1
|
Veríssimo LF, Alves FHF, Estrada VB, da Costa Marques LA, Andrade KC, Bonancea AM, Okano NT, Corrêa FMDA, Pelosi GG. Cardiovascular effects of early maternal separation and escitalopram treatment in rats with depressive-like behaviour. Auton Neurosci 2024; 256:103223. [PMID: 39616948 DOI: 10.1016/j.autneu.2024.103223] [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: 07/17/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024]
Abstract
Depression and cardiovascular diseases are two of the world's major health problems. Escitalopram (ESC) is widely used because of its safety in relation to other drugs in that class; however, it can affect the cardiovascular system. The present study evaluated the cardiovascular parameters of depressive-like male rats and the cardiovascular effects of ESC treatment on that condition. The EMS protocol consisted of separating the litter from the dam for 3 h over 13 days. Animals were anesthetized with tribromoethanol (250 mg/kg, intraperitoneally) and the catheters were inserted into the femoral and into the femoral vein. Depressive-like rats showed an increase in the pressor response to phenylephrine (Emax:depressive = 50.36 ± 2.997 mmHg; non-depressive = 39.51 ± 3.328 mmHg; p < 0.05) and a reduction in the EC50 (depressive = 0.6203 ± 0.03005 μg/kg; non-depressive = 0.7320 ± 0.03519 μg/kg; p < 0.05) with no change in the other cardiovascular parameters. After treatment with ESC, a reduction of intrinsic heart rate was observed in the depressive-like rats (control: 342 ± 6 bpm; ESC: 316 ± 5 bpm; p < 0.05). In addition, ESC treatment increased the bradycardic (control: -97.81 ± 8.3 bpm; ESC: -137.1 ± 12.31 bpm; p = 0.0236; t = 2.502) during the baroreflex response, caused by an increase in cardiac parasympathetic modulation in the heart, in depressive-like rats (p < 0.001). The findings suggest that depressive-like rats showed cardiovascular changes, and that ESC treatment was able to reverse these changes, suggesting that ESC has a good safety profile for depressive patients with cardiovascular disease due to increased parasympathetic modulation.
Collapse
Affiliation(s)
- Luiz Fernando Veríssimo
- Department of Physiological Sciences, Center of Biological Sciences, State University of Londrina, Paraná, Brazil
| | | | - Viviane Batista Estrada
- Department of Physiological Sciences, Center of Biological Sciences, State University of Londrina, Paraná, Brazil
| | | | - Karoliny Coelho Andrade
- Department of Health Sciences Faculty of Medicine Federal University of Lavras (UFLA), Lavras, Minas Gerais, Brazil
| | - Amanda Monteiro Bonancea
- Department of Physiological Sciences, Center of Biological Sciences, State University of Londrina, Paraná, Brazil
| | - Natália Tavares Okano
- Department of Physiological Sciences, Center of Biological Sciences, State University of Londrina, Paraná, Brazil
| | | | - Gislaine Garcia Pelosi
- Department of Physiological Sciences, Center of Biological Sciences, State University of Londrina, Paraná, Brazil
| |
Collapse
|
2
|
Vachalova V, Kumnova F, Synova T, Anandam KY, Abad C, Karahoda R, Staud F. Metformin inhibits OCT3-mediated serotonin transport in the placenta. Biomed Pharmacother 2024; 179:117399. [PMID: 39243433 DOI: 10.1016/j.biopha.2024.117399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/19/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024] Open
Abstract
Proper fetal development requires tight regulation of serotonin concentrations within the fetoplacental unit. This homeostasis is partly maintained by the placental transporter OCT3/SLC22A3, which takes up serotonin from the fetal circulation. Metformin, an antidiabetic drug commonly used to treat gestational diabetes mellitus, was shown to inhibit OCT3. We, therefore, hypothesized that its use during pregnancy could disrupt placental serotonin homeostasis. This hypothesis was tested using three experimental model systems: primary trophoblast cells isolated from the human term placenta, fresh villous human term placenta fragments, and rat term placenta perfusions. Inhibition of serotonin transport by metformin at three concentrations (1 μM, 10 μM, and 100 μM) was assessed in all three models. The OCT3 inhibitor decynium-22 (100 μM) and paroxetine (100 μM), a dual inhibitor of SERT and OCT3, were used as controls. In primary trophoblasts, paroxetine exhibited the strongest inhibition of serotonin uptake, followed by decynium-22. Metformin showed a concentration-dependent effect, reducing serotonin uptake by up to 57 % at the highest concentration. Its inhibitory effect was less pronounced in fresh villous fragments but remained statistically significant at all concentrations. In the perfused rat placenta, metformin demonstrated a concentration-dependent effect, reducing placental serotonin uptake by 44 % at the highest concentration tested. Our findings across all experimental models show inhibition of placental OCT3 by metformin, resulting in reduced serotonin uptake by the trophoblast. This sheds light on mechanisms that may underpin metformin-mediated effects on fetal development.
Collapse
Affiliation(s)
- Veronika Vachalova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Fiona Kumnova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Tetiana Synova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Kasin Yadunandam Anandam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic.
| |
Collapse
|
3
|
Ramkumar R, Edge-Partington M, Terstege DJ, Adigun K, Ren Y, Khan NS, Rouhi N, Jamani NF, Tsutsui M, Epp JR, Sargin D. Long-Term Impact of Early-Life Stress on Serotonin Connectivity. Biol Psychiatry 2024; 96:287-299. [PMID: 38316332 DOI: 10.1016/j.biopsych.2024.01.024] [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: 08/10/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND Chronic childhood stress is a prominent risk factor for developing affective disorders, yet mechanisms underlying this association remain unclear. Maintenance of optimal serotonin (5-HT) levels during early postnatal development is critical for the maturation of brain circuits. Understanding the long-lasting effects of early-life stress (ELS) on serotonin-modulated brain connectivity is crucial to develop treatments for affective disorders arising from childhood stress. METHODS Using a mouse model of chronic developmental stress, we determined the long-lasting consequences of ELS on 5-HT circuits and behavior in females and males. Using FosTRAP mice, we cross-correlated regional c-Fos density to determine brain-wide functional connectivity of the raphe nucleus. We next performed in vivo fiber photometry to establish ELS-induced deficits in 5-HT dynamics and optogenetics to stimulate 5-HT release to improve behavior. RESULTS Adult female and male mice exposed to ELS showed heightened anxiety-like behavior. ELS further enhanced susceptibility to acute stress by disrupting the brain-wide functional connectivity of the raphe nucleus and the activity of 5-HT neuron population, in conjunction with increased orbitofrontal cortex (OFC) activity and disrupted 5-HT release in medial OFC. Optogenetic stimulation of 5-HT terminals in the medial OFC elicited an anxiolytic effect in ELS mice in a sex-dependent manner. CONCLUSIONS These findings suggest a significant disruption in 5-HT-modulated brain connectivity in response to ELS, with implications for sex-dependent vulnerability. The anxiolytic effect of the raphe-medial OFC circuit stimulation has potential implications for developing targeted stimulation-based treatments for affective disorders that arise from early life adversities.
Collapse
Affiliation(s)
- Raksha Ramkumar
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Moriah Edge-Partington
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Dylan J Terstege
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kabirat Adigun
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yi Ren
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nazmus S Khan
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Nahid Rouhi
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Naila F Jamani
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Mio Tsutsui
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jonathan R Epp
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Derya Sargin
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
4
|
Chhabria K, Selvaraj S, Refuerzo J, Truong C, Cazaban CG. Investigating the association between metabolic syndrome conditions and perinatal mental illness: a national administrative claims study. BMC Pregnancy Childbirth 2024; 24:409. [PMID: 38849738 PMCID: PMC11157911 DOI: 10.1186/s12884-024-06542-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/25/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Although the association between mental disorder and metabolic syndrome as a bidirectional relationship has been demonstrated, there is little knowledge of the cumulative and individual effect of these conditions on peripartum mental health. This study aims to investigate the association between metabolic syndrome conditions (MetS-C) and maternal mental illness in the perinatal period, while exploring time to incident mental disorder diagnosis in postpartum women. METHODS This observational study identified perinatal women continuously enrolled 1 year prior to and 1 year post-delivery using Optum's de-identified Clinformatics® Data Mart Database (CDM) from 2014 to 2019 with MetS-C i.e. obesity, diabetes, high blood pressure, high triglycerides, or low HDL (1-year prior to delivery); perinatal comorbidities (9-months prior to and 4-month postpartum); and mental disorder (1-year prior to and 1-year post-delivery). Additionally, demographics and the number of days until mental disorder diagnosis were evaluated in this cohort. The analysis included descriptive statistics and multivariable logistic regression. MetS-C, perinatal comorbidities, and mental disorder were assessed using the International Classification of Diseases, Ninth, and Tenth Revision diagnosis codes. RESULTS 372,895 deliveries met inclusion/exclusion criteria. The prevalence of MetS-C was 13.43%. Multivariable logistic regression revealed prenatal prevalence (1.64, CI = 1.59-1.70) and postpartum incident (1.30, CI = 1.25-1.34) diagnosis of mental health disorder were significantly higher in those with at least one MetS-C. Further, the adjusted odds of having postpartum incident mental illness were 1.51 times higher (CI = 1.39-1.66) in those with 2 MetS-C and 2.12 times higher (CI = 1.21-4.01) in those with 3 or more MetS-C. Young women (under the age of 18 years) were more likely to have an incident mental health diagnosis as opposed to other age groups. Lastly, time from hospital discharge to incident mental disorder diagnosis revealed an average of 157 days (SD = 103 days). CONCLUSION The risk of mental disorder (both prenatal and incident) has a significant association with MetS-C. An incremental relationship between incident mental illness diagnosis and the number of MetS-C, a significant association with younger mothers along with a relatively long period of diagnosis mental illness highlights the need for more screening and treatment during pregnancy and postpartum.
