM, Sorbonne
Université, 47 Boulevard de l’Hopital, 75013 Paris, France
| | - Patricia Gaspar
- ICM−Paris
Brain Institute, CNRS, INSERM, Sorbonne
Université, 47 Boulevard de l’Hopital, 75013 Paris, France
| | - Alberto Bacci
- ICM−Paris
Brain Institute, CNRS, INSERM, Sorbonne
Université, 47 Boulevard de l’Hopital, 75013 Paris, France
| |
Collapse
|
13
|
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
|
14
|
Kisner A, Polter AM. Maturation of glutamatergic transmission onto dorsal raphe serotonergic neurons. J Neurophysiol 2024; 131:626-637. [PMID: 38380827 PMCID: PMC11305679 DOI: 10.1152/jn.00037.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/01/2024] [Accepted: 02/19/2024] [Indexed: 02/22/2024] Open
Abstract
Serotonergic neurons in the dorsal raphe nucleus (DRN) play important roles early in postnatal development in the maturation and modulation of higher-order emotional, sensory, and cognitive circuitry. The pivotal functions of these cells in brain development make them a critical substrate by which early experience can be wired into the brain. In this study, we investigated the maturation of synapses onto dorsal raphe serotonergic neurons in typically developing male and female mice using whole cell patch-clamp recordings in ex vivo brain slices. We show that while inhibition of these neurons is relatively stable across development, glutamatergic synapses greatly increase in strength between postnatal day 6 (P6) and P21-23. In contrast to forebrain regions, where the components making up glutamatergic synapses are dynamic across early life, we find that DRN excitatory synapses maintain a very high ratio of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) to N-methyl-d-aspartate (NMDA) receptors and a rectifying component of the AMPA response until adulthood. Overall, these findings reveal that the development of serotonergic neurons is marked by a significant refinement of glutamatergic synapses during the first three postnatal weeks. This suggests this time is a sensitive period of heightened plasticity for the integration of information from upstream brain areas. Genetic and environmental insults during this period could lead to alterations in serotonergic output, impacting both the development of forebrain circuits and lifelong neuromodulatory actions.NEW & NOTEWORTHY Serotonergic neurons are regulators of both the development of and ongoing activity in neuronal circuits controlling affective, cognitive, and sensory processing. Here, we characterize the maturation of extrinsic synaptic inputs onto these cells, showing that the first three postnatal weeks are a period of synaptic refinement and a potential window for experience-dependent plasticity in response to both enrichment and adversity.
Collapse
Affiliation(s)
- Alexandre Kisner
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Abigail M Polter
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| |
Collapse
|
15
|
Ogelman R, Gomez Wulschner LE, Hoelscher VM, Hwang IW, Chang VN, Oh WC. Serotonin modulates excitatory synapse maturation in the developing prefrontal cortex. Nat Commun 2024; 15:1368. [PMID: 38365905 PMCID: PMC10873381 DOI: 10.1038/s41467-024-45734-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 02/02/2024] [Indexed: 02/18/2024] Open
Abstract
Serotonin (5-HT) imbalances in the developing prefrontal cortex (PFC) are linked to long-term behavioral deficits. However, the synaptic mechanisms underlying 5-HT-mediated PFC development are unknown. We found that chemogenetic suppression and enhancement of 5-HT release in the PFC during the first two postnatal weeks decreased and increased the density and strength of excitatory spine synapses, respectively, on prefrontal layer 2/3 pyramidal neurons in mice. 5-HT release on single spines induced structural and functional long-term potentiation (LTP), requiring both 5-HT2A and 5-HT7 receptor signals, in a glutamatergic activity-independent manner. Notably, LTP-inducing 5-HT stimuli increased the long-term survival of newly formed spines ( ≥ 6 h) via 5-HT7 Gαs activation. Chronic treatment of mice with fluoxetine, a selective serotonin-reuptake inhibitor, during the first two weeks, but not the third week of postnatal development, increased the density and strength of excitatory synapses. The effect of fluoxetine on PFC synaptic alterations in vivo was abolished by 5-HT2A and 5-HT7 receptor antagonists. Our data describe a molecular basis of 5-HT-dependent excitatory synaptic plasticity at the level of single spines in the PFC during early postnatal development.
Collapse
Affiliation(s)
- Roberto Ogelman
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Luis E Gomez Wulschner
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Victoria M Hoelscher
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - In-Wook Hwang
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Victoria N Chang
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Won Chan Oh
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
16
|
Jaber M, Kahwaji H, Nasr S, Baz R, Kim YK, Fakhoury M. Precision Medicine in Depression: The Role of Proteomics and Metabolomics in Personalized Treatment Approaches. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1456:359-378. [PMID: 39261438 DOI: 10.1007/978-981-97-4402-2_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Depression, or major depressive disorder (MDD), is a widespread mental health condition marked by enduring feelings of sorrow and loss of interest. Treatment of depression frequently combines psychotherapy, medication, and lifestyle modifications. However, the occurrence of treatment resistance in certain individuals makes it difficult for physicians to effectively manage this disorder, calling for the implementation of alternative therapeutic strategies. Recently, precision medicine has gained increased attention in the field of mental health, paving the way for more personalized and effective therapeutic interventions in depression. Also known as personalized medicine, this approach relies on genetic composition, molecular profiles, and environmental variables to customize therapies to individual patients. In particular, precision medicine has offered novel viewpoints on depression through two specific domains: proteomics and metabolomics. On one hand, proteomics is the thorough study of proteins in a biological system, while metabolomics focuses on analyzing the complete set of metabolites in a living being. In the past few years, progress in research has led to the identification of numerous depression-related biomarkers using proteomics and metabolomics techniques, allowing for early identification, precise diagnosis, and improved clinical outcome. However, despite significant progress in these techniques, further efforts are required for advancing precision medicine in the diagnosis and treatment of depression. The overarching goal of this chapter is to provide the current state of knowledge regarding the use of proteomics and metabolomics in identifying biomarkers related to depression. It also highlights the potential of proteomics and metabolomics in elucidating the intricate processes underlying depression, opening the door for tailored therapies that could eventually enhance clinical outcome in depressed patients. This chapter finally discusses the main challenges in the use of proteomics and metabolomics and discusses potential future research directions.
Collapse
Affiliation(s)
- Mohamad Jaber
- School of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hamza Kahwaji
- School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Sirine Nasr
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Reine Baz
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Yong-Ku Kim
- Department of Psychiatry, Korea University College of Medicine, Seoul, Republic of Korea
| | - Marc Fakhoury
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon.
| |
Collapse
|
17
|
Sharp T, Collins H. Mechanisms of SSRI Therapy and Discontinuation. Curr Top Behav Neurosci 2024; 66:21-47. [PMID: 37955823 DOI: 10.1007/7854_2023_452] [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] [Indexed: 11/14/2023]
Abstract
SSRIs are one of the most widely used drug therapies in primary care and psychiatry, and central to the management of the most common mental health problems in today's society. Despite this, SSRIs suffer from a slow onset of therapeutic effect and relatively poor efficacy as well as adverse effects, with recent concerns being focused on a disabling SSRI discontinuation syndrome. The mechanism underpinning their therapeutic effect has long shifted away from thinking that SSRIs act simply by increasing 5-HT in the synapse. Rather, a current popular view is that increased 5-HT is just the beginning of a series of complex downstream signalling events, which trigger changes in neural plasticity at the functional and structural level. These changes in plasticity are then thought to interact with neuropsychological processes to enhance re-learning of emotional experiences that ultimately brings about changes in mood. This compelling view of SSRI action is underpinning attempts to understand fast-acting antidepressants, such as ketamine and psychedelic drugs, and aid the development of future therapies. An important gap in the theory is evidence that changes in plasticity are causally linked to relevant behavioural effects. Also, predictions that the SSRI-induced neural plasticity might have applicability in other areas of medicine have not yet been borne out. In contrast to the sophisticated view of the antidepressant action of SSRIs, the mechanism underpinning SSRI discontinuation is little explored. Nevertheless, evidence of rebound increases in 5-HT neuron excitability immediately on cessation of SSRI treatment provide a starting point for future investigation. Indeed, this evidence allows formulation of a mechanistic explanation of SSRI discontinuation which draws on parallels with the withdrawal states of other psychotropic drugs.
Collapse
Affiliation(s)
- Trevor Sharp
- Department of Pharmacology, University of Oxford, Oxford, UK.
| | - Helen Collins
- Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
18
|
Sun ZW, Wang X, Zhao Y, Sun ZX, Wu YH, Hu H, Zhang L, Wang SD, Li F, Wei AJ, Feng H, Xie F, Qian LJ. Blood-brain barrier dysfunction mediated by the EZH2-Claudin-5 axis drives stress-induced TNF-α infiltration and depression-like behaviors. Brain Behav Immun 2024; 115:143-156. [PMID: 37848095 DOI: 10.1016/j.bbi.2023.10.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/19/2023] [Accepted: 10/14/2023] [Indexed: 10/19/2023] Open
Abstract
Growing evidence suggests that neurovascular dysfunction characterized by blood-brain barrier (BBB) breakdown underlies the development of psychiatric disorders, such as major depressive disorder (MDD). Tight junction (TJ) proteins are critical modulators of homeostasis and BBB integrity. TJ protein Claudin-5 is the most dominant BBB component and is downregulated in numerous depression models; however, the underlying mechanisms remain elusive. Here, we demonstrate a molecular basis of BBB breakdown that links stress and depression. We implemented an animal model of depression, chronic unpredictable mild stress (CUMS) in male C57BL/6 mice, and showed that hippocampal BBB breakdown was closely associated with stress vulnerability. Concomitantly, we found that dysregulated Cldn5 level coupled with repression of the histone methylation signature at its promoter contributed to stress-induced BBB dysfunction and depression. Moreover, histone methyltransferase enhancer of zeste homolog 2 (EZH2) knockdown improved Cldn5 expression and alleviated depression-like behaviors by suppressing the tri-methylation of lysine 27 on histone 3 (H3K27me3) in chronically stressed mice. Furthermore, the stress-induced excessive transfer of peripheral cytokine tumor necrosis factor-α (TNF-α) into the hippocampus was prevented by Claudin-5 overexpression and EZH2 knockdown. Interestingly, antidepressant treatment could inhibit H3K27me3 deposition at the Cldn5 promoter, reversing the loss of the encoded protein and BBB damage. Considered together, these findings reveal the importance of the hippocampal EZH2-Claudin-5 axis in regulating neurovascular function and MDD development, providing potential therapeutic targets for this psychiatric illness.