Collapse
Affiliation(s)
- Karishma Chhabria
- Division of Management Policy and Community Health, Center for Healthcare Data Research, The University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA.
- Department of Public Health, Usha Kundu MD College of Health, University of West Florida 11000 University Pkwy, Pensacola, FL, 32514, USA.
| | - Sudhakar Selvaraj
- Louis Faillace, MD, Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Clinical Development, Intra-Cellular Therapies, Inc., 430 East 29th Street, New York, NY, United States
| | - Jerrie Refuerzo
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chau Truong
- Division of Management Policy and Community Health, Center for Healthcare Data Research, The University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Cecilia Ganduglia Cazaban
- Division of Management Policy and Community Health, Center for Healthcare Data Research, The University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| |
Collapse
|
5
|
Ye X, Ghosh S, Shin BC, Ganguly A, Maggiotto L, Jacobs JP, Devaskar SU. Brain serotonin and serotonin transporter expression in male and female postnatal rat offspring in response to perturbed early life dietary exposures. Front Neurosci 2024; 18:1363094. [PMID: 38576870 PMCID: PMC10991790 DOI: 10.3389/fnins.2024.1363094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/29/2024] [Indexed: 04/06/2024] Open
Abstract
Introduction Serotonin (5-HT) is critical for neurodevelopment and the serotonin transporter (SERT) modulates serotonin levels. Perturbed prenatal and postnatal dietary exposures affect the developing offspring predisposing to neurobehavioral disorders in the adult. We hypothesized that the postnatal brain 5-HT-SERT imbalance associated with gut dysbiosis forms the contributing gut-brain axis dependent mechanism responsible for such ultimate phenotypes. Methods Employing maternal diet restricted (IUGR, n=8) and high fat+high fructose (HFhf, n=6) dietary modifications, rodent brain serotonin was assessed temporally by ELISA and SERT by quantitative Western blot analysis. Simultaneously, colonic microbiome studies were performed. Results At early postnatal (P) day 2 no changes in the IUGR, but a ~24% reduction in serotonin (p = 0.00005) in the HFhf group occurred, particularly in the males (p = 0.000007) revealing a male versus female difference (p = 0.006). No such changes in SERT concentrations emerged. At late P21 the IUGR group reared on HFhf (IUGR/HFhf, (n = 4) diet revealed increased serotonin by ~53% in males (p = 0.0001) and 36% in females (p = 0.023). While only females demonstrated a ~40% decrease in serotonin (p = 0.010), the males only trended lower without a significant change within the HFhf group (p = 0.146). SERT on the other hand was no different in HFhf or IUGR/RC, with only the female IUGR/HFhf revealing a 28% decrease (p = 0.036). In colonic microbiome studies, serotonin-producing Bacteriodes increased with decreased Lactobacillus at P2, while the serotonin-producing Streptococcus species increased in IUGR/HFhf at P21. Sex-specific changes emerged in association with brain serotonin or SERT in the case of Alistipase, Anaeroplasma, Blautia, Doria, Lactococcus, Proteus, and Roseburia genera. Discussion We conclude that an imbalanced 5-HT-SERT axis during postnatal brain development is sex-specific and induced by maternal dietary modifications related to postnatal gut dysbiosis. We speculate that these early changes albeit transient may permanently alter critical neural maturational processes affecting circuitry formation, thereby perturbing the neuropsychiatric equipoise.
Collapse
Affiliation(s)
- Xin Ye
- Department of Pediatrics, Division of Neonatology & Developmental Biology and The Neonatal Research Center of the Children's Discovery & Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Shubhamoy Ghosh
- Department of Pediatrics, Division of Neonatology & Developmental Biology and The Neonatal Research Center of the Children's Discovery & Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Bo-Chul Shin
- Department of Pediatrics, Division of Neonatology & Developmental Biology and The Neonatal Research Center of the Children's Discovery & Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Amit Ganguly
- Department of Pediatrics, Division of Neonatology & Developmental Biology and The Neonatal Research Center of the Children's Discovery & Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Liesbeth Maggiotto
- Department of Pediatrics, Division of Neonatology & Developmental Biology and The Neonatal Research Center of the Children's Discovery & Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Jonathan P. Jacobs
- The Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Sherin U. Devaskar
- Department of Pediatrics, Division of Neonatology & Developmental Biology and The Neonatal Research Center of the Children's Discovery & Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| |
Collapse
|
6
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
7
|
Pyatha S, Kim H, Lee D, Kim K. Co-exposure to lead, mercury, and cadmium induces neurobehavioral impairments in mice by interfering with dopaminergic and serotonergic neurotransmission in the striatum. Front Public Health 2023; 11:1265864. [PMID: 38026429 PMCID: PMC10662100 DOI: 10.3389/fpubh.2023.1265864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Humans are exposed to lead (Pb), mercury (Hg), and cadmium (Cd) through various routes, including drinking water, and such exposure can lead to a range of toxicological effects. However, few studies have investigated the toxic effects of exposure to mixtures of metals, particularly in relation to neurotoxicity. In this study, 7-week-old male mice were exposed to Pb, Hg, and Cd individually or in combination through their drinking water for 28 days. The mice exposed to the metal mixture exhibited significantly reduced motor coordination and impaired learning and memory abilities compared to the control group and each of the single metal exposure groups, indicating a higher level of neurotoxicity of the metal mixture. The dopamine content in the striatum was significantly lower in the metal mixture exposure group than in the single metal exposure groups and the control group. Furthermore, compared to the control group, the metal mixture exposure group showed a significantly lower expression level of tyrosine hydroxylase (TH) and significantly higher expression levels of dopamine transporter (DAT), tryptophan hydroxylase 1 (TPH1), and serotonin reuptake transporter (SERT). Notably, there were no significant differences in SERT expression between the single metal exposure groups and the control group, but SERT expression was significantly higher in the metal mixture exposure group than in the single metal and control groups. These findings suggest that the key proteins involved in the synthesis and reuptake of dopamine (TH and DAT, respectively), as well as in the synthesis and reuptake of serotonin (TPH1 and SERT, respectively), play crucial roles in the neurotoxic effects associated with exposure to metal mixtures. In conclusion, this study demonstrates that simultaneous exposure to different metals can impact key enzymes involved in dopaminergic and serotonergic neurotransmission processes, leading to disruptions in dopamine and serotonin homeostasis and consequently a range of detrimental neurobehavioral effects.
Collapse
Affiliation(s)
| | | | | | - Kisok Kim
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| |
Collapse
|
8
|
Zosen D, Kondratskaya E, Kaplan-Arabaci O, Haugen F, Paulsen RE. Antidepressants escitalopram and venlafaxine up-regulate BDNF promoter IV but down-regulate neurite outgrowth in differentiating SH-SY5Y neurons. Neurochem Int 2023; 169:105571. [PMID: 37451345 DOI: 10.1016/j.neuint.2023.105571] [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: 01/24/2023] [Revised: 06/26/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Antidepressants are used to treat depression and some anxiety disorders, including use in pregnant patients. The pharmacological actions of these drugs generally determine the uptake and metabolism of a series of neurotransmitters, such as serotonin, norepinephrine, or dopamine, along with an increase in BDNF expression. However, many aspects of antidepressant action remain unknown, particularly whether antidepressants interfere with normal neurodevelopment when taken by pregnant women. In order to reveal cellular and molecular implications crucial to the functioning of pathways related to antidepressant effects, we performed an investigation on neuronally differentiating human SH-SY5Y cells. To our knowledge, this is the first time human SH-SY5Y cells in cultures of purely neuronal cells induced by controlled differentiation with retinoic acid are followed by short-term 48-h exposure to 0.1-10 μM escitalopram or venlafaxine. Treatment with antidepressants (1 μM) did not affect the electrophysiological properties of SH-SY5Y cells. However, the percentage of mature neurons exhibiting voltage-gated sodium currents was substantially higher in cultures pre-treated with either antidepressant. After exposure to escitalopram or venlafaxine, we observed a concentration-dependent increase in activity-dependent BDNF promoter IV activation. The assessment of neurite metrics showed significant down-regulation of neurite outgrowth upon exposure to venlafaxine. Identified changes may represent links to molecular processes of importance to depression and be involved in neurodevelopmental alterations observed in postpartum children exposed to antidepressants antenatally.