Collapse
Affiliation(s)
- Zhao-Wei Sun
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Xue Wang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Yun Zhao
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Zhao-Xin Sun
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin 301617, China
| | - Yu-Han Wu
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Hui Hu
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Ling Zhang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Shi-Da Wang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Feng Li
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Ai-Jun Wei
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Hong Feng
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin 301617, China
| | - Fang Xie
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China.
| | - Ling-Jia Qian
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China.
| |
Collapse
|
19
|
Carlos-Lima E, Higa GSV, Viana FJC, Tamais AM, Cruvinel E, Borges FDS, Francis-Oliveira J, Ulrich H, De Pasquale R. Serotonergic Modulation of the Excitation/Inhibition Balance in the Visual Cortex. Int J Mol Sci 2023; 25:519. [PMID: 38203689 PMCID: PMC10778629 DOI: 10.3390/ijms25010519] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/18/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Serotonergic neurons constitute one of the main systems of neuromodulators, whose diffuse projections regulate the functions of the cerebral cortex. Serotonin (5-HT) is known to play a crucial role in the differential modulation of cortical activity related to behavioral contexts. Some features of the 5-HT signaling organization suggest its possible participation as a modulator of activity-dependent synaptic changes during the critical period of the primary visual cortex (V1). Cells of the serotonergic system are among the first neurons to differentiate and operate. During postnatal development, ramifications from raphe nuclei become massively distributed in the visual cortical area, remarkably increasing the availability of 5-HT for the regulation of excitatory and inhibitory synaptic activity. A substantial amount of evidence has demonstrated that synaptic plasticity at pyramidal neurons of the superficial layers of V1 critically depends on a fine regulation of the balance between excitation and inhibition (E/I). 5-HT could therefore play an important role in controlling this balance, providing the appropriate excitability conditions that favor synaptic modifications. In order to explore this possibility, the present work used in vitro intracellular electrophysiological recording techniques to study the effects of 5-HT on the E/I balance of V1 layer 2/3 neurons, during the critical period. Serotonergic action on the E/I balance has been analyzed on spontaneous activity, evoked synaptic responses, and long-term depression (LTD). Our results pointed out that the predominant action of 5-HT implies a reduction in the E/I balance. 5-HT promoted LTD at excitatory synapses while blocking it at inhibitory synaptic sites, thus shifting the Hebbian alterations of synaptic strength towards lower levels of E/I balance.
Collapse
Affiliation(s)
- Estevão Carlos-Lima
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (E.C.-L.); (G.S.V.H.); (E.C.); (J.F.-O.)
| | - Guilherme Shigueto Vilar Higa
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (E.C.-L.); (G.S.V.H.); (E.C.); (J.F.-O.)
- Departamento de Bioquímica, Instituto de Química (USP), São Paulo 05508-900, SP, Brazil;
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo 09210-580, SP, Brazil
| | - Felipe José Costa Viana
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (E.C.-L.); (G.S.V.H.); (E.C.); (J.F.-O.)
| | - Alicia Moraes Tamais
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (E.C.-L.); (G.S.V.H.); (E.C.); (J.F.-O.)
| | - Emily Cruvinel
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (E.C.-L.); (G.S.V.H.); (E.C.); (J.F.-O.)
| | - Fernando da Silva Borges
- Department of Physiology & Pharmacology, SUNY Downstate Health Sciences University, New York, NY 11203, USA;
| | - José Francis-Oliveira
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (E.C.-L.); (G.S.V.H.); (E.C.); (J.F.-O.)
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química (USP), São Paulo 05508-900, SP, Brazil;
| | - Roberto De Pasquale
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (E.C.-L.); (G.S.V.H.); (E.C.); (J.F.-O.)
| |
Collapse
|
20
|
Gianni G, Pasqualetti M. Wiring and Volume Transmission: An Overview of the Dual Modality for Serotonin Neurotransmission. ACS Chem Neurosci 2023; 14:4093-4104. [PMID: 37966717 DOI: 10.1021/acschemneuro.3c00648] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023] Open
Abstract
Serotonin is a neurotransmitter involved in the modulation of a multitude of physiological and behavioral processes. In spite of the relatively reduced number of serotonin-producing neurons present in the mammalian CNS, a complex long-range projection system provides profuse innervation to the whole brain. Heterogeneity of serotonin receptors, grouped in seven families, and their spatiotemporal expression pattern account for its widespread impact. Although neuronal communication occurs primarily at tiny gaps called synapses, wiring transmission, another mechanism based on extrasynaptic diffusion of neuroactive molecules and referred to as volume transmission, has been described. While wiring transmission is a rapid and specific one-to-one modality of communication, volume transmission is a broader and slower mode in which a single element can simultaneously act on several different targets in a one-to-many mode. Some experimental evidence regarding ultrastructural features, extrasynaptic localization of receptors and transporters, and serotonin-glia interactions collected over the past four decades supports the existence of a serotonergic system of a dual modality of neurotransmission, in which wiring and volume transmission coexist. To date, in spite of the radical difference in the two modalities, limited information is available on the way they are coordinated to mediate the specific activities in which serotonin participates. Understanding how wiring and volume transmission modalities contribute to serotonergic neurotransmission is of utmost relevance for the comprehension of serotonin functions in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Giulia Gianni
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56127 Pisa, Italy
| | - Massimo Pasqualetti
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56127 Pisa, Italy
- Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy
- Centro per l'Integrazione della Strumentazione Scientifica dell'Università di Pisa (CISUP), 56126 Pisa, Italy
| |
Collapse
|
21
|
Teng L, Qin Q, Zhou Z, Zhou F, Cao C, Yang J, Ding J. Glutamate secretion by embryonic stem cells as an autocrine signal to promote proliferation. Sci Rep 2023; 13:19069. [PMID: 37925518 PMCID: PMC10625544 DOI: 10.1038/s41598-023-46477-2] [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: 07/05/2023] [Accepted: 11/01/2023] [Indexed: 11/06/2023] Open
Abstract
Glutamate, the major excitatory neurotransmitter in the central nervous system, has also been found to play a role in embryonic stem (ES) cells. However, the exact mechanism and function of glutamatergic signaling in ES cells remain poorly understood. In this study, we identified a glutamatergic transmission circuit in ES cells that operates through an autocrine mechanism and regulates cell proliferation. We performed biological analyses to identify the key components involved in glutamate biosynthesis, packaging for secretion, reaction, and reuptake in ES cells, including glutaminase, vesicular glutamate transporter, glutamate N-methyl-D-aspartate (NMDA) receptor, and cell membrane excitatory amino-acid transporter (EAAT). We directly quantified the released glutamate signal using microdialysis-high performance liquid chromatography-tandem mass spectrometry (MD-HPLC-MS-MS). Pharmacological inhibition of endogenous glutamate release and the resulting tonic activation of NMDA receptors significantly affected ES cell proliferation, suggesting that ES cells establish a glutamatergic autocrine niche via releasing and responding to the transmitter for their own regulation.
Collapse
Affiliation(s)
- Lin Teng
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
- College of Basic Medical Sciences, Hubei Key Laboratory of Tumor Microencironment and Immunotherapy, China Three Gorges University, Yichang, 443000, Hubei, China
| | - Qin Qin
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
| | - Ziyi Zhou
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
| | - Fei Zhou
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
| | - Chunyu Cao
- College of Basic Medical Sciences, Hubei Key Laboratory of Tumor Microencironment and Immunotherapy, China Three Gorges University, Yichang, 443000, Hubei, China
| | - Jian Yang
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
| | - Jiawang Ding
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China.
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China.
| |
Collapse
|
22
|
Heesbeen EJ, Bijlsma EY, Verdouw PM, van Lissa C, Hooijmans C, Groenink L. The effect of SSRIs on fear learning: a systematic review and meta-analysis. Psychopharmacology (Berl) 2023; 240:2335-2359. [PMID: 36847831 PMCID: PMC10593621 DOI: 10.1007/s00213-023-06333-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 01/31/2023] [Indexed: 03/01/2023]
Abstract
RATIONALE Selective serotonin reuptake inhibitors (SSRIs) are considered first-line medication for anxiety-like disorders such as panic disorder, generalized anxiety disorder, and post-traumatic stress disorder. Fear learning plays an important role in the development and treatment of these disorders. Yet, the effect of SSRIs on fear learning are not well known. OBJECTIVE We aimed to systematically review the effect of six clinically effective SSRIs on acquisition, expression, and extinction of cued and contextual conditioned fear. METHODS We searched the Medline and Embase databases, which yielded 128 articles that met the inclusion criteria and reported on 9 human and 275 animal experiments. RESULTS Meta-analysis showed that SSRIs significantly reduced contextual fear expression and facilitated extinction learning to cue. Bayesian-regularized meta-regression further suggested that chronic treatment exerts a stronger anxiolytic effect on cued fear expression than acute treatment. Type of SSRI, species, disease-induction model, and type of anxiety test used did not seem to moderate the effect of SSRIs. The number of studies was relatively small, the level of heterogeneity was high, and publication bias has likely occurred which may have resulted in an overestimation of the overall effect sizes. CONCLUSIONS This review suggests that the efficacy of SSRIs may be related to their effects on contextual fear expression and extinction to cue, rather than fear acquisition. However, these effects of SSRIs may be due to a more general inhibition of fear-related emotions. Therefore, additional meta-analyses on the effects of SSRIs on unconditioned fear responses may provide further insight into the actions of SSRIs.
Collapse
Affiliation(s)
- Elise J Heesbeen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Elisabeth Y Bijlsma
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - P Monika Verdouw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Caspar van Lissa
- Department of Methodology, Tilburg University, Tilburg, Netherlands
| | - Carlijn Hooijmans
- Department of Anaesthesiology, Pain and Palliative Care, Radboud University Medical Center, Nijmegen, Netherlands
| | - Lucianne Groenink
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.
| |
Collapse
|
23
|
Heuvelman H, Davies NM, Ben-Shlomo Y, Emond A, Evans J, Gunnell D, Liebling R, Morris R, Payne R, Storey C, Viner M, Rai D. Antidepressants in pregnancy: applying causal epidemiological methods to understand service-use outcomes in women and long-term neurodevelopmental outcomes in exposed children. Health Technol Assess 2023; 27:1-83. [PMID: 37842916 DOI: 10.3310/aqtf4490] [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] [Indexed: 10/17/2023] Open
Abstract
Background Antidepressants are commonly prescribed during pregnancy, despite a lack of evidence from randomised trials on the benefits or risks. Some studies have reported associations of antidepressants during pregnancy with adverse offspring neurodevelopment, but whether or not such associations are causal is unclear. Objectives To study the associations of antidepressants for depression in pregnancy with outcomes using multiple methods to strengthen causal inference. Design This was an observational cohort design using multiple methods to strengthen causal inference, including multivariable regression, propensity score matching, instrumental variable analysis, negative control exposures, comparison across indications and exposure discordant pregnancies analysis. Setting This took place in UK general practice. Participants Participants were pregnant women with depression. Interventions The interventions were initiation of antidepressants in pregnancy compared with no initiation, and continuation of antidepressants in pregnancy compared with discontinuation. Main outcome measures The maternal outcome measures were the use of primary care and secondary mental health services during pregnancy, and during four 6-month follow-up periods up to 24 months after pregnancy, and antidepressant prescription status 24 months following pregnancy. The child outcome measures were diagnosis of autism, diagnosis of attention deficit hyperactivity disorder and intellectual disability. Data sources UK Clinical Practice Research Datalink. Results Data on 80,103 pregnancies were used to study maternal primary care outcomes and were linked to 34,274 children with at least 4-year follow-up for neurodevelopmental outcomes. Women who initiated or continued antidepressants during pregnancy were more likely to have contact with primary and secondary health-care services during and after pregnancy and more likely to be prescribed an antidepressant 2 years following the end of pregnancy than women who did not initiate or continue antidepressants during pregnancy (odds ratioinitiation 2.16, 95% confidence interval 1.95 to 2.39; odds ratiocontinuation 2.40, 95% confidence interval 2.27 to 2.53). There was little evidence for any substantial association with autism (odds ratiomultivariableregression 1.10, 95% confidence interval 0.90 to 1.35; odds ratiopropensityscore 1.06, 95% confidence interval 0.84 to 1.32), attention deficit hyperactivity disorder (odds ratiomultivariableregression 1.02, 95% confidence interval 0.80 to 1.29; odds ratiopropensityscore 0.97, 95% confidence interval 0.75 to 1.25) or intellectual disability (odds ratiomultivariableregression 0.81, 95% confidence interval 0.55 to 1.19; odds ratiopropensityscore 0.89, 95% confidence interval 0.61 to 1.31) in children of women who continued antidepressants compared with those who discontinued antidepressants. There was inconsistent evidence of an association between initiation of antidepressants in pregnancy and diagnosis of autism in offspring (odds ratiomultivariableregression 1.23, 95% confidence interval 0.85 to 1.78; odds ratiopropensityscore 1.64, 95% confidence interval 1.01 to 2.66) but not attention deficit hyperactivity disorder or intellectual disability; however, but results were imprecise owing to smaller numbers. Limitations Several causal-inference analyses lacked precision owing to limited numbers. In addition, adherence to the prescribed treatment was not measured. Conclusions Women prescribed antidepressants during pregnancy had greater service use during and after pregnancy than those not prescribed antidepressants. The evidence against any substantial association with autism, attention deficit hyperactivity disorder or intellectual disability in the children of women who continued compared with those who discontinued antidepressants in pregnancy is reassuring. Potential association of initiation of antidepressants during pregnancy with offspring autism needs further investigation. Future work Further research on larger samples could increase the robustness and precision of these findings. These methods applied could be a template for future pharmaco-epidemiological investigation of other pregnancy-related prescribing safety concerns. Funding This project was funded by the National Institute for Health and Care Research (NIHR) Health Technology Assessment programme (15/80/19) and will be published in full in Health Technology Assessment; Vol. 27, No. 15. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- Hein Heuvelman
- Department of Population Health Sciences, University of Bristol, Bristol, UK
| | - Neil M Davies
- Department of Population Health Sciences, University of Bristol, Bristol, UK
| | - Yoav Ben-Shlomo
- Department of Population Health Sciences, University of Bristol, Bristol, UK
| | - Alan Emond
- Department of Population Health Sciences, University of Bristol, Bristol, UK
| | - Jonathan Evans
- Department of Population Health Sciences, University of Bristol, Bristol, UK
| | - David Gunnell
- Department of Population Health Sciences, University of Bristol, Bristol, UK
| | - Rachel Liebling
- Fetal Medicine Unit, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Richard Morris
- Department of Population Health Sciences, University of Bristol, Bristol, UK
| | - Rupert Payne
- Department of Population Health Sciences, University of Bristol, Bristol, UK
| | | | | | - Dheeraj Rai
- Department of Population Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
24
|
Støier JF, Jørgensen TN, Sparsø T, Rasmussen HB, Kumar V, Newman AH, Blakely RD, Werge T, Gether U, Herborg F. Disruptive mutations in the serotonin transporter associate serotonin dysfunction with treatment-resistant affective disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.08.29.23294386. [PMID: 37693601 PMCID: PMC10491376 DOI: 10.1101/2023.08.29.23294386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Affective or mood disorders are a leading cause of disability worldwide. The serotonergic system has been heavily implicated in the complex etiology and serves as a therapeutic target. The serotonin transporter (SERT) is a major regulator of serotonin neurotransmission, yet the disease-relevance of impaired SERT function remains unknown. Here, we present the first identification and functional characterization of disruptive coding SERT variants found in patients with psychiatric diseases. In a unique cohort of 144 patients characterized by treatment-resistant chronic affective disorders with a lifetime history of electroconvulsive therapy, we identified two previously uncharacterized coding SERT variants: SERT-N217S and SERT-A500T. Both variants were significantly enriched in the patient cohort compared to GnomAD (SERT-N217S: OR = 151, P = 0.0001 and SERT-A500T: OR = 1348, P = 0.0022) and ethnicity-matched healthy controls (SERT-N217S: OR ≥ 17.7, P ≤ 0.013 and SERT-A500T: OR = ∞, P = 0.029). Functional investigations revealed that the mutations exert distinct perturbations to SERT function, but their overall effects converge on a partial loss-of-function molecular phenotype. Thus, the SERT-A500T variant compromises the catalytic activity, while SERT-N217S disrupts proper glycosylation of SERT with a resulting dominant-negative trafficking deficiency. Moreover, we demonstrate that the trafficking deficiency of SERT-N217S is amenable to pharmacochaperoning by noribogaine. Collectively, our findings describe the first disease-associated loss-of-function SERT variants and implicate serotonergic disturbances arising from SERT dysfunction as a risk factor for chronic affective disorders.
Collapse
|
25
|
Bahi A, Dreyer JL. Anxiety and ethanol consumption in socially defeated mice; effect of hippocampal serotonin transporter knockdown. Behav Brain Res 2023; 451:114508. [PMID: 37244437 DOI: 10.1016/j.bbr.2023.114508] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/13/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023]
Abstract
The comorbidity of generalized anxiety disorders (GAD) with alcohol use disorders (AUD) is common and there is an association between the serotonin transporter (SERT) genetic variation and the comorbid conditions of GAD and AUD. However, few mechanistic studies have systematically explored the role of direct SERT manipulation in stress-elicited mood disorders. Therefore, the aim of this study was to determine whether reductions in SERT expression in the hippocampus were sufficient to ameliorate anxiety- and ethanol-related behaviors in socially defeated mice. Following stress exposure, and using stereotaxic surgery, SERT was knocked down using specific shRNA-expressing lentiviral vectors and anxiety-like behavior was evaluated by open-field, elevated plus maze, and marbles burying test. The two-bottle choice (TBC) drinking paradigm was used to assess stress-induced voluntary ethanol intake and preference. Results showed that hippocampal SERT loss-of-function prevented stress-elicited anxiogenic-like effects with no differences in spontaneous locomotor activity. Moreover, in the TBC paradigm, SERT shRNA-injected mice consistently showed a significantly decreased consumption and preference for ethanol when compared to Mock-injected controls. In contrast to ethanol, SERT shRNA-injected mice exhibited similar consumption and preference for saccharin and quinine. Interestingly, we confirmed that SERT hippocampal mRNA expression correlated with measures of anxiety- and ethanol-related behaviors by Pearson correlation analysis. Our findings show that social defeat recruits hippocampal serotoninergic system and that these neuroadaptations mediate the heightened anxiety-like behavior and voluntary alcohol intake observed following stress exposure, suggesting that this system represents a major brain stress element responsible for the negative reinforcement associated with the "dark side" of alcohol addiction.
Collapse
Affiliation(s)
- Amine Bahi
- College of Medicine, Ajman University, Ajman, UAE; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, UAE; Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, UAE.
| | - Jean-Luc Dreyer
- Division of Biochemistry, Department of Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| |
Collapse
|
26
|
Esen BÖ, Ehrenstein V, Nørgaard M, Sørensen HT, Pedersen L. Prenatal Antidepressant Exposure and the Risk of Attention-Deficit/Hyperactivity Disorder in Childhood: Accounting for Misclassification of Exposure. Epidemiology 2023; 34:476-486. [PMID: 36976730 DOI: 10.1097/ede.0000000000001615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
BACKGROUND Many studies of prenatal antidepressant exposure and the risk of attention-deficit/hyperactivity disorder (ADHD) have done little to reduce bias from exposure misclassification. We assessed the prenatal antidepressant-ADHD effect by incorporating information on repeatedly redeemed prescriptions and redemptions of drug classes commonly used in pregnancy in the analyses to reduce bias from exposure misclassification. METHODS Using population-based registries, we conducted a nationwide cohort study of all children born in Denmark from 1997 to 2017. In a former-user analysis, we compared children prenatally exposed, defined by a redeemed prescription by the mother during pregnancy, to a comparison cohort consisting of prenatally unexposed children whose mothers had redeemed a prescription before pregnancy. We incorporated information on repeatedly redeemed prescriptions and redemptions of drug classes commonly used in pregnancy in the analyses to reduce bias from exposure misclassification. We used incidence rate ratios (IRRs) and incidence rate differences (IRDs) as effect measures. RESULTS The cohort included 1,253,362 children, among whom 24,937 were prenatally exposed to antidepressants. The comparison cohort consisted of 25,698 children. During follow-up, 1,183 exposed children and 1,291 children in the comparison cohort developed ADHD yielding an IRR of 1.05 (95% confidence interval [CI] = 0.96, 1.15) and an IRD of 0.28 (95% CI = -0.20, 0.80) pr. 1,000 person-years. IRRs from analyses attempting to reduce exposure misclassification varied from 1.03 to 1.07. CONCLUSIONS Our results were not consistent with the hypothesized effect of prenatal antidepressant exposure on the risk of ADHD. Attempts to reduce exposure misclassification did not alter this finding.
Collapse
Affiliation(s)
- Buket Öztürk Esen
- From the Department of Clinical Epidemiology, Aarhus University, Aarhus, Denmark
| | | | | | | | | |
Collapse
|
27
|
Bliddal M, Wesselhoeft R, Strandberg-Larsen K, Ernst MT, Weissman MM, Gingrich JA, Talati A, Pottegård A. Prenatal antidepressant exposure and emotional disorders until age 22: a danish register study. Child Adolesc Psychiatry Ment Health 2023; 17:73. [PMID: 37328889 DOI: 10.1186/s13034-023-00624-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/30/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Selective serotonin reuptake inhibitors (SSRIs) are the most frequently prescribed antidepressants in pregnancy. Animal and some clinical studies have suggested potential increases in depression and anxiety following prenatal SSRI exposure, but the extent to which these are driven by the medication remains unclear. We used Danish population data to test associations between maternal SSRI use during pregnancy and children outcomes up to age 22. METHODS We prospectively followed 1,094,202 single-birth Danish children born 1997-2015. The primary exposure was ≥ 1 SSRI prescription filled during pregnancy; the primary outcome, first diagnosis of a depressive, anxiety, or adjustment disorder, or redeemed prescription for an antidepressant medication. We used propensity score weights to adjust potential confounders, and incorporated data from the Danish National Birth Cohort (1997-2003) to further quantify potential residual confounding by subclinical factors. RESULTS The final dataset included 15,651 exposed and 896,818 unexposed, children. After adjustments, SSRI-exposed had higher rates of the primary outcome than those of mothers who either did not use an SSRI (HR = 1.55 [95%CI:1.44,1.67] or discontinued the SSRI use ≥ 3 months prior to conception (HR = 1.23 [1.13,1.34]). Age of onset was earlier among exposed (9 [IQR:7-13] years) versus unexposed (12 [IQR:12-17] years) children (p < 0.01). Paternal SSRI use in the absence of maternal use during the index pregnancy (HR = 1.46 [1.35,1.58]) and maternal SSRI use only after pregnancy (HR = 1.42 [1.35,1.49]) were each also associated with these outcomes. CONCLUSIONS While SSRI exposure was associated with increased risk in the children, this risk may be driven at least partly by underlying severity of maternal illness or other confounding factors.