Collapse
Affiliation(s)
- Denis Zosen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Elena Kondratskaya
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; NORMENT, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Oykum Kaplan-Arabaci
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Fred Haugen
- Department of Work Psychology and Physiology, National Institute of Occupational Health (STAMI), Oslo, Norway
| | - Ragnhild Elisabeth Paulsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
9
|
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
|
10
|
Han J, Stine JM, Chapin AA, Ghodssi R. A portable electrochemical sensing platform for serotonin detection based on surface-modified carbon fiber microelectrodes. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:1096-1104. [PMID: 36723293 DOI: 10.1039/d2ay01627c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Serotonin (5-HT) is one of the key neurotransmitters in the human body, regulating numerous physiological functions. A disruption in 5-HT homeostasis could result in serious health problems, including neurodegenerative disorders, depression, and 5-HT syndrome. Detection of 5-HT concentrations in biological fluids, such as urine, is a potential solution for early diagnosis of these diseases. In this study, we developed a novel, simple, and low-cost electrochemical sensing platform consisting of a portable workstation with customized electrodes for 5-HT detection in artificial biological fluids. Nafion/carbon nanotubes (CNTs) and electrochemically modified carbon fiber microelectrodes (Nafion-CNT/EC CFMEs) displayed improved 5-HT sensitivity and selectivity. Together with a customized Ag/AgCl reference electrode and Pt counter electrode, the portable 5-HT sensing platform had a sensitivity of 0.074 μA μM-1 and a limit of detection (LOD) of 140 nM. This system was also assessed to measure 5-HT spiked in artificial urine samples, showing nearly full recovery rates. These satisfactory results demonstrated that the portable system exhibits outstanding performance and confirmed the feasibility of 5-HT detection, which can be used to provide point-of-care analysis in actual biological samples.
Collapse
Affiliation(s)
- Jinjing Han
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD 20742, USA,.
- Institute for Systems Research, University of Maryland, College Park, MD 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, USA
| | - Justin M Stine
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD 20742, USA,.
- Institute for Systems Research, University of Maryland, College Park, MD 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, USA
| | - Ashley A Chapin
- Institute for Systems Research, University of Maryland, College Park, MD 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Reza Ghodssi
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD 20742, USA,.
- Institute for Systems Research, University of Maryland, College Park, MD 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
11
|
Bhat RS, Alonazi M, Al-Daihan S, El-Ansary A. Prenatal SSRI Exposure Increases the Risk of Autism in Rodents via Aggravated Oxidative Stress and Neurochemical Changes in the Brain. Metabolites 2023; 13:metabo13020310. [PMID: 36837929 PMCID: PMC9963091 DOI: 10.3390/metabo13020310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
The mechanisms underlying selective serotonin reuptake inhibitor (SSRI) use during pregnancy as a major autism risk factor are unclear. Here, brain neurochemical changes following fluoxetine exposure and in an autism model were compared to determine the effects on autism risk. The study was performed on neonatal male western albino rats which were divided into Groups one (control), two (propionic acid [PPA]-induced autism model), and three (prenatal SSRI-exposed newborn rats whose mothers were exposed to 5 mg/kg of fluoxetine over gestation days 10-20). SSRI (fluoxetine) induced significant neurochemical abnormalities in the rat brain by increasing lipid peroxide (MDA), Interferon-gamma (IFN-γ), and caspase-3 levels and by depleting Glutathione (GSH), Glutathione S-transferases (GST), Catalase, potassium (K+), and Creatine kinase (CK) levels, similarly to what has been discovered in the PPA model of autism when compared with control. Prenatal fluoxetine exposure plays a significant role in asset brain damage in newborns; further investigation of fluoxetine as an autism risk factor is thus warranted.
Collapse
Affiliation(s)
- Ramesa Shafi Bhat
- Biochemistry Department, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
- Correspondence:
| | - Mona Alonazi
- Biochemistry Department, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
| | - Sooad Al-Daihan
- Biochemistry Department, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
| | - Afaf El-Ansary
- Central Research Laboratory, Female Campus, King Saud University, Riyadh 11495, Saudi Arabia
| |
Collapse
|
12
|
Positive Effects of α-Lactalbumin in the Management of Symptoms of Polycystic Ovary Syndrome. Nutrients 2022; 14:nu14153220. [PMID: 35956395 PMCID: PMC9370664 DOI: 10.3390/nu14153220] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 11/18/2022] Open
Abstract
To date, the involvement of α-Lactalbumin (α-LA) in the management of polycystic ovary syndrome (PCOS) refers to its ability to improve intestinal absorption of natural molecules like inositols, overcoming the inositol resistance. However, due to its own aminoacidic building blocks, α-LA is involved in various biological processes that can open new additional applications. A great portion of women with PCOS exhibit gastrointestinal dysbiosis, which is in turn one of the triggering mechanisms of the syndrome. Due to its prebiotic effect, α-LA can recover dysbiosis, also improving the insulin resistance, obesity and intestinal inflammation frequently associated with PCOS. Further observations suggest that altered gut microbiota negatively influence mental wellbeing. Depressive mood and low serotonin levels are indeed common features of women with PCOS. Thanks to its content of tryptophan, which is the precursor of serotonin, and considering the strict link between gut and brain, using α-LA contributes to preserving mental well-being by maintaining high levels of serotonin. In addition, considering women with PCOS seeking pregnancy, both altered microbiota and serotonin levels can induce later consequences in the offspring. Therefore, a deeper knowledge of potential applications of α-LA is required to transition to preclinical and clinical studies extending its therapeutic advantages in PCOS.
Collapse
|
13
|
Saeed R, Mahmood K, Ali SB, Haleem DJ. Behavioral, Hormonal, and Serotonergic Responses to Different Restricted Feeding Schedules in Rats. Int J Tryptophan Res 2022; 15:11786469221104729. [PMID: 35757086 PMCID: PMC9218908 DOI: 10.1177/11786469221104729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 05/16/2022] [Indexed: 11/16/2022] Open
Abstract
To determine the effect of long-term restricted feeding schedules on behavior, serotonergic responses, and neuro-endocrine functions, metabolism of serotonin (5-HT) in the striatum, expression of serotonin-1A (5-HT1A) auto-receptor in the raphe nuclei and circulating levels of leptin and corticosterone were determined in female Wistar rats kept on excessive food restriction schedule. Due to a role of dietary deficiency of tryptophan (Trp) in influencing serotonergic neurotransmission, circulating levels of Trp were also determined. Estimations were done in 2 different restricted feeding models: time-restricted feeding (TRF) and diet restricted (DR). TRF animals were given access to food ad libitum only for 2 hours/day. The DR animals were given a small calculated amount of food each day. We found that chronic food restriction for 5 weeks cause a significant decrease in the body weight and produced hyperactivity in both, TRF and DR animals. Levels of Trp were declined in circulation and in the striatum. Similarly, the levels of 5-HT and its metabolite 5-hydroxyindoleacetic acid (5-HIAA) were decreased in the striatum. Also, the expression of 5-HT1A auto-receptor was declined in the raphe nuclei. These changes in 5-HT metabolism and 5-HT1A auto-receptor expression were more profound in DR animals as compare to TRF animals. Similarly, hypoleptinemia and increased corticosterone found in both models was higher in DR animals. Effect of dietary deficiency of Trp in the modulation of striatal 5-HT metabolism and its consequences on circulating leptin and corticosterone are discussed.
Collapse
Affiliation(s)
- Raheel Saeed
- Neuroscience Research Laboratory, Dr. Panjwani Center for Molecular Medicine & Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Pakistan
| | - Khalid Mahmood
- Neuroscience Research Laboratory, Dr. Panjwani Center for Molecular Medicine & Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Pakistan
| | - Sadia Basharat Ali
- Neuroscience Research Laboratory, Dr. Panjwani Center for Molecular Medicine & Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Pakistan
| | - Darakhshan Jabeen Haleem
- Neuroscience Research Laboratory, Dr. Panjwani Center for Molecular Medicine & Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Pakistan
| |
Collapse
|
14
|
Laureano-Melo R, Dos-Santos RC, da Conceição RR, de Souza JS, da Silva Almeida C, Reis LC, Marinho BG, Giannocco G, Ahmed RG, da Silva Côrtes W. Neonatal D-fenfluramine treatment promotes long-term behavioral changes in adult mice. Int J Dev Neurosci 2022; 82:486-498. [PMID: 35718760 DOI: 10.1002/jdn.10204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/27/2022] [Accepted: 06/01/2022] [Indexed: 11/10/2022] Open
Abstract
Serotonin exerts a significant role in the mammalian central nervous system embryogenesis and brain ontogeny. Therefore, we investigate the effect of neonatal treatment of d-fenfluramine (d-FEN), a serotonin (5-HT) releaser, on the behavioral expression of adult male Swiss mice. For this purpose, we divided pregnant female Swiss mice into two groups (n = 6 each and ~35 g). Their offspring were treated with d-FEN (3 mg/kg, s.c.) from postnatal days (PND) 5 to 20. At PND 21, one male puppy of each litter was euthanized; the midbrain and the hippocampus were dissected for RNA analysis. At PND 70, the male offspring underwent a behavioral assessment in the open field, elevated plus-maze, light-dark box, tail suspension, and rotarod test. The programmed animals had a decrease in 5HT1a, serotonin transporter (SERT), and brain-derived neurotrophic factor (BDNF) expression in the mesencephalic raphe region. Alternatively, there was a reduction only in the tryptophan hydroxylase (TPH2) and BDNF expression in the hippocampus. In the light-dark box test, offspring of the treated group had higher latency to light and less time on the light side than the control. Also, it was observed less time of immobility in the tail suspension test. We also observed low motor skill learning in the rotarod test. These findings suggest that programming with d-FEN during the neonatal period alters a mesencephalic and hippocampal serotonergic system, promoting anxiety, antidepressant behavior, low coordination, and motor learning in adults.
Collapse
Affiliation(s)
- Roberto Laureano-Melo
- Multicenter and Regular Graduate Program in Physiological Sciences, Department of Physiological Sciences, Institute of Biology, Universidade Federal Rural do Rio de Janeiro, Seropedica, Brazil.,Behavioral Physiopharmacology Laboratory, Barra Mansa Center University, Rio de Janeiro, Brazil
| | - Raoni Conceição Dos-Santos
- Multicenter and Regular Graduate Program in Physiological Sciences, Department of Physiological Sciences, Institute of Biology, Universidade Federal Rural do Rio de Janeiro, Seropedica, Brazil
| | - Rodrigo Rodrigues da Conceição
- Molecular and Translational Endocrinology Laboratory, Department of Medicine, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Janaina Sena de Souza
- Molecular and Translational Endocrinology Laboratory, Department of Medicine, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Claudio da Silva Almeida
- Multicenter and Regular Graduate Program in Physiological Sciences, Department of Physiological Sciences, Institute of Biology, Universidade Federal Rural do Rio de Janeiro, Seropedica, Brazil
| | - Luís Carlos Reis
- Multicenter and Regular Graduate Program in Physiological Sciences, Department of Physiological Sciences, Institute of Biology, Universidade Federal Rural do Rio de Janeiro, Seropedica, Brazil
| | - Bruno Guimarães Marinho
- Multicenter and Regular Graduate Program in Physiological Sciences, Department of Physiological Sciences, Institute of Biology, Universidade Federal Rural do Rio de Janeiro, Seropedica, Brazil
| | - Gisele Giannocco
- Molecular and Translational Endocrinology Laboratory, Department of Medicine, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Ragab Gaber Ahmed
- Division of Anatomy and Embryology, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Wellington da Silva Côrtes
- Multicenter and Regular Graduate Program in Physiological Sciences, Department of Physiological Sciences, Institute of Biology, Universidade Federal Rural do Rio de Janeiro, Seropedica, Brazil
| |
Collapse
|
15
|
Graf AV, Maslova MV, Artiukhov AV, Ksenofontov AL, Aleshin VA, Bunik VI. Acute Prenatal Hypoxia in Rats Affects Physiology and Brain Metabolism in the Offspring, Dependent on Sex and Gestational Age. Int J Mol Sci 2022; 23:2579. [PMID: 35269722 PMCID: PMC8910449 DOI: 10.3390/ijms23052579] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 01/27/2023] Open
Abstract
Hypoxia is damaging to the fetus, but the developmental impact may vary, with underlying molecular mechanisms unclear. We demonstrate the dependence of physiological and biochemical effects of acute prenatal hypoxia (APH) on sex and gestational age. Compared to control rats, APH on the 10th day of pregnancy (APH-10) increases locomotion in both the male and female offspring, additionally increasing exploratory activity and decreasing anxiety in the males. Compared to APH-10, APH on the 20th day of pregnancy (APH-20) induces less behavioral perturbations. ECG is changed similarly in all offspring only by APH-10. Sexual dimorphism in the APH outcome on behavior is also observed in the brain acetylation system and 2-oxoglutarate dehydrogenase reaction, essential for neurotransmitter metabolism. In view of the perturbed behavior, more biochemical parameters in the brains are assessed after APH-20. Of the six enzymes, APH-20 significantly decreases the malic enzyme activity in both sexes. Among 24 amino acids and dipeptides, APH-20 increases the levels of only three amino acids (Phe, Thr, and Trp) in male offspring, and of seven amino acids (Glu, Gly, Phe, Trp, Ser, Thr, Asn) and carnosine in the female offspring. Thus, a higher reactivity of the brain metabolism to APH stabilizes the behavior. The behavior and brain biochemistry demonstrate sexually dimorphic responses to APH at both gestational stages, whereas the APH effects on ECG depend on gestational age rather than sex.
Collapse
Affiliation(s)
- Anastasia V. Graf
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.V.G.); (M.V.M.)
- Department of Biokinetics, A. N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.V.A.); (A.L.K.); (V.A.A.)
| | - Maria V. Maslova
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.V.G.); (M.V.M.)
| | - Artem V. Artiukhov
- Department of Biokinetics, A. N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.V.A.); (A.L.K.); (V.A.A.)
- Department of Biochemistry, Sechenov University, 119048 Moscow, Russia
| | - Alexander L. Ksenofontov
- Department of Biokinetics, A. N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.V.A.); (A.L.K.); (V.A.A.)
| | - Vasily A. Aleshin
- Department of Biokinetics, A. N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.V.A.); (A.L.K.); (V.A.A.)
- Department of Biochemistry, Sechenov University, 119048 Moscow, Russia
| | - Victoria I. Bunik
- Department of Biokinetics, A. N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.V.A.); (A.L.K.); (V.A.A.)
- Department of Biochemistry, Sechenov University, 119048 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
16
|
Sakala K, Kasearu K, Katus U, Veidebaum T, Harro J. Association between platelet MAO activity and lifetime drug use in a longitudinal birth cohort study. Psychopharmacology (Berl) 2022; 239:327-337. [PMID: 35001146 DOI: 10.1007/s00213-021-06035-y] [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: 08/23/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022]
Abstract
RATIONALE Platelet monoamine oxidase (MAO) activity, a marker of central serotonergic capacity, has been associated with a variety of problem behaviours. However, studies on platelet MAO activity and addictive drugs have not consistently linked MAO activity with addiction or reported to predict illicit substance use initiation or frequency. OBJECTIVES Platelet MAO activity and illicit drug use was examined in a longitudinal birth cohort study. METHODS The sample included both birth cohorts (original n = 1238) of the Estonian Children Personality Behaviour and Health Study. Longitudinal association from age 15 to 25 years between platelet MAO activity and lifetime drug use was analysed by mixed-effects regression models. Differences at ages 15, 18 and 25 were analysed by t-test. Cox proportional hazard regression analysis was used to assess the association between platelet MAO activity and the age of drug use initiation. RESULTS Male subjects who reported at least one drug use event had lower platelet MAO activity compared to nonusers, both in cross-sectional and longitudinal analyses. Males with low platelet MAO activity had started to use drugs at a younger age. Moreover, in male subjects who had experimented with illicit drugs only once in lifetime, low platelet MAO activity was also associated with higher risk at a younger age. In females, platelet MAO activity was not associated with drug use. CONCLUSION In males, low platelet MAO activity is associated with drug abuse primarily owing to risk-taking at early age.
Collapse
Affiliation(s)
- Katre Sakala
- Department of Chronic Diseases, National Institute for Health Development, Hiiu 42, 11619, Tallinn, Estonia.,Institute of Family Medicine and Public Health, Faculty of Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia.,School of Natural Sciences and Health, Tallinn University, Narva Road 29, 10120, Tallinn, Estonia
| | - Kairi Kasearu
- Institute of Social Studies, Faculty of Social Sciences, University of Tartu, Lossi 36, 51003, Tartu, Estonia
| | - Urmeli Katus
- Institute of Family Medicine and Public Health, Faculty of Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Toomas Veidebaum
- Department of Chronic Diseases, National Institute for Health Development, Hiiu 42, 11619, Tallinn, Estonia
| | - Jaanus Harro
- School of Natural Sciences and Health, Tallinn University, Narva Road 29, 10120, Tallinn, Estonia. .,Chair of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Ravila 14a, 50411, Tartu, Estonia.
| |
Collapse
|
17
|
de Mendonça CR, Manhães-de-Castro R, de Santana BJRC, Olegário da Silva L, Toscano AE, Guzmán-Quevedo O, Monteiro Galindo LC. Effects of flavonols on emotional behavior and compounds of the serotonergic system: A preclinical systematic review. Eur J Pharmacol 2021; 916:174697. [PMID: 34954234 DOI: 10.1016/j.ejphar.2021.174697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 12/21/2022]
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is a neurotransmitter that regulates multiple psychophysiological functions. An imbalance of 5-HT in the brain can modulate emotional behavior such as depression and anxiety. Substances, such as flavonols, naturally found in some plants and foods have beneficial effects on psychiatric disorders, have been studied. The aim of this systematic review was to investigate the effects of flavonols on morphological, physiological, and cellular aspects of the serotonergic system as well as on some behaviors modulated by this system. Literature searches were performed in the LILACS, Web of Science, Scopus, PubMed and Sigle via Open Grey databases, from which 1725 studies were found. Using a predefined protocol registered on the CAMARADES website, 18 studies were chosen for qualitative synthesis. Internal validity was assessed using the SYRCLE's risk of bias tool. The Kappa index was also measured to analyze agreement among the reviewers. The results of this systematic review showed that flavonols have been reported to modify physiological aspects of the serotonergic system, increasing levels of serotonin and decreasing levels of its metabolite, 5-hydroxyindoleacetic acid (5-HIAA) and promoting antioxidant effects in encephalic regions. Moreover, the results showed that flavonols can also modulate of the serotonergic system, being associated with antidepressant and anxiolytic activities. Additionally, flavonols were found to not have psychostimulant effect; they can, however, reverse damage to locomotor activity.