Collapse
Affiliation(s)
- Mette Bliddal
- Research unit OPEN, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, University of Southern Denmark, Odense, Denmark
| | - Rikke Wesselhoeft
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, University of Southern Denmark, Odense, Denmark
- Department of Child and Adolescent Psychiatry, Odense Mental Health Services in the Region of Southern Denmark, Odense, Denmark
| | | | - Martin T Ernst
- Research unit OPEN, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, University of Southern Denmark, Odense, Denmark
| | - Myrna M Weissman
- Department of Psychiatry, Columbia University Irving Medical Center and Vagelos College of Physicians and Surgeons, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Jay A Gingrich
- Department of Psychiatry, Columbia University Irving Medical Center and Vagelos College of Physicians and Surgeons, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Ardesheer Talati
- Department of Psychiatry, Columbia University Irving Medical Center and Vagelos College of Physicians and Surgeons, New York, NY, USA.
- New York State Psychiatric Institute, New York, NY, USA.
- Columbia University and New York State Psychiatric Institute, 1051 Riverside Drive, Suite 6402B / Unit 24, New York, NY, 10032, USA.
| | - Anton Pottegård
- Research unit OPEN, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
28
|
Wang A, Wan X, Zhuang P, Jia W, Ao Y, Liu X, Tian Y, Zhu L, Huang Y, Yao J, Wang B, Wu Y, Xu Z, Wang J, Yao W, Jiao J, Zhang Y. High fried food consumption impacts anxiety and depression due to lipid metabolism disturbance and neuroinflammation. Proc Natl Acad Sci U S A 2023; 120:e2221097120. [PMID: 37094155 PMCID: PMC10160962 DOI: 10.1073/pnas.2221097120] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/20/2023] [Indexed: 04/26/2023] Open
Abstract
Western dietary patterns have been unfavorably linked with mental health. However, the long-term effects of habitual fried food consumption on anxiety and depression and underlying mechanisms remain unclear. Our population-based study with 140,728 people revealed that frequent fried food consumption, especially fried potato consumption, is strongly associated with 12% and 7% higher risk of anxiety and depression, respectively. The associations were more pronounced among male and younger consumers. Consistently, long-term exposure to acrylamide, a representative food processing contaminant in fried products, exacerbates scototaxis and thigmotaxis, and further impairs exploration ability and sociality of adult zebrafish, showing anxiety- and depressive-like behaviors. Moreover, treatment with acrylamide significantly down-regulates the gene expression of tjp2a related to the permeability of blood-brain barrier. Multiomics analysis showed that chronic exposure to acrylamide induces cerebral lipid metabolism disturbance and neuroinflammation. PPAR signaling pathway mediates acrylamide-induced lipid metabolism disorder in the brain of zebrafish. Especially, chronic exposure to acrylamide dysregulates sphingolipid and phospholipid metabolism, which plays important roles in the development of anxiety and depression symptoms. In addition, acrylamide promotes lipid peroxidation and oxidation stress, which participate in cerebral neuroinflammation. Acrylamide dramatically increases the markers of lipid peroxidation, including (±)5-HETE, 11(S)-HETE, 5-oxoETE, and up-regulates the expression of proinflammatory lipid mediators such as (±)12-HETE and 14(S)-HDHA, indicating elevated cerebral inflammatory status after chronic exposure to acrylamide. Together, these results both epidemiologically and mechanistically provide strong evidence to unravel the mechanism of acrylamide-triggered anxiety and depression, and highlight the significance of reducing fried food consumption for mental health.
Collapse
Affiliation(s)
- Anli Wang
- Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang310058, China
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310003, China
| | - Xuzhi Wan
- Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang310058, China
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310003, China
| | - Pan Zhuang
- Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang310058, China
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310003, China
| | - Wei Jia
- Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang310058, China
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310003, China
| | - Yang Ao
- Department of Nutrition, School of Public Health, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310058, China
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310009, China
| | - Xiaohui Liu
- Department of Nutrition, School of Public Health, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310058, China
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310009, China
| | - Yimei Tian
- Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang310058, China
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310003, China
| | - Li Zhu
- Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang310058, China
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310003, China
| | - Yingyu Huang
- Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang310058, China
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310003, China
| | - Jianxin Yao
- Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang310058, China
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310003, China
| | - Binjie Wang
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou, Zhejiang310053, China
| | - Yuanzhao Wu
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou, Zhejiang310053, China
| | - Zhongshi Xu
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou, Zhejiang310053, China
| | - Jiye Wang
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou, Zhejiang310053, China
| | - Weixuan Yao
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou, Zhejiang310053, China
| | - Jingjing Jiao
- Department of Nutrition, School of Public Health, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310058, China
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310009, China
| | - Yu Zhang
- Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang310058, China
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310003, China
| |
Collapse
|
29
|
Moreau AL, Voss M, Hansen I, Paul SE, Barch DM, Rogers CE, Bogdan R. Prenatal Selective Serotonin Reuptake Inhibitor Exposure, Depression, and Brain Morphology in Middle Childhood: Results From the ABCD Study. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:243-254. [PMID: 37124359 PMCID: PMC10140451 DOI: 10.1016/j.bpsgos.2022.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/14/2021] [Accepted: 02/04/2022] [Indexed: 11/16/2022] Open
Abstract
Background Prenatal selective serotonin reuptake inhibitor (SSRI) exposure has been inconsistently linked to depression, and little is known about neural correlates. We examined whether prenatal SSRI exposure is associated with depressive symptoms and brain structure during middle childhood. Methods Prenatal SSRI exposure (retrospective caregiver report), depressive symptoms (caregiver-reported Child Behavior Checklist), and brain structure (magnetic resonance imaging-derived subcortical volume; cortical thickness and surface area) were assessed in children (analytic ns = 5420-7528; 235 with prenatal SSRI exposure; 9-10 years of age) who completed the baseline Adolescent Brain Cognitive Development Study session. Linear mixed-effects models nested data. Covariates included familial, pregnancy, and child variables. Matrix spectral decomposition adjusted for multiple testing. Results Prenatal SSRI exposure was not independently associated with depression after accounting for recent maternal depressive symptoms. Prenatal SSRI exposure was associated with greater left superior parietal surface area (b = 145.3 mm2, p = .00038) and lateral occipital cortical thickness (b = 0.0272 mm, p = .0000079); neither was associated with child depressive symptoms. Child depression was associated with smaller global brain structure. Conclusions Our findings, combined with adverse outcomes of exposure to maternal depression and the utility of SSRIs for treating depression, suggest that risk for depression during middle childhood should not discourage SSRI use during pregnancy. Associations between prenatal SSRI exposure and brain structure were small in magnitude and not associated with depression. It will be important for future work to examine associations between prenatal SSRI exposure and depression through young adulthood, when risk for depression increases.
Collapse
Affiliation(s)
- Allison L. Moreau
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri
| | - Michaela Voss
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri
| | - Isabella Hansen
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri
| | - Sarah E. Paul
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri
| | - Deanna M. Barch
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, Missouri
- Department of Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Cynthia E. Rogers
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Ryan Bogdan
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri
| |
Collapse
|
30
|
Radford-Smith DE, Anthony DC. Mechanisms of Maternal Diet-Induced Obesity Affecting the Offspring Brain and Development of Affective Disorders. Metabolites 2023; 13:455. [PMID: 36984895 PMCID: PMC10053489 DOI: 10.3390/metabo13030455] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/16/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Depression and metabolic disease are common disorders that share a bidirectional relationship and continue to increase in prevalence. Maternal diet and maternal behaviour both profoundly influence the developmental trajectory of offspring during the perinatal period. At an epidemiological level, both maternal depression and obesity during pregnancy have been shown to increase the risk of neuropsychiatric disease in the subsequent generation. Considerable progress has been made to understand the mechanisms by which maternal obesity disrupts the developing offspring gut-brain axis, priming offspring for the development of affective disorders. This review outlines such mechanisms in detail, including altered maternal care, the maternal microbiome, inflammation, breast milk composition, and maternal and placental metabolites. Subsequently, offspring may be prone to developing gut-brain interaction disorders with concomitant changes to brain energy metabolism, neurotransmission, and behaviour, alongside gut dysbiosis. The gut microbiome may act as a key modifiable, and therefore treatable, feature of the relationship between maternal obesity and the offspring brain function. Further studies examining the relationship between maternal nutrition, the maternal microbiome and metabolites, and offspring neurodevelopment are warranted to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Daniel E. Radford-Smith
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford OX37JX, UK
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford OX13TA, UK
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX13QT, UK
| | - Daniel C. Anthony
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX13QT, UK
| |
Collapse
|
31
|
Hutchison SM, Brain U, Grunau RE, Kuzeljevic B, Irvine M, Mâsse LC, Oberlander TF. Associations between maternal depressive symptoms and selective serotonin reuptake inhibitor antidepressant treatment on internalising and anxiety behaviours in children: 12-year longitudinal study. BJPsych Open 2023; 9:e26. [PMID: 36721917 PMCID: PMC9970165 DOI: 10.1192/bjo.2022.623] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Prenatal selective serotonin reuptake inhibitor (SSRI) antidepressant exposure is associated with increased internalising and anxious behaviours in young children; whether this continues into early adolescence is unknown. Also, it is not well established whether it is the in utero exposure to SSRIs or the underlying maternal mood that contributes more to these associations. AIMS To examine associations between maternal depressive symptoms, prenatal SSRI antidepressant treatment and internalising and anxiety behaviours from childhood into pre-adolescence. METHOD From a prospective longitudinal cohort, measures of maternal depressive symptoms and SSRI use and child outcomes (n = 191 births) were obtained from the second trimester to 12 years. Maternal reports of internalising and anxiety behaviours in children were obtained at 3, 6 and 12 years. RESULTS Multilevel mixed-effects models revealed that maternal depressed mood at the third trimester assessment, not prenatal SSRI exposure, was associated with longitudinal patterns of higher levels of internalising and anxiety behaviours across childhood from 3 to 12 years of age. At each age, hierarchical regressions showed that maternal mood at the third trimester, compared with current maternal depression or prenatal SSRI exposure, explained a greater proportion of the variance in internalising and anxiety behaviours. CONCLUSIONS Even with prenatal SSRI treatment, maternal depressed mood during the third trimester still had an enduring effect as it was associated with increased levels of internalising and anxiety behaviours across childhood and into early adolescence. Importantly, we found no evidence of a 'main effect' association between prenatal SSRI exposure and internalising and anxiety behaviours in children.
Collapse
Affiliation(s)
- Sarah M Hutchison
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Ursula Brain
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Ruth E Grunau
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Boris Kuzeljevic
- (deceased), Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Mike Irvine
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Louise C Mâsse
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Tim F Oberlander
- Department of Pediatrics, University of British Columbia, Vancouver, Canada; and School of Population and Public Health, University of British Columbia, Vancouver, Canada
| |
Collapse
|
32
|
Zengeler KE, Shapiro DA, Bruch KR, Lammert CR, Ennerfelt H, Lukens JR. SSRI treatment modifies the effects of maternal inflammation on in utero physiology and offspring neurobiology. Brain Behav Immun 2023; 108:80-97. [PMID: 36343752 PMCID: PMC10291741 DOI: 10.1016/j.bbi.2022.10.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/17/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Perturbations to the in utero environment can dramatically change the trajectory of offspring neurodevelopment. Insults commonly encountered in modern human life such as infection, toxins, high-fat diet, prescription medications, and others are increasingly linked to behavioral alterations in prenatally-exposed offspring. While appreciation is expanding for the potential consequence that these triggers can have on embryo development, there is a paucity of information concerning how the crucial maternal-fetal interface (MFI) responds to these various insults and how it may relate to changes in offspring neurodevelopment. Here, we found that the MFI responds both to an inflammatory state and altered serotonergic tone in pregnant mice. Maternal immune activation (MIA) triggered an acute inflammatory response in the MFI dominated by interferon signaling that came at the expense of ordinary development-related transcriptional programs. The major MFI compartments, the decidua and the placenta, each responded in distinct manners to MIA. MFIs exposed to MIA were also found to have disrupted sex-specific gene expression and heightened serotonin levels. We found that offspring exposed to MIA had sex-biased behavioral changes and that microglia were not transcriptionally impacted. Moreover, the combination of maternal inflammation in the presence of pharmacologic inhibition of serotonin reuptake further transformed MFI physiology and offspring neurobiology, impacting immune and serotonin signaling pathways alike. In all, these findings highlight the complexities of evaluating diverse environmental impacts on placental physiology and neurodevelopment.
Collapse
Affiliation(s)
- Kristine E Zengeler
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA; Cell and Molecular Biology Graduate Training Program, University of Virginia, Charlottesville, VA 22908, USA.
| | - Daniel A Shapiro
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Katherine R Bruch
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Catherine R Lammert
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
| | - Hannah Ennerfelt
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA; Cell and Molecular Biology Graduate Training Program, University of Virginia, Charlottesville, VA 22908, USA
| | - John R Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA; Cell and Molecular Biology Graduate Training Program, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
33
|
Kisner A, Polter AM. Maturation of glutamatergic transmission onto dorsal raphe serotonergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.524776. [PMID: 36711665 PMCID: PMC9882295 DOI: 10.1101/2023.01.19.524776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Serotonergic neurons in the dorsal raphe nucleus (DRN) play important roles early in postnatal development in the maturation and modulation of higher order emotional, sensory, and cognitive circuitry. This unique position makes these cells a substrate by which early experience can be wired into brain. In this study, we have investigated the maturation of synapses onto dorsal raphe serotonergic neurons in typically developing male and female mice using whole-cell patch-clamp recordings in ex vivo brain slices. We show that while inhibition of these neurons is relatively stable across development, glutamatergic synapses greatly increase in strength between P6 and P21-23. In contrast to forebrain regions, where the components making up glutamatergic synapses are dynamic across early life, we find that the makeup of these synapses onto DRN serotonergic neurons is largely stable after P15. DRN excitatory synapses maintain a very high ratio of AMPA to NMDA receptors and a rectifying component of the AMPA response throughout the lifespan. Overall, these findings reveal that the development of serotonergic neurons is marked by a significant refinement of glutamatergic synapses during the first 3 postnatal weeks. This suggests this time as a sensitive period of heightened plasticity for integration of information from upstream brain areas and that genetic and environmental insults during this period could lead to alterations in serotonergic output, impacting both the development of forebrain circuits and lifelong neuromodulatory actions.
Collapse
Affiliation(s)
- Alexandre Kisner
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
- Current address: Department of Neuroscience, American University, Washington DC 20016
| | - Abigail M. Polter
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| |
Collapse
|
34
|
Bravo K, González-Ortiz M, Beltrán-Castillo S, Cáceres D, Eugenín J. Development of the Placenta and Brain Are Affected by Selective Serotonin Reuptake Inhibitor Exposure During Critical Periods. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:179-198. [PMID: 37466774 DOI: 10.1007/978-3-031-32554-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are usually prescribed to treat major depression and anxiety disorders. Fetal brain development exhibits dependency on serotonin (5-hydroxytryptamine, 5-HT) from maternal, placental, and fetal brain sources. At very early fetal stages, fetal serotonin is provided by maternal and placental sources. However, in later fetal stages, brain sources are indispensable for the appropriate development of neural circuitry and the rise of emergent functions implied in behavior acquisition. Thus, susceptible serotonin-related critical periods are recognized, involving the early maternal and placental 5-HT synthesis and the later endogenous 5-HT synthesis in the fetal brain. Acute and chronic exposure to SSRIs during these critical periods may result in short- and long-term placental and brain dysfunctions affecting intrauterine and postnatal life. Maternal and fetal cells express serotonin receptors which make them susceptible to changes in serotonin levels influenced by SSRIs. SSRIs block the serotonin transporter (SERT), which is required for 5-HT reuptake from the synaptic cleft into the presynaptic neuron. Chronic SSRI administration leads to pre- and postsynaptic 5-HT receptor rearrangement. In this review, we focus on the effects of SSRIs administered during critical periods upon placentation and brain development to be considered in evaluating the risk-safety balance in the clinical use of SSRIs.
Collapse
Affiliation(s)
- Karina Bravo
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile USACH, Santiago, Chile.
- Facultad de Ingeniería, Universidad Autónoma de Chile, Santiago, Chile.
| | - Marcelo González-Ortiz
- Laboratorio de Investigación Materno-Fetal (LIMaF), Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Concepción, Concepción, Chile
| | - Sebastian Beltrán-Castillo
- Centro integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | - Daniela Cáceres
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile USACH, Santiago, Chile
| | - Jaime Eugenín
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile USACH, Santiago, Chile
| |
Collapse
|
35
|
Thanos PK, McCarthy M, Senior D, Watts S, Connor C, Hammond N, Blum K, Hadjiargyrou M, Komatsu D, Steiner H. Combined Chronic Oral Methylphenidate and Fluoxetine Treatment During Adolescence: Effects on Behavior. Curr Pharm Biotechnol 2023; 24:1307-1314. [PMID: 36306463 DOI: 10.2174/1389201024666221028092342] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/14/2022] [Accepted: 09/22/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Attention Deficit Hyperactivity Disorder (ADHD) can be comorbid with depression, often leading to the prescription of both methylphenidate (MP) and selective serotonin reuptake inhibitor (SSRI) antidepressants, such as fluoxetine (FLX). Moreover, these drugs are often misused as cognitive enhancers. This study examined the effects of chronic oral co-administration of MP and FLX on depressive- and anxiety-like behaviors. METHODS Adolescent rats received daily either water (control), MP, FLX, or the combination of MP plus FLX in their drinking water over the course of 4 weeks. RESULTS Data analysis shows a decrease in food consumption and body weight for rats exposed to FLX or the combination of MP and FLX. Sucrose consumption was significantly greater in FLX or MP+FLX groups compared to controls. FLX-treated rats showed no effect in the elevated plus maze (EPM; open arm time) and forced swim test (FST; latency to immobility). However, rats treated with the combination (MP+FLX) showed significant anxiolytic-like and anti-depressive-like behaviors (as measured by EPM and FST), as well as significant increases in overall activity (distance traveled in open field test). Finally, the combined MP+FLX treatment induced a decrease in anxiety and depressive- like behaviors significantly greater than the response from either of these drugs alone. CONCLUSION These behavioral results characterize the long-term effects of these drugs (orally administered) that are widely co-administered and co-misused and provide important insight into the potential neurobiological and neurochemical effects. Future research will determine the potential risks of the long-term use of MP and FLX together.
Collapse
Affiliation(s)
- Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory (BNNL), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14051, USA
- Department of Psychology, University at Buffalo, Buffalo, NY, 14203, USA
| | - Madison McCarthy
- Behavioral Neuropharmacology and Neuroimaging Laboratory (BNNL), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14051, USA
| | - Daniela Senior
- Behavioral Neuropharmacology and Neuroimaging Laboratory (BNNL), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14051, USA
| | - Samantha Watts
- Behavioral Neuropharmacology and Neuroimaging Laboratory (BNNL), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14051, USA
| | - Carly Connor
- Behavioral Neuropharmacology and Neuroimaging Laboratory (BNNL), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14051, USA
| | - Nikki Hammond
- Behavioral Neuropharmacology and Neuroimaging Laboratory (BNNL), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14051, USA
| | - Kenneth Blum
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Michael Hadjiargyrou
- Department of Life Sciences, New York Institute of Technology, Old Westbury, NY, USA
| | - David Komatsu
- Department of Orthopedics, Stony Brook University, Stony Brook, NY, USA
| | - Heinz Steiner
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| |
Collapse
|
36
|
Edem EE, Okhonmina UE, Nebo KE, Akinluyi ET, Ikuelogbon DA, Fafure AA, Olabiyi AA, Adedokun MA. Combined Exposure to Chronic Sleep Deprivation and Caffeine Potentiates Behavioural Deficits by Altering Neurochemical Profile and Synaptophysin Expression in Long-Evans Rats. Neurotox Res 2022; 40:2001-2015. [PMID: 36434357 DOI: 10.1007/s12640-022-00589-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/22/2022] [Accepted: 10/08/2022] [Indexed: 11/27/2022]
Abstract
Using the Unpredictable Chronic Sleep Deprivation (UCSD) paradigm we developed, the combined effects of chronic sleep deprivation and high caffeine intake on prefrontal cortical synaptophysin expression, neurochemical profiles, and behavioural outcomes in Long-Evans rats were evaluated. The combination of chronic sleep deprivation and high-dose caffeine treatment produced varying degrees of behavioural impairments, depletion of antioxidants, serotonin, and an upregulation of acetylcholinesterase (AChE) activity in the prefrontal cortex. An immunohistochemical assessment revealed a reduction in synaptophysin protein expression in the prefrontal cortex following exposure to high-dose caffeine and chronic sleep deprivation. Overall, our findings support the advocacy for adequate sleep for optimal mental performance as a high intake of caffeine to attenuate the effects of sleep deprivation that may alter the neurochemical profile and synaptic plasticity in the prefrontal cortex, significantly increasing the risk of neuropsychiatric/degenerative disorders.