Collapse
Affiliation(s)
| | - Raul Manhães-de-Castro
- Postgraduate Program in Nutrition, Universidade Federal de Pernambuco, 55670-901, Recife, PE, Brazil; Studies in Nutrition and Phenotypic Plasticity Unit, Brazil
| | | | - Luana Olegário da Silva
- Postgraduate Program in Nutrition, Physical Activity and Phenotypic Plasticity, Universidade Federal de Pernambuco, 55608-680, Vitória de Santo Antão, PE, Brazil
| | - Ana Elisa Toscano
- Postgraduate Program in Nutrition, Universidade Federal de Pernambuco, 55670-901, Recife, PE, Brazil; Postgraduate Program in Neuropsychiatry and Behavioral Sciences, Universidade Federal de Pernambuco, 55608-680, Recife, PE, Brazil; Studies in Nutrition and Phenotypic Plasticity Unit, Brazil
| | - Omar Guzmán-Quevedo
- Postgraduate Program in Neuropsychiatry and Behavioral Sciences, Universidade Federal de Pernambuco, 55608-680, Recife, PE, Brazil; Laboratory of Neuronutrition and Metabolic Disorders, Instituto Tecnológico Superior de Tacámbaro, 61650, Tacámbaro, Michoacán, Mexico; Centro de Investigación Biomédica de Michoacán, Instituto Mexicano Del Seguro Social, 58341, Morelia, Michoacán, Mexico
| | - Lígia Cristina Monteiro Galindo
- Postgraduate Program in Nutrition, Physical Activity and Phenotypic Plasticity, Universidade Federal de Pernambuco, 55608-680, Vitória de Santo Antão, PE, Brazil; Department of Anatomy, Universidade Federal de Pernambuco, Brazil; Studies in Nutrition and Phenotypic Plasticity Unit, Brazil.
| |
Collapse
|
18
|
van Onselen R, Downing TG. Neonatal Reserpine Administration Produces Widespread Neuronal Losses and ⍺-Synuclein Inclusions in a Rat Model. Neurotox Res 2021; 39:1762-1770. [PMID: 34727322 DOI: 10.1007/s12640-021-00434-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 10/19/2022]
Abstract
Historically, reserpine was widely used as an antihypertensive drug. However, severe motor and non-motor symptoms such as dyskinesia and depression led to the discontinuation of reserpine as a first-line treatment for hypertension. Reserpine functions by inhibiting vesicular monoamine transporter 2 (VMAT2), reducing sequestration of monoamines into synaptic vesicles. The consequent reduction in monoamines, most notably dopamine, serotonin and norepinephrine, in the central nervous system, causes well-defined symptoms such as catalepsy, hypoactivity and sedation in animals, and these motor and non-motor symptoms are well defined for reserpine treatment. However, no gross neuropathological changes in response to reserpine treatment have been reported previously in any animal model. In contrast, reducing VMAT2 expression in genetically modified VMAT2 LO mice leads to the production of ⍺-synuclein-positive aggregates and progressive nigrostriatal neuronal loss. These VMAT2 LO mice have reduced VMAT2 functionality during critical brain developmental stages and this could be the key to producing a reserpine model with matching histopathologies. The aim of this study was therefore to investigate the effect of neonatal reserpine administration on brain histology. We report here that a single dose of 5 mg kg-1 reserpine administered subcutaneously to neonatal rats on postnatal day 3 leads to widespread neuronal loss in various brain regions including the substantia nigra pars compacta, ventral tegmental area, striatum, hippocampus, locus coeruleus, amygdala and cerebral cortex, and the presence of ⍺-synuclein-positive inclusions in the substantia nigra pars compacta and the dorsal striatum within 30 days of administration.
Collapse
Affiliation(s)
- Rianita van Onselen
- Department of Microbiology and Biochemistry, Nelson Mandela University, Port Elizabeth, 6019, South Africa
| | - Tim G Downing
- Department of Microbiology and Biochemistry, Nelson Mandela University, Port Elizabeth, 6019, South Africa.
| |
Collapse
|
19
|
Courtiol E, Menezes EC, Teixeira CM. Serotonergic regulation of the dopaminergic system: Implications for reward-related functions. Neurosci Biobehav Rev 2021; 128:282-293. [PMID: 34139249 PMCID: PMC8335358 DOI: 10.1016/j.neubiorev.2021.06.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/11/2021] [Accepted: 06/10/2021] [Indexed: 12/17/2022]
Abstract
Serotonin is a critical neuromodulator involved in development and behavior. Its role in reward is however still debated. Here, we first review classical studies involving electrical stimulation protocols and pharmacological approaches. Contradictory results on the serotonergic' involvement in reward emerge from these studies. These differences might be ascribable to either the diversity of cellular types within the raphe nuclei or/and the specific projection pathways of serotonergic neurons. We continue to review more recent work, using optogenetic approaches to activate serotonergic cells in the Raphe to VTA pathway. From these studies, it appears that activation of this pathway can lead to reinforcement learning mediated through the excitation of dopaminergic neurons by serotonergic neurons co-transmitting glutamate. Finally, given the importance of serotonin during development on adult emotion, the effect of abnormal early-life levels of serotonin on the dopaminergic system will also be discussed. Understanding the interaction between the serotonergic and dopaminergic systems during development and adulthood is critical to gain insight into the specific facets of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Emmanuelle Courtiol
- Lyon Neuroscience Research Center, UMR 5292- INSERM U1028- Université Lyon 1, 69675 Bron Cedex, France
| | - Edenia C Menezes
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, United States
| | - Catia M Teixeira
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, United States; Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, United States.
| |
Collapse
|
20
|
Cunha C, Smiley JF, Chuhma N, Shah R, Bleiwas C, Menezes EC, Seal RP, Edwards RH, Rayport S, Ansorge MS, Castellanos FX, Teixeira CM. Perinatal interference with the serotonergic system affects VTA function in the adult via glutamate co-transmission. Mol Psychiatry 2021; 26:4795-4812. [PMID: 32398719 PMCID: PMC7657958 DOI: 10.1038/s41380-020-0763-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 04/07/2020] [Accepted: 04/27/2020] [Indexed: 11/29/2022]
Abstract
Serotonin and dopamine are associated with multiple psychiatric disorders. How they interact during development to affect subsequent behavior remains unknown. Knockout of the serotonin transporter or postnatal blockade with selective serotonin reuptake inhibitors (SSRIs) leads to novelty-induced exploration deficits in adulthood, potentially involving the dopamine system. Here, we show in the mouse that raphe nucleus serotonin neurons activate ventral tegmental area dopamine neurons via glutamate co-transmission and that this co-transmission is reduced in animals exposed postnatally to SSRIs. Blocking serotonin neuron glutamate co-transmission mimics this SSRI-induced hypolocomotion, while optogenetic activation of dopamine neurons reverses this hypolocomotor phenotype. Our data demonstrate that serotonin neurons modulate dopamine neuron activity via glutamate co-transmission and that this pathway is developmentally malleable, with high serotonin levels during early life reducing co-transmission, revealing the basis for the reduced novelty-induced exploration in adulthood due to postnatal SSRI exposure.
Collapse
Affiliation(s)
- Catarina Cunha
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
- Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - John F Smiley
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
- Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Nao Chuhma
- Department of Psychiatry, Columbia University and New York State Psychiatric Institute, New York, NY, 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Relish Shah
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
| | - Cynthia Bleiwas
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
| | - Edenia C Menezes
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
| | - Rebecca P Seal
- Department of Neurobiology and Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15260, USA
| | - Robert H Edwards
- Departments of Neurology and Physiology, University of California, San Francisco School of Medicine, San Francisco, CA, 94143, USA
| | - Stephen Rayport
- Department of Psychiatry, Columbia University and New York State Psychiatric Institute, New York, NY, 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Mark S Ansorge
- Department of Psychiatry, Columbia University and New York State Psychiatric Institute, New York, NY, 10032, USA
- Department of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Francisco X Castellanos
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
- Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Catia M Teixeira
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA.
- Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
21
|
Moraga-Amaro R, Díaz-Galarce R, Donoso-Ramos JP, Ugalde V, Linsambarth S, Doorduin J, de Vries EF, Ampuero E, Peña F, Pacheco R, Wyneken U, Stehberg J. Prenatal fluoxetine impairs non-hippocampal but not hippocampal memory in adult male rat offspring. Neuropharmacology 2021; 197:108751. [PMID: 34375626 DOI: 10.1016/j.neuropharm.2021.108751] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 01/07/2023]
Abstract
Fluoxetine is often prescribed to treat depression during pregnancy. Rodent studies have shown that fluoxetine exposure during early development can induce persistent changes in the emotional behavior of the offspring. However, the effects of prenatal fluoxetine on memory have not been elucidated. This study evaluates the memory of adult male offspring from rat dams orally administered with a clinically relevant dose of 0.7 mg/kg fluoxetine from 9 weeks before pregnancy to 1 week before delivery. Hippocampal-dependent (Morris Water Maze, MWM) and non-hippocampal-dependent (Novel Object Recognition, NOR) memory paradigms were assessed. Anxiety- and depressive-like symptoms were also evaluated using the Open Field Test, Tail Suspension Test and Sucrose Preference Test. Male rats exposed to fluoxetine during gestation displayed NOR memory impairments during adulthood, as well as increased anxiety- and depressive-like symptoms. In the MWM, the offspring of fluoxetine-treated dams did not show learning deficits. However, a retention impairment was found on remote memory, 15 days after the end of training. Molecular analyses showed increased expression of NMDA subunit NR2B, and a decrease in NR2A-to- NR2B ratio in the temporal cortex, but not in the hippocampus, suggesting changes in NMDA receptor composition. These results suggest that in utero exposure to fluoxetine induces detrimental effects on non-hippocampal memory and in remote retention of hippocampal-dependent memory, which is believed to be stored in the temporal cortex, possibly due to changes in cortical NMDA receptor subunit stoichiometry. The present results warrant the need for studies on potential remote memory deficits in human offspring exposed to fluoxetine in utero.
Collapse
Affiliation(s)
- Rodrigo Moraga-Amaro
- Laboratorio de Neurobiologia, Instituto de Ciencias Biomédicas, Universidad Andres Bello, Santiago, Chile; Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Raul Díaz-Galarce
- Laboratorio de Neurobiologia, Instituto de Ciencias Biomédicas, Universidad Andres Bello, Santiago, Chile
| | - Juan P Donoso-Ramos
- Laboratorio de Neurobiologia, Instituto de Ciencias Biomédicas, Universidad Andres Bello, Santiago, Chile
| | - Valentina Ugalde
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Santiago, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, 7510156, Santiago, Chile
| | - Sergio Linsambarth
- Laboratorio de Neurobiologia, Instituto de Ciencias Biomédicas, Universidad Andres Bello, Santiago, Chile
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Erik Fj de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Estibaliz Ampuero
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile (USACH), Alameda 3363, Estación Central, Santiago, 9170022, Chile
| | - Francisca Peña
- Laboratorio de Neurobiologia, Instituto de Ciencias Biomédicas, Universidad Andres Bello, Santiago, Chile
| | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Santiago, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, 7510156, Santiago, Chile
| | - Ursula Wyneken
- Laboratorio de Neurociencias, Universidad de los Andes, Chile
| | - Jimmy Stehberg
- Laboratorio de Neurobiologia, Instituto de Ciencias Biomédicas, Universidad Andres Bello, Santiago, Chile.
| |
Collapse
|
22
|
Gawlińska K, Gawliński D, Filip M, Przegaliński E. Maternal feeding patterns affect the offspring's brain: focus on serotonin 5-HT 2C and 5-HT 2A receptors. Pharmacol Rep 2021; 73:1170-1178. [PMID: 34146338 PMCID: PMC8413174 DOI: 10.1007/s43440-021-00298-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Recent studies have shown a relationship between the composition of the maternal diet and acquiring a risk of mental illnesses through changes in the offspring's brain. This study assessed the role of a modified maternal diet on the levels of serotonin (5-HT)2C and 5-HT2A receptors in the offspring brain. METHODS Wistar rat dams during gestation and lactation were maintained either on a standard (SD) or special diets: high-fat (HFD), high-carbohydrate (rich in sucrose, HCD) or mixed (MD). Offspring were weaned to SD after lactation, and at postnatal days (PNDs) 28 and 63 changes in the 5-HT2C and 5-HT2A receptor levels were evaluated in their prefrontal cortex (PFCx), nucleus accumbens (NAc), dorsal striatum (DSTR) and hippocampus (HIP). RESULTS Maternal HFD reduced the expression of 5-HT2C receptors in male rats at PND 28 in the PFCx, NAc, and DSTR but increased it at PND 63 in male animals in the NAc and DSTR. HCD induced a decrease in the expression of 5-HT2C receptors in male offspring at PND 28 but increased it in female rats at PND 63 in the PFCx. MD reduced 5-HT2C receptor expression in males at PND 28 in the PFCx and increased it in male and female offspring at PND 28 in the HIP. Moreover, maternal HFD reduced 5-HT2A receptor levels within the PFCx in adolescent male offspring. CONCLUSION Our findings indicate that a modified maternal diet induces age- and sex-specific adaptive changes mainly in 5-HT2C receptors, which may contribute to disturbances in the offspring brain.
Collapse
Affiliation(s)
- Kinga Gawlińska
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland.
| | - Dawid Gawliński
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - Małgorzata Filip
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - Edmund Przegaliński
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| |
Collapse
|
23
|
Meurer YDSR, Linhares SSG, Lima ADC, de Aquino ACQ, Brandão LEM, Nôga DA, Campelo CLDC, Lima RH, Cavalcante JDS, Engelberth RCGJ, Ribeiro AM, Silva RH. Postnatal exposure to fluoxetine led to cognitive-emotional alterations and decreased parvalbumin positive neurons in the hippocampus of juvenile Wistar rats. Int J Dev Neurosci 2021; 81:616-632. [PMID: 34196404 DOI: 10.1002/jdn.10139] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/29/2021] [Accepted: 06/24/2021] [Indexed: 12/31/2022] Open
Abstract
The exposure to selective serotonin reuptake inhibitors (SSRIs) during development results in behavioural impairment in adulthood in humans and animal models. Indeed, serotonergic overexpression in early life leads to structural and functional changes in brain circuits that control cognition and emotion. However, the effects of developmental exposure to these substances on the behaviour of adolescent rats are conflicting and remain poorly characterised. We performed a behavioural screening to investigate the effects of postnatal exposure to fluoxetine on memory and behaviours related to anxiety, anhedonia, and depression, as well we evaluate the parvalbumin expression in hippocampus of juvenile (~PND45) female and male rats. Fluoxetine (daily 20 mg/kg s.c. injections from PND7-PND21)- or vehicle-treated adolescent rats went through several behavioural tasks (from PND 38 to PND52) and were subject to transcardial perfusion and brain removal for immunohistochemical analysis (PND53). We found that postnatal exposure to fluoxetine increased anxiety- and depression-like behaviours in the open field and sucrose preference and forced swimming tests, respectively. In addition, this treatment induced working memory and short-term (but not long-term) recognition memory impairments, and reduced parvalbumin-positive interneurons in the hippocampus. In addition, the results revealed developmental sex-dependent effects of fluoxetine postnatal treatment on adolescent rats' behaviour. These outcomes indicate that affective disorders and mnemonic alterations caused by SSRIs perinatal exposure can be present at adolescence.
Collapse
Affiliation(s)
- Ywlliane da Silva Rodrigues Meurer
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil.,Memory and Cognition Studies Laboratory, Post-graduate Program of Cognitive Neuroscience and Behavior, Department of Psychology, Federal University of Paraíba, João Pessoa, Brazil.,Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Sara Sophia Guedes Linhares
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Alvaro da Costa Lima
- Memory and Cognition Studies Laboratory, Post-graduate Program of Cognitive Neuroscience and Behavior, Department of Psychology, Federal University of Paraíba, João Pessoa, Brazil
| | - Antonio Carlos Queiroz de Aquino
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | | | | | - Ramon Hypólito Lima
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, Brazil
| | - Jeferson de Souza Cavalcante
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | - Alessandra Mussi Ribeiro
- Laboratory of Neuroscience and Bioprospecting of Natural Products, Department of Biosciences, Federal University of São Paulo, Santos, Brazil
| | - Regina Helena Silva
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
24
|
Liu K, Garcia A, Park JJ, Toliver AA, Ramos L, Aizenman CD. Early Developmental Exposure to Fluoxetine and Citalopram Results in Different Neurodevelopmental Outcomes. Neuroscience 2021; 467:110-121. [PMID: 34048796 DOI: 10.1016/j.neuroscience.2021.05.023] [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: 10/10/2020] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 11/25/2022]
Abstract
Although selective serotonin reuptake inhibitors are commonly prescribed for prenatal depression, there exists controversy over adverse effects of SSRI use on fetal development. Few studies have adequately isolated outcomes due to SSRI exposure and those due to maternal psychiatric conditions. Here, we directly investigated outcomes of exposure to widely-used SSRIs Fluoxetine and Citalopram on the developing nervous system of Xenopus laevis tadpoles, using an integrative experimental approach. We exposed tadpoles to low doses of Citalopram and Fluoxetine during a critical developmental period and found that different experimental groups displayed opposing behavioral effects. While both groups showed reduced schooling behavior, the Fluoxetine group showed increased seizure susceptibility and reduced startle habituation. In contrast, Citalopram treated tadpoles had decreased seizure susceptibility and increased habituation. Both groups had abnormal dendritic morphology in the optic tectum, a brain area important for behaviors tested. Whole-cell electrophysiological recordings of tectal neurons showed no differences in synaptic function; however, tectal cells from Fluoxetine-treated tadpoles had decreased voltage gated K+ currents while cells in the Citalopram group had increased K+ currents. Both behavioral and electrophysiological findings indicate that cells and circuits in the Fluoxetine treated optic tecta are hyperexcitable, while the Citalopram group exhibits decreased excitability. Taken together, these results show that early developmental exposure to SSRIs is sufficient to induce neurodevelopmental effects, however these effects can be complex and vary depending on the SSRI. This may explain some discrepancies across human studies, and further underscores the importance of serotonergic signaling for the developing nervous system.