Collapse
Affiliation(s)
- Edem Ekpenyong Edem
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria. .,Department of Anatomy, College of Medicine, University of Lagos, Idi-Araba, Lagos State, Nigeria.
| | - Uyi Emmanuel Okhonmina
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Kate Eberechukwu Nebo
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Elizabeth Toyin Akinluyi
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | | | - Adedamola Adediran Fafure
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Ayodeji Augustine Olabiyi
- Department of Medical Biochemistry, College of Medicine and Health Sciences, Afe Babalola University, Ekiti State, Ado-Ekiti, Nigeria
| | - Mujeeb Adekunle Adedokun
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| |
Collapse
|
37
|
Silveira JS, Júnior OVR, Schmitz F, Ferreira FS, Rodrigues FC, Deon M, Ribas G, Coutinho-Silva R, Vargas CR, Savio LEB, Wyse AT. High-protein nutrition during pregnancy increases neuroinflammation and homocysteine levels and impairs behavior in male adolescent rats offspring. Life Sci 2022; 310:121084. [DOI: 10.1016/j.lfs.2022.121084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/04/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022]
|
38
|
Linhares SSG, Meurer YDSR, de Aquino ACQ, Câmara DDA, Brandão LEM, Fiuza FP, Lima RH, Engelberth RCJG, Cavalcante JS. Prenatal exposure to fluoxetine modulates emotionality and aversive memory in male and female rat offspring. Behav Pharmacol 2022; 33:575-588. [PMID: 36256730 DOI: 10.1097/fbp.0000000000000705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During pregnancy, women are prone to depression, for which selective serotonin reuptake inhibitors (SSRIs), such as fluoxetine, are usually the first-line treatment. However, fluoxetine can cross the placental barrier and affect fetuses, causing changes in serotonin levels early in life. Long-term effects in the brain circuits that control cognitive and emotional behavior are related to early fluoxetine exposure during development. In this study, we aimed to investigate whether fluoxetine exposure (10 mg/kg/day) from the 13th gestational day (GD13) to GD21 may lead to behavioral emotional-cognitive changes in male and female rat offspring approximately 90 days postnatally (~PN90). We have analyzed the performance of individuals in the open field and in the plus-maze discriminative avoidance task, which assesses anxiety and learning/memory processing behaviors. We have found that prenatal (GD13-GD21) exposure to fluoxetine strengthened aversive memory and induced higher anxiety levels in males, and quick extinction of aversive memory in females. Taken together, these results suggest that early exposure to fluoxetine impairs the basal state of anxiety and the cognitive functions of rats during adulthood, which may be in a sex-specific manner because males appear more susceptible than females.
Collapse
Affiliation(s)
- Sarah Sophia G Linhares
- Laboratory of Neurochemical Studies, Department of Physiology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Ywlliane da Silva R Meurer
- Laboratory of Neurochemical Studies, Department of Physiology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Antônio Carlos Queiroz de Aquino
- Laboratory of Neurochemical Studies, Department of Physiology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Diego de Aquino Câmara
- Laboratory of Neurochemical Studies, Department of Physiology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | - Felipe Porto Fiuza
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, Brazil
| | - Ramón Hypolito Lima
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, Brazil
| | - Rovena Clara J G Engelberth
- Laboratory of Neurochemical Studies, Department of Physiology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Jeferson Souza Cavalcante
- Laboratory of Neurochemical Studies, Department of Physiology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| |
Collapse
|
39
|
Schreiner C, Powell TL, Palmer C, Jansson T. Placental proteins with predicted roles in fetal development decrease in premature infants. Pediatr Res 2022; 92:1316-1324. [PMID: 35132128 PMCID: PMC9357234 DOI: 10.1038/s41390-022-01942-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/15/2021] [Accepted: 12/23/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Emerging evidence from animal experiments indicate that factors secreted by the placenta are critical for normal fetal organ development. Our objective was to characterize the umbilical vein and artery proteome in preterm infants and identify proteins that decrease in the neonatal circulation following delivery. METHODS Cord blood at delivery and neonatal blood at 48-72 h of life was collected in 25 preterm infants. Plasma protein abundance was determined using the SomaLogic platform. RESULTS When comparing protein levels of umbilical venous to arterial cord blood, 434 proteins were significantly higher indicating placental secretion into the fetal circulation. Moreover, when comparing neonatal blood to umbilical vein levels, 142 proteins were significantly lower. These proteins included Endoplasmic reticulum resident protein 29, CD59, Fibroblast growth factor 2 and Dynactin subunit 2, which are involved in brain development and prevention of brain damage as well as Fibroblast growth factor 1 which prevents lung fibrosis. CONCLUSIONS The late second trimester human placenta secretes proteins into the fetal circulation which decrease following delivery. Many of these proteins are predicted to be important in the development of fetal organs. Further studies are needed to directly link placental proteins to organ development and poor outcomes in preterm infants. IMPACT Prematurity remains a leading cause of morbidity and mortality requiring the development of novel treatments. Emerging evidence from animal studies suggest that factors secreted from the placenta may be critical in the development of the fetus. We report that the preterm human placenta secretes an array of proteins into the fetal circulation. Some of these proteins are predicted to be involved in the development of the brain and the lung. When born prematurely, infants are deprived of these placental proteins, which may contribute to their poor outcomes.
Collapse
Affiliation(s)
- Cynthia Schreiner
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Department of Pediatrics at Renown Children's Hospital, Reno, NV, USA.
| | - Theresa L Powell
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Obstetrics, Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Claire Palmer
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas Jansson
- Department of Obstetrics, Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
40
|
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
|
41
|
Fabio MC, Servin-Bernal IJC, Degano AL, Pautassi RM. Serotonin disruption at gestation alters expression of genes associated with serotonin synthesis and reuptake at weaning. Psychopharmacology (Berl) 2022; 239:3355-3366. [PMID: 36063206 DOI: 10.1007/s00213-022-06228-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/27/2022] [Indexed: 12/28/2022]
Abstract
RATIONALE Serotonin (5-HT) is a monoamine neuromodulator that plays a key role in the organization of the central nervous system. 5-HT alterations may be associated to the emergence of social deficits and psychiatric disorders, including anxiety, depression, and substance abuse disorders. Notably, disruption of the 5-HT system during sensitive periods of development seems to exert long-term consequences, including altered anxiety responses and problematic use of alcohol. OBJECTIVE We analyzed, in mice, the effects of transient 5-HT depletion at gestation (a developmental stage when medial prefrontal cortex (mPFC) 5-HT levels depend exclusively on placental 5-HT availability) on 5-HT central synthesis and reuptake at weaning. We also explored if 5-HT disruption at the embryonic stage influences behavioral outcomes that may serve as a proxy for autistic- or anxiety-like phenotypes. METHODS C57/BL6 male and female mice, born from dams treated with a 5-HT synthesis inhibitor (PCPA; 4-Chloro-DL-phenylalanine methyl ester hydrochloride) at gestational days (G)13.5-16.5, were subjected to a behavioral battery that assesses social preference and novelty, compulsive behavior, stereotypies, and ethanol's anti-anxiety effects, at postnatal days (P) 21-28. Afterwards, expression of the genes that encode for 5-HT synthesis (Tph2) and SERT (5-HT transporter) were analyzed in mPFC via real-time RT-PCR. Dopamine 2 receptor (D2R) expression was also analyzed via RT-PCR to further explore possible effects of PCPA on dopaminergic transmission. RESULTS Transient 5-HT disruption at G13.5-16.5 reduced Tph2 expression of both male and female mice in mPFC at P23. Notably, female mice also exhibited higher SERT expression and reduced D2R expression in mPFC. Mice derived from 5-HT depleted dams displayed heightened compulsive behavior at P21, when compared to control mice. Alcohol anti-anxiety effects at early adolescence (P28) were exhibited by mice derived from 5-HT depleted dams, but not by control counterparts. No social deficits or stereotyped behaviors were observed. CONCLUSION Transient 5-HT inhibition at gestation resulted in altered expression of genes involved in 5-HT synthesis and reuptake in mPFC at weaning, a period in which the 5-HT system is still developing. These alterations may exert lingering effects, which translate to significant compulsivity and heightened sensitivity to the anxiolytic effects of alcohol at early adolescence.
Collapse
Affiliation(s)
- M C Fabio
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Cordoba, Argentina. .,Facultad de Psicología, Universidad Nacional de Córdoba, Cordoba, Argentina.
| | - I J C Servin-Bernal
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Cordoba, Argentina
| | - A L Degano
- Departamento de Química Biológica Ranwel CaputtoFacultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba, Argentina.,Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Universidad Nacional de Córdoba, Cordoba, Argentina
| | - R M Pautassi
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Cordoba, Argentina.,Facultad de Psicología, Universidad Nacional de Córdoba, Cordoba, Argentina
| |
Collapse
|
42
|
Effect of early-life stress or fluoxetine exposure on later-life conditioned taste aversion learning in Sprague-Dawley rats. Neurosci Lett 2022; 787:136818. [PMID: 35931277 DOI: 10.1016/j.neulet.2022.136818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/12/2022] [Accepted: 07/23/2022] [Indexed: 01/06/2023]
Abstract
In rodents, early-life exposure to environmental stress or antidepressant medication treatment has been shown to induce similar long-term consequences on memory- and depression-related behavior in adulthood. To expand on this line of work, we evaluated how juvenile exposure to chronic variable stress (CVS) or the selective serotonin reuptake inhibitor fluoxetine (FLX) influences conditioned taste aversion (CTA) learning in adulthood. To do this, in Experiment 1, we examined how adolescent CVS alone (postnatal day [PND] 35-48), or with prenatal stress (PNS) history (PNS + CVS), influenced the acquisition and extinction of CTA in adult male Sprague Dawley rats. Specifically, at PND70+ (adulthood), rats were presented with 0.15 % saccharin followed by an intraperitoneal (i.p.) injection of lithium chloride (LiCl) to induce visceral malaise. A total of four saccharin (conditioned stimulus) and LiCl (unconditioned stimulus) pairings occurred across the CTA acquisition phase. Next, saccharin was presented without aversive consequences, and intake was measured across consecutive days of the extinction phase. No differences in body weight gain across the experimental days, rate of CTA acquisition, or extinction of CTA, were observed among the experimental groups (control, n = 7; CVS, n = 12; PNS + CVS, n = 9). In Experiment 2, we evaluated if early-life FLX exposure alters CTA learning in adulthood. Specifically, adolescent stress naïve male and female rats received FLX (0 or 20 mg/kg/i.p) once daily for 15 consecutive days (PND35-49). During antidepressant exposure, FLX decreased body weight gain in both male (n = 7) and female rats (n = 7), when compared to respective controls (male control, n = 8; female control, n = 8). However, juvenile FLX exposure decreased body weight-gain in adult male, but not female, rats. Lastly, adolescent FLX history had no effect on CTA acquisition or extinction in adulthood (PND70), in neither male nor female rats. Together, the data indicate that juvenile FLX exposure results in a long-term decrease of body weight-gain in a male-specific manner. Yet, independent of sex, neither early-life stress nor FLX exposure alters CTA learning in adulthood.
Collapse
|
43
|
Uzungil V, Tran H, Aitken C, Wilson C, Opazo CM, Li S, Payet JM, Mawal CH, Bush AI, Hale MW, Hannan AJ, Renoir T. Novel Antidepressant-Like Properties of the Iron Chelator Deferiprone in a Mouse Model of Depression. Neurotherapeutics 2022; 19:1662-1685. [PMID: 35861925 PMCID: PMC9606181 DOI: 10.1007/s13311-022-01257-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 10/17/2022] Open
Abstract
Depressed individuals who carry the short allele for the serotonin-transporter-linked promotor region of the gene are more vulnerable to stress and have reduced response to first-line antidepressants such as selective serotonin reuptake inhibitors. Since depression severity has been reported to correlate with brain iron levels, the present study aimed to characterise the potential antidepressant properties of the iron chelator deferiprone. Using the serotonin transporter knock-out (5-HTT KO) mouse model, we assessed the behavioural effects of acute deferiprone on the Porsolt swim test (PST) and novelty-suppressed feeding test (NSFT). Brain and blood iron levels were also measured following acute deferiprone. To determine the relevant brain regions activated by deferiprone, we then measured c-Fos expression and applied network-based analyses. We found that deferiprone reduced immobility time in the PST in 5-HTT KO mice and reduced latency to feed in the NSFT in both genotypes, suggesting potential antidepressant-like effects. There was no effect on brain or blood iron levels following deferiprone treatment, potentially indicating an acute iron-independent mechanism. Deferiprone reversed the increase in c-Fos expression induced by swim stress in 5-HTT KO mice in the lateral amygdala. Functional network analyses suggest that hub regions of activity in mice treated with deferiprone include the caudate putamen and prefrontal cortex. The PST-induced increase in network modularity in wild-type mice was not observed in 5-HTT KO mice. Altogether, our data show that the antidepressant-like effects of deferiprone could be acting via an iron-independent mechanism and that these therapeutic effects are underpinned by changes in neuronal activity in the lateral amygdala.
Collapse
Affiliation(s)
- Volkan Uzungil
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Harvey Tran
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Connor Aitken
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Carey Wilson
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Carlos M Opazo
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Shanshan Li
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Jennyfer M Payet
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Celeste H Mawal
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Matthew W Hale
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Anthony J Hannan
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Thibault Renoir
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia.
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia.
| |
Collapse
|
44
|
Strużyńska L, Dąbrowska-Bouta B, Sulkowski G. Developmental neurotoxicity of silver nanoparticles: the current state of knowledge and future directions. Nanotoxicology 2022; 16:1-26. [PMID: 35921173 DOI: 10.1080/17435390.2022.2105172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/17/2022] [Accepted: 07/20/2022] [Indexed: 10/16/2022]
Abstract
The increasing production and use of silver nanoparticles (AgNPs) as an antimicrobial agent in an array of medical and commercial products, including those designed for infants and children, poses a substantial risk of exposure during the developmental period. This review summarizes current knowledge on developmental neurotoxicity of AgNPs in both pre- and post-natal stages with a focus on the biological specificity of immature organisms that predisposes them to neurotoxic insults as well as the molecular mechanisms underlying AgNP-induced neurotoxicity. The current review revealed that AgNPs increase the permeability of the blood-brain barrier (BBB) and selectively damage neurons in the brain of immature rats exposed pre and postnatally. Among the AgNP-induced molecular mechanisms underlying toxic insult is cellular stress, which can consequently lead to cell death. Glutamatergic neurons and NMDAR-mediated neurotransmission also appear to be a target for AgNPs during the postnatal period of exposure. Collected data indicate also that our current knowledge of the impact of AgNPs on the developing nervous system remains insufficient and further studies are required during different stages of development with investigation of environmentally-relevant doses of exposure.
Collapse
Affiliation(s)
- Lidia Strużyńska
- Department of Neurochemistry, Laboratory of Pathoneurochemistry, Mossakowski Medical, Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Beata Dąbrowska-Bouta
- Department of Neurochemistry, Laboratory of Pathoneurochemistry, Mossakowski Medical, Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Grzegorz Sulkowski
- Department of Neurochemistry, Laboratory of Pathoneurochemistry, Mossakowski Medical, Research Institute, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
45
|
Collins HM, Pinacho R, Ozdemir D, Bannerman DM, Sharp T. Effect of selective serotonin reuptake inhibitor discontinuation on anxiety-like behaviours in mice. J Psychopharmacol 2022; 36:794-805. [PMID: 35607713 PMCID: PMC9247435 DOI: 10.1177/02698811221093032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Abrupt cessation of therapy with a selective serotonin reuptake inhibitor (SSRI) is associated with a discontinuation syndrome, typified by numerous disabling symptoms, including anxiety. Surprisingly, little is known of the behavioural effect of SSRI discontinuation in animals. AIM Here, the effect of SSRI discontinuation on anxiety-like behaviour was systematically investigated in mice. METHODS Experiments were based on a three-arm experimental design comprising saline, continued SSRI and discontinued SSRI. Mice were assessed 2 days after SSRI discontinuation over a 5-day period using the elevated plus maze (EPM) and other anxiety tests. RESULTS An exploratory experiment found cessation of paroxetine (12 days) was associated with decreased open-arm exploration and reduced total distance travelled, in male but not female mice. Follow-up studies confirmed a discontinuation effect on the EPM in male mice after paroxetine (12 days) and also citalopram (12 days). Mice receiving continued paroxetine (but not citalopram) also showed decreased open-arm exploration but this was dissociable from the effects of discontinuation. The discontinuation response to paroxetine did not strengthen after 28 days of treatment but was absent after 7 days of treatment. A discontinuation response was not discernible in other anxiety and fear-learning tests applied 3-5 days after treatment cessation. Finally, discontinuation effects on the EPM were typically associated with decreased locomotion on the test. However, separate locomotor testing implicated anxiety-provoked behavioural inhibition rather than a general reduction in motor activity. CONCLUSION Overall, this study provides evidence for a short-lasting behavioural discontinuation response to cessation of SSRI treatment in mice.
Collapse
Affiliation(s)
- Helen M Collins
- Department of Pharmacology,
University of Oxford, Oxford, UK,Department of Experimental
Psychology, University of Oxford, Oxford, UK
| | - Raquel Pinacho
- Department of Pharmacology,
University of Oxford, Oxford, UK,Department of Experimental
Psychology, University of Oxford, Oxford, UK
| | - Dersu Ozdemir
- Department of Pharmacology,
University of Oxford, Oxford, UK
| | - David M Bannerman
- Department of Experimental
Psychology, University of Oxford, Oxford, UK
| | - Trevor Sharp
- Department of Pharmacology,
University of Oxford, Oxford, UK,Trevor Sharp, Department of
Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT,
UK.
| |
Collapse
|
46
|
Armand S, Ozenne B, Svart N, Frøkjaer VG, Knudsen GM, Fisher PM, Stenbaek DS. Brain serotonin transporter is associated with cognitive-affective biases in healthy individuals. Hum Brain Mapp 2022; 43:4174-4184. [PMID: 35607850 PMCID: PMC9374883 DOI: 10.1002/hbm.25946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/21/2022] [Accepted: 05/02/2022] [Indexed: 12/26/2022] Open
Abstract
Cognitive affective biases describe the tendency to process negative information or positive information over the other. These biases can be modulated by changing extracellular serotonin (5-HT) levels in the brain, for example, by pharmacologically blocking and downregulating the 5-HT transporter (5-HTT), which remediates negative affective bias. This suggests that higher levels of 5-HTT are linked to a priority of negative information over positive, but this link remains to be tested in vivo in healthy individuals. We, therefore, evaluated the association between 5-HTT levels, as measured with [11 C]DASB positron emission tomography (PET), and affective biases, hypothesising that higher 5-HTT levels are associated with a more negative bias. We included 98 healthy individuals with measures of [11 C]DASB binding potential (BPND ) and affective biases using The Emotional Faces Identification Task by subtracting the per cent hit rate for happy from that of sad faces (EFITAB ). We evaluated the association between [11 C]DASB BPND and EFITAB in a linear latent variable model, with the latent variable (5-HTTLV ) modelled from [11 C]DASB BPND in the fronto-striatal and fronto-limbic networks implicated in affective cognition. We observed an inverse association between 5-HTTLV and EFITAB (β = -8% EFITAB per unit 5-HTTLV , CI = -14% to -3%, p = .002). These findings show that higher 5-HTT levels are linked to a more negative bias in healthy individuals. High 5-HTT supposedly leads to high clearance of 5-HT, and thus, a negative bias could result from low extracellular 5-HT. Future studies must reveal if a similar inverse association exists in individuals with affective disorders.
Collapse
Affiliation(s)
- Sophia Armand
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Brice Ozenne
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Public Health, Section of Biostatistics, University of Copenhagen, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Nanna Svart
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Vibe G Frøkjaer
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Psychiatric Center Copenhagen, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Gitte M Knudsen
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Patrick M Fisher
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Dea S Stenbaek
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Psychology, University of Copenhagen, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
47
|
Guedes Linhares SS, da Silva Rodrigues Meurer Y, Aquino A, Aquino Câmara D, Mateus Brandão LE, Dierschnabel AL, Porto Fiuza F, Hypólito Lima R, Engelberth RC, Cavalcante JS. Effects of prenatal exposure to fluoxetine on circadian rhythmicity in the locomotor activity and neuropeptide Y and 5-HT expression in male and female adult Wistar rats. Int J Dev Neurosci 2022; 82:407-422. [PMID: 35481929 DOI: 10.1002/jdn.10189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/04/2022] [Accepted: 04/03/2022] [Indexed: 11/07/2022] Open
Abstract
Serotonin (5-HT) reuptake inhibitors, such as fluoxetine, are the most prescribed antidepressant for maternal depression. In this sense, it exposes mothers and the brains of infants to increased modulatory and trophic effects of serotonergic neurotransmission. 5-HT promotes essential brain changes throughout its development, which include neuron migration, differentiation, and organization of neural circuitries related to emotional, cognitive, and circadian behavior. Early exposure to the SSRIs induces long-term effects on behavioral and neural serotonergic signalization. We have aimed to evaluate the circadian rhythm of locomotor activity and the neurochemical content, neuropeptide Y (NPY) and 5-HT in three brain areas: intergeniculate leaflet (IGL), suprachiasmatic nuclei (SCN) and raphe nuclei (RN), at two zeitgebers (ZT6 and ZT18), in male and female rat's offspring early exposed (developmental period GD13-GD21) to fluoxetine (20mg/kg). First, we have conducted daily records of the locomotor activity rhythm using activity sensors coupled to individual cages over four weeks. We have lastly evaluated the immunoreactivity of NPY in both SCN and IGL, and as well the 5-HT expression in the dorsal and medial RN. In summary, our results showed that (1) prenatal fluoxetine affects phase entrainment of the rest/activity rhythm at ZT6 and ZT18, more in male than female specimens, and (2) modulates the NPY and 5-HT expression. Here, we show male rats are more susceptible to phase entrainment and the NPY and 5-HT misexpression compared to female ones. The sex differences induced by early exposure to fluoxetine in both the circadian rhythm of locomotor activity and the neurochemical expression into SCN, IGL, and midbrain raphe are an important highlight in the present work. Thus, our results may help to improve the knowledge on neurobiological mechanisms of circadian rhythms and are relevant to understanding the "broken brains" and behavioral abnormalities of offspring early exposed to antidepressants.