Collapse
Affiliation(s)
- Karine Liu
- Department of Neuroscience, Brown University, United States
| | - Alfonso Garcia
- Department of Neuroscience, Brown University, United States
| | - Jenn J Park
- Department of Neuroscience, Brown University, United States
| | | | | | | |
Collapse
|
25
|
Postnatal Fluoxetine Treatment Alters Perineuronal Net Formation and Maintenance in the Hippocampus. eNeuro 2021; 8:ENEURO.0424-20.2021. [PMID: 33622703 PMCID: PMC8046023 DOI: 10.1523/eneuro.0424-20.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 01/20/2023] Open
Abstract
Elevation of serotonin via postnatal fluoxetine (PNFlx) treatment during critical temporal windows is hypothesized to perturb the development of limbic circuits thus establishing a substratum for persistent disruption of mood-related behavior. We examined the impact of PNFlx treatment on the formation and maintenance of perineuronal nets (PNNs), extracellular matrix (ECM) structures that deposit primarily around inhibitory interneurons, and mark the closure of critical period plasticity. PNFlx treatment evoked a significant decline in PNN number, with a robust reduction in PNNs deposited around parvalbumin (PV) interneurons, within the CA1 and CA3 hippocampal subfields at postnatal day (P)21 in Sprague Dawley rat pups. While the reduction in CA1 subfield PNN number was still observed in adulthood, we observed no change in colocalization of PV-positive interneurons with PNNs in the hippocampi of adult PNFlx animals. PNFlx treatment did not alter hippocampal PV, calretinin (CalR), or Reelin-positive neuron numbers in PNFlx animals at P21 or in adulthood. We did observe a small, but significant increase in somatostatin (SST)-positive interneurons in the DG subfield of PNFlx-treated animals in adulthood. This was accompanied by altered GABA-A receptor subunit composition, increased dendritic complexity of apical dendrites of CA1 pyramidal neurons, and enhanced neuronal activation revealed by increased c-Fos-positive cell numbers within hippocampi of PNFlx-treated animals in adulthood. These results indicate that PNFlx treatment alters the formation of PNNs within the hippocampus, raising the possibility of a disruption of excitation-inhibition (E/I) balance within this key limbic brain region.
Collapse
|
26
|
Glazova NY, Manchenko DM, Volodina MA, Merchieva SA, Andreeva LA, Kudrin VS, Myasoedov NF, Levitskaya NG. Semax, synthetic ACTH(4-10) analogue, attenuates behavioural and neurochemical alterations following early-life fluvoxamine exposure in white rats. Neuropeptides 2021; 86:102114. [PMID: 33418449 DOI: 10.1016/j.npep.2020.102114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 12/20/2020] [Accepted: 12/25/2020] [Indexed: 11/17/2022]
Abstract
Selective serotonin reuptake inhibitors (SSRI) are commonly used to treat depression during pregnancy. SSRIs cross the placenta and may influence the maturation of the foetal brain. Clinical and preclinical findings suggest long-term consequences of SSRI perinatal exposure for the offspring. The mechanisms of SSRI effects on developing brain remain largely unknown and there are no directional approaches for prevention of the consequences of maternal SSRI treatment during pregnancy. The heptapeptide Semax (MEHFPGP) is a synthetic analogue of ACTH(4-10) which exerts marked nootropic and neuroprotective activities. The aim of the present study was to investigate the long-term effects of neonatal exposure to the SSRI fluvoxamine (FA) in white rats. Additionally, the study examined the potential for Semax to prevent the negative consequences of neonatal FA exposure. Rat pups received FA or vehicle injections on postnatal days 1-14, a time period equivalent to 27-40 weeks of human foetal age. After FA treatment, rats were administered with Semax or vehicle on postnatal days 15-28. During the 2nd month of life, the rats underwent behavioural testing, and monoamine levels in brain structures were measured. It was shown that neonatal FA exposure leads to the impaired emotional response to stress and novelty and delayed acquisition of food-motivated maze task in adolescent and young adult rats. Furthermore, FA exposure induced alterations in the monoamine levels in brains of 1- and 2- month-old rats. Semax administration reduced the anxiety-like behaviour, improved learning abilities and normalized the levels of brain biogenic amines impaired by the FA exposure. The results demonstrate that early-life FA exposure in rat pups produces long-term disturbances in their anxiety-related behaviour, learning abilities, and brain monoamines content. Semax exerts a favourable effect on behaviour and biogenic amine system of rats exposed to the antidepressant. Thus, peptide Semax can prevent behavioural deficits caused by altered 5-HT levels during development.
Collapse
Affiliation(s)
- Nataliya Yu Glazova
- Institute of Molecular Genetics, RAS, 2 Akademika Kurchatova square, Moscow 123182, Russia.
| | - Daria M Manchenko
- Lomonosov Moscow State University, Biological Faculty, 1-12 Leninskie gori, Moscow 119234, Russia
| | - Maria A Volodina
- Lomonosov Moscow State University, Biological Faculty, 1-12 Leninskie gori, Moscow 119234, Russia; Institute of Cognitive Neuroscience, Centre for Bioelectric Interfaces, NRU HSE, 13-4 Myasnitskaya, Moscow 109028, Russia
| | - Svetlana A Merchieva
- Lomonosov Moscow State University, Biological Faculty, 1-12 Leninskie gori, Moscow 119234, Russia
| | - Ludmila A Andreeva
- Institute of Molecular Genetics, RAS, 2 Akademika Kurchatova square, Moscow 123182, Russia
| | - Vladimir S Kudrin
- Zakusov Research Institute of Pharmacology RAMS, 8 Baltiyskaya, Moscow 125315, Russia
| | - Nikolai F Myasoedov
- Institute of Molecular Genetics, RAS, 2 Akademika Kurchatova square, Moscow 123182, Russia
| | - Natalia G Levitskaya
- Lomonosov Moscow State University, Biological Faculty, 1-12 Leninskie gori, Moscow 119234, Russia; Institute of Molecular Genetics, RAS, 2 Akademika Kurchatova square, Moscow 123182, Russia
| |
Collapse
|
27
|
Ye X, Shin BC, Baldauf C, Ganguly A, Ghosh S, Devaskar SU. Developing Brain Glucose Transporters, Serotonin, Serotonin Transporter, and Oxytocin Receptor Expression in Response to Early-Life Hypocaloric and Hypercaloric Dietary, and Air Pollutant Exposures. Dev Neurosci 2021; 43:27-42. [PMID: 33774619 DOI: 10.1159/000514709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 01/20/2021] [Indexed: 12/18/2022] Open
Abstract
Perturbed maternal diet and prenatal exposure to air pollution (AP) affect the fetal brain, predisposing to postnatal neurobehavioral disorders. Glucose transporters (GLUTs) are key in fueling neurotransmission; deficiency of the neuronal isoform GLUT3 culminates in autism spectrum disorders. Along with the different neurotransmitters, serotonin (5-HT) and oxytocin (OXT) are critical for the development of neural connectivity. Serotonin transporter (SERT) modulates synaptic 5-HT levels, while the OXT receptor (OXTR) mediates OXT action. We hypothesized that perturbed brain GLUT1/GLUT3 regulated 5-HT-SERT imbalance, which serves as a contributing factor to postnatal neuropsychiatric phenotypes, with OXT/OXTR providing a counterbalance. Employing maternal diet restriction (intrauterine growth restriction [IUGR]), high-fat (HF) dietary modifications, and prenatal exposure to simulated AP, fetal (E19) murine brain 5-HT was assessed by ELISA with SERT and OXTR being localized by immunohistochemistry and measured by quantitative Western blot analysis. IUGR with lower head weights led to a 48% reduction in male and female fetal brain GLUT3 with no change in GLUT1, when compared to age- and sex-matched controls, with no significant change in OXTR. In addition, a ∼50% (p = 0.005) decrease in 5-HT and SERT concentrations was displayed in fetal IUGR brains. In contrast, despite emergence of microcephaly, exposure to a maternal HF diet or AP caused no significant changes. We conclude that in the IUGR during fetal brain development, reduced GLUT3 is associated with an imbalanced 5-HT-SERT axis. We speculate that these early changes may set the stage for altering the 5HT-SERT neural axis with postnatal emergence of associated neurodevelopmental disorders.
Collapse
Affiliation(s)
- Xin Ye
- Department of Pediatrics, Division of Neonatology & Developmental Biology and the Neonatal Research Center of the UCLA Children's Discovery & Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Bo-Chul Shin
- Department of Pediatrics, Division of Neonatology & Developmental Biology and the Neonatal Research Center of the UCLA Children's Discovery & Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Claire Baldauf
- Department of Pediatrics, Division of Neonatology & Developmental Biology and the Neonatal Research Center of the UCLA Children's Discovery & Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Amit Ganguly
- Department of Pediatrics, Division of Neonatology & Developmental Biology and the Neonatal Research Center of the UCLA Children's Discovery & Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Shubhamoy Ghosh
- Department of Pediatrics, Division of Neonatology & Developmental Biology and the Neonatal Research Center of the UCLA Children's Discovery & Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Sherin U Devaskar
- Department of Pediatrics, Division of Neonatology & Developmental Biology and the Neonatal Research Center of the UCLA Children's Discovery & Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
28
|
Park S, Kim Y, Lee J, Lee JY, Kim H, Lee S, Oh CM. A Systems Biology Approach to Investigating the Interaction between Serotonin Synthesis by Tryptophan Hydroxylase and the Metabolic Homeostasis. Int J Mol Sci 2021; 22:ijms22052452. [PMID: 33671067 PMCID: PMC7957782 DOI: 10.3390/ijms22052452] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/11/2021] [Accepted: 02/24/2021] [Indexed: 12/19/2022] Open
Abstract
Obesity has become a global public health and economic problem. Obesity is a major risk factor for a number of complications, such as type 2 diabetes, cardiovascular disease, fatty liver disease, and cancer. Serotonin (5-hydroxytryptamine [5-HT]) is a biogenic monoamine that plays various roles in metabolic homeostasis. It is well known that central 5-HT regulates appetite and mood. Several 5-HT receptor agonists and selective serotonin receptor uptake inhibitors (SSRIs) have shown beneficial effects on appetite and mood control in clinics. Although several genetic polymorphisms related to 5-HT synthesis and its receptors are strongly associated with obesity, there is little evidence of the role of peripheral 5-HT in human metabolism. In this study, we performed a systemic analysis of transcriptome data from the Genotype-Tissue Expression (GTEX) database. We investigated the expression of 5-HT and tryptophan hydroxylase (TPH), the rate-limiting enzyme of 5-HT biosynthesis, in the human brain and peripheral tissues. We also performed differential gene expression analysis and predicted changes in metabolites by comparing gene expressions of tissues with high TPH expression to the gene expressions of tissues with low TPH expression. Our analyses provide strong evidence that serotonin plays an important role in the regulation of metabolic homeostasis in humans.
Collapse
Affiliation(s)
- Suhyeon Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Korea; (S.P.); (Y.K.); (J.L); (J.Y.L.)
| | - Yumin Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Korea; (S.P.); (Y.K.); (J.L); (J.Y.L.)
| | - Jibeom Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Korea; (S.P.); (Y.K.); (J.L); (J.Y.L.)
| | - Jeong Yun Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Korea; (S.P.); (Y.K.); (J.L); (J.Y.L.)
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea;
| | - Sunjae Lee
- Department of School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
- Correspondence: (S.L.); (C.-M.O.); Tel.: +82-10-7304-1213 (S.L.)
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Korea; (S.P.); (Y.K.); (J.L); (J.Y.L.)
- Correspondence: (S.L.); (C.-M.O.); Tel.: +82-10-7304-1213 (S.L.)
| |
Collapse
|
29
|
Tiwari P, Fanibunda SE, Kapri D, Vasaya S, Pati S, Vaidya VA. GPCR signaling: role in mediating the effects of early adversity in psychiatric disorders. FEBS J 2021; 288:2602-2621. [DOI: 10.1111/febs.15738] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/11/2021] [Accepted: 01/27/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Praachi Tiwari
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| | - Sashaina E. Fanibunda
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
- Medical Research Centre Kasturba Health Society Mumbai India
| | - Darshana Kapri
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| | - Shweta Vasaya
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| | - Sthitapranjya Pati
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| | - Vidita A. Vaidya
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| |
Collapse
|
30
|
Implication of 5-HT7 receptor in prefrontal circuit assembly and detrimental emotional effects of SSRIs during development. Neuropsychopharmacology 2020; 45:2267-2277. [PMID: 32688364 PMCID: PMC7784885 DOI: 10.1038/s41386-020-0775-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/06/2020] [Accepted: 07/09/2020] [Indexed: 01/09/2023]
Abstract
Altered development of prefrontal cortex (PFC) circuits can have long-term consequences on adult emotional behavior. Changes in serotonin homeostasis during critical periods produced by genetic or pharmacological inactivation of the serotonin transporter (SERT, or Slc6a4), have been involved in such developmental effects. In mice, selective serotonin reuptake inhibitors (SSRIs), administered during postnatal development cause exuberant synaptic connectivity of the PFC to brainstem dorsal raphe nucleus (DRN) circuits, and increase adult risk for developing anxiety and depressive symptoms. SERT is transiently expressed in the glutamate neurons of the mouse PFC, that project to the DRN. Here, we find that 5-HTR7 is transiently co-expressed with SERT by PFC neurons, and it plays a key role in the maturation of PFC-to-DRN synaptic circuits during early postnatal life. 5-HTR7-KO mice show reduced PFC-to-DRN synaptic density (as measured by array-tomography and VGLUT1/synapsin immunocytochemistry). Conversely, 5-HTR7 over-expression in the developing PFC increased PFC-to-DRN synaptic density. Long-term consequences on depressive-like and anxiogenic behaviors were observed in adults. 5-HTR7 over-expression in the developing PFC, results in depressive-like symptoms in adulthood. Importantly, the long-term depressive-like and anxiogenic effects of SSRIs (postnatal administration of fluoxetine from P2 to P14) were not observed in 5-HTR7-KO mice, and were prevented by co-administration of the selective inhibitor of 5-HTR7, SB269970. This study identifies a new role 5-HTR7 in the postnatal maturation of prefrontal descending circuits. Furthermore, it shows that 5-HTR7 in the PFC is crucially required for the detrimental emotional effects caused by SSRI exposure during early postnatal life.
Collapse
|
31
|
Béchade C, D'Andrea I, Etienne F, Verdonk F, Moutkine I, Banas SM, Kolodziejczak M, Diaz SL, Parkhurst CN, Gan WB, Maroteaux L, Roumier A. The serotonin 2B receptor is required in neonatal microglia to limit neuroinflammation and sickness behavior in adulthood. Glia 2020; 69:638-654. [PMID: 33095507 DOI: 10.1002/glia.23918] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/11/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022]
Abstract
Severe peripheral infections induce an adaptive sickness behavior and an innate immune reaction in various organs including the brain. On the long term, persistent alteration of microglia, the brain innate immune cells, is associated with an increased risk of psychiatric disorders. It is thus critical to identify genes and mechanisms controlling the intensity and duration of the neuroinflammation induced by peripheral immune challenges. We tested the hypothesis that the 5-HT2B receptor, the main serotonin receptor expressed by microglia, might represent a valuable candidate. First, we observed that Htr2b-/- mice, knock-out for the 5-HT2B receptor gene, developed, when exposed to a peripheral lipopolysaccharide (LPS) challenge, a stronger weight loss compared to wild-type mice; in addition, comparison of inflammatory markers in brain, 4 and 24 hr after LPS injection, showed that Htr2b deficiency leads to a prolonged neuroinflammation. Second, to assess the specific contribution of the microglial 5-HT2B receptor, we investigated the response to LPS of conditional knock-out mice invalidated for Htr2b in microglia only. We found that deletion of Htr2b in microglia since birth is sufficient to cause enhanced weight loss and increased neuroinflammatory response upon LPS injection at adult stage. In contrast, mice deleted for microglial Htr2b in adulthood responded normally to LPS, revealing a neonatal developmental effect. These results highlight the role of microglia in the response to a peripheral immune challenge and suggest the existence of a developmental, neonatal period, during which instruction of microglia through 5-HT2B receptors is necessary to prevent microglia overreactivity in adulthood.
Collapse
Affiliation(s)
- Catherine Béchade
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Ivana D'Andrea
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Fanny Etienne
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Franck Verdonk
- Experimental Neuropathology, Infection and Epidemiology Department, Institut Pasteur, Paris, France
| | - Imane Moutkine
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Sophie M Banas
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Marta Kolodziejczak
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Silvina L Diaz
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Christopher N Parkhurst
- Molecular Neurobiology Program, The Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, New York, New York, USA
| | - Wenbiao B Gan
- Molecular Neurobiology Program, The Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, New York, New York, USA
| | - Luc Maroteaux
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Anne Roumier
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| |
Collapse
|
32
|
Kheirouri S, Alizadeh M. Maternal excessive gestational weight gain as a risk factor for autism spectrum disorder in offspring: a systematic review. BMC Pregnancy Childbirth 2020; 20:645. [PMID: 33092575 PMCID: PMC7579946 DOI: 10.1186/s12884-020-03324-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Background Abnormal gestational weight gain (GWG) is a prenatal complication that may contribute to long-term behavioral and neurodevelopmental differences in offspring. This systematic review summarizes research on the association between maternal GWG and risk of autism spectrum disorder (ASD) in offspring. Methods Google and electronic databases, including PubMed, SCOPUS, Embase, Cochrane Library and Google Scholar, were searched for original human studies published in English through June 2020. Articles that examined the association between GWG and risk of ASD in offspring were included. Duplicate and irrelevant studies were removed; and data were obtained through critical analysis. Results Of 96 articles searched, eight studies were included in the final review. All studies (n = 7) investigating the association of maternal excessive GWG with risk of ASD in offspring indicated that high GWG was independently associated with an increased risk of ASD. Of five studies investigating the association of inadequate GWG with the risk of ASD, four indicated that low GWG was not associated with an increased risk of ASD. Of seven studies examining the association of maternal pre-pregnancy BMI or weight with the risk of ASD, five reported that maternal pre-pregnancy BMI or weight did not appear to be independently associated with risk of ASD. The GWG-ASD association is independent of maternal BMI and child’s intellectual disability, but offspring’s genetic susceptibility connection to the GWG-ASD association remains a topic of debate. Conclusions The findings suggest that maternal excessive GWG may be associated with increased risk of ASD in offspring. However, insufficient GWG does not appear to have such association.
Collapse
Affiliation(s)
- Sorayya Kheirouri
- Department of Nutrition, Tabriz University of Medical Sciences, Attar Nishabouri St., POBOX: 14711, Tabriz, IR, Postal code: 5166614711, Iran