Collapse
Affiliation(s)
- Sara Sophia Guedes Linhares
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Ywlliane da Silva Rodrigues Meurer
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Antonio Aquino
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Diego Aquino Câmara
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | - Aline Lima Dierschnabel
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Felipe Porto Fiuza
- Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Graduate Program in Neuroengineering, Macaíba, Brazil
| | - Ramon Hypólito Lima
- Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Graduate Program in Neuroengineering, Macaíba, Brazil
| | - Rovena Clara Engelberth
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Jeferson Souza Cavalcante
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| |
Collapse
|
48
|
Wang X, Wang K, Wu X, Huang W, Yang L. Role of the cAMP-PKA-CREB-BDNF pathway in abnormal behaviours of serotonin transporter knockout mice. Behav Brain Res 2022; 419:113681. [PMID: 34838579 DOI: 10.1016/j.bbr.2021.113681] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 11/02/2022]
Abstract
Serotonin transporter gene-linked polymorphic region polymorphisms are associated with anxiety, neuroticism, affective disorders and vulnerability to stressful life events; however, the relevant physiological mechanisms are not well understood. Serotonin transporter knockout mice have been widely used as a model of allelic variation of serotonin transporter function in humans; herein, wild-type mice and heterozygous and homozygous knockout mice models were established to explore the behavioural changes related to different genotypes and the possible physiological mechanisms. Behavioural changes were assessed using behavioural tests, namely, elevated plus maze, open field, Morris water maze and rotarod tests. Serum indicators were detected using the enzyme-linked immunosorbent assay. Compared with wild-type mice, homozygous mice showed significant anxiety-like behaviours in the plus maze and open field tests; conversely, anxiety-like behaviours in heterozygous mice were less pronounced. Homozygous mice also showed cognitive impairment and motor inhibition in the Morris water maze and rotarod tests. Serotonin levels decreased in both heterozygous and homozygous mice, and 5-hydroxytryptophan, protein kinase A, adenylyl cyclase, cyclic adenosine monophosphate response element-binding protein and brain-derived neurotrophic factor levels were lower in homozygous mice than in wild-type and heterozygous mice, whereas no statistical differences were found between wild-type and heterozygous mice. Additionally, there was a correlation between serological and behavioural indicators. This study provided experimental evidence that the cyclic adenosine monophosphate-protein kinase A-cyclic adenosine monophosphate response element-binding protein-brain-derived neurotrophic factor pathway may be involved in the regulation of polymorphism to stress and enriched the behavioural and physiological characteristics of serotonin transporter knockout mice.
Collapse
Affiliation(s)
- Xiaomin Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning, PR China
| | - Ke Wang
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, PR China
| | - Xiangmin Wu
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, PR China
| | - Wenxiu Huang
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, PR China
| | - Li Yang
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, PR China.
| |
Collapse
|
49
|
Silveira JS, Ramires Júnior OV, Schmitz F, Ferreira FS, Rodrigues FC, Silva RC, Savio LEB, Wyse ATS. Folic acid supplementation during pregnancy alters behavior in male rat offspring: nitrative stress and neuroinflammatory implications. Mol Neurobiol 2022; 59:2150-2170. [PMID: 35044624 DOI: 10.1007/s12035-022-02724-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/30/2021] [Indexed: 12/27/2022]
Abstract
Pregnancy diet can impact offspring's neurodevelopment, metabolism, redox homeostasis, and inflammatory status. In pregnancy, folate demand is increased due to the requirement for one-carbon transfer reactions. The present study was proposed to investigate the effect of folic acid supplementation throughout pregnancy on a battery of behavior tests (olfactory preference, motor activity, exploratory capacity, habituation, memory, anxiety- and depression-like behavior). Redox homeostasis and neuroinflammatory status in cerebral cortex were also investigated. After pregnancy confirmation, the pregnant rats were randomly divided into two groups, according to the diet: group 1, (control) standard diet (2 mg/kg diet of folic acid) and group 2, supplemented diet with 4 mg/kg diet of folic acid. Throughout the gestational period, the pregnant rats received experimental diets. Results show that the supplemented diet with 4 mg/kg diet of folic acid throughout pregnancy impaired memory and motricity of the offspring when compared with control (standard diet). It was also observed an increase in anxiety- and depression-like behavior in this group. Nitrite levels increased in cerebral cortex of the offspring, when compared to control group. In contrast, iNOS expression and immunocontent were not altered. Moreover, we identify an increase in TNF-α, IL-1β, IL-6, IL-10, and MCP-1 gene expression in the cerebral cortex. In conclusion, our study showed that the supplemented diet with 4 mg/kg diet of folic acid throughout pregnancy may cause behavioral and biochemical changes in the male offspringGraphical abstract After pregnancy confirmation, the pregnant rats were randomly divided into two groups, according to the diet: group 1, (control) standard diet (2 mg/kg diet of folic acid) and group 2, supplemented diet with 4 mg/kg diet of folic acid. Throughout the gestational period, the pregnant rats received experimental diets. Results show that folic acid supplementation did not impair the mother-pup relationship. We showed that supplemented diet with 4 mg/kg diet of folic acid during pregnancy impairs memory and motricity of the offspring when compared with standard diet. It was also observed an increase in anxiety- and depression-like behavior in this group. Nitrative stress and neuroinflammation parameters were increased in the cerebral cortex of the offspring. ROS, reactive oxygen species.
Collapse
Affiliation(s)
- Josiane Silva Silveira
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Laboratório de Neuroproteção E Doenças Metabólicas (Wyse's Lab), Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, Rio Grande do Sul, 90035003, Brazil
| | - Osmar Vieira Ramires Júnior
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Laboratório de Neuroproteção E Doenças Metabólicas (Wyse's Lab), Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, Rio Grande do Sul, 90035003, Brazil
| | - Felipe Schmitz
- Laboratório de Neuroproteção E Doenças Metabólicas (Wyse's Lab), Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, Rio Grande do Sul, 90035003, Brazil
| | - Fernanda Silva Ferreira
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Laboratório de Neuroproteção E Doenças Metabólicas (Wyse's Lab), Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, Rio Grande do Sul, 90035003, Brazil
| | - Fabiana Cristina Rodrigues
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Bloco G. Av. Carlos Chagas Filho, 373. Cidade Universitária, Ilha Do Fundão, Rio de Janeiro, 21941-902, Brazil
| | - Robson Coutinho Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Bloco G. Av. Carlos Chagas Filho, 373. Cidade Universitária, Ilha Do Fundão, Rio de Janeiro, 21941-902, Brazil
| | - Luiz Eduardo Baggio Savio
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Bloco G. Av. Carlos Chagas Filho, 373. Cidade Universitária, Ilha Do Fundão, Rio de Janeiro, 21941-902, Brazil
| | - Angela T S Wyse
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Rio Grande do Sul, Brazil. .,Laboratório de Neuroproteção E Doenças Metabólicas (Wyse's Lab), Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, Rio Grande do Sul, 90035003, Brazil.
| |
Collapse
|
50
|
Lee GS, Graham DL, Noble BL, Trammell TS, McCarthy DM, Anderson LR, Rubinstein M, Bhide PG, Stanwood GD. Behavioral and Neuroanatomical Consequences of Cell-Type Specific Loss of Dopamine D2 Receptors in the Mouse Cerebral Cortex. Front Behav Neurosci 2022; 15:815713. [PMID: 35095443 PMCID: PMC8793809 DOI: 10.3389/fnbeh.2021.815713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/22/2021] [Indexed: 11/13/2022] Open
Abstract
Developmental dysregulation of dopamine D2 receptors (D2Rs) alters neuronal migration, differentiation, and behavior and contributes to the psychopathology of neurological and psychiatric disorders. The current study is aimed at identifying how cell-specific loss of D2Rs in the cerebral cortex may impact neurobehavioral and cellular development, in order to better understand the roles of this receptor in cortical circuit formation and brain disorders. We deleted D2R from developing cortical GABAergic interneurons (Nkx2.1-Cre) or from developing telencephalic glutamatergic neurons (Emx1-Cre). Conditional knockouts (cKO) from both lines, Drd2fl/fl, Nkx2.1-Cre+ (referred to as GABA-D2R-cKO mice) or Drd2fl/fl, Emx1-Cre+ (referred to as Glu-D2R-cKO mice), exhibited no differences in simple tests of anxiety-related or depression-related behaviors, or spatial or nonspatial working memory. Both GABA-D2R-cKO and Glu-D2R-cKO mice also had normal basal locomotor activity, but GABA-D2R-cKO mice expressed blunted locomotor responses to the psychotomimetic drug MK-801. GABA-D2R-cKO mice exhibited improved motor coordination on a rotarod whereas Glu-D2R-cKO mice were normal. GABA-D2R-cKO mice also exhibited spatial learning deficits without changes in reversal learning on a Barnes maze. At the cellular level, we observed an increase in PV+ cells in the frontal cortex of GABA-D2R-cKO mice and no noticeable changes in Glu-D2R-cKO mice. These data point toward unique and distinct roles for D2Rs within excitatory and inhibitory neurons in the regulation of behavior and interneuron development, and suggest that location-biased D2R pharmacology may be clinically advantageous to achieve higher efficacy and help avoid unwanted effects.
Collapse
Affiliation(s)
- Gloria S. Lee
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Devon L. Graham
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
- Center for Brain Repair, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Brenda L. Noble
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Taylor S. Trammell
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Deirdre M. McCarthy
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
- Center for Brain Repair, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Lisa R. Anderson
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Marcelo Rubinstein
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas and Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pradeep G. Bhide
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
- Center for Brain Repair, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Gregg D. Stanwood
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
- Center for Brain Repair, Florida State University College of Medicine, Tallahassee, FL, United States
- *Correspondence: Gregg D. Stanwood
| |
Collapse
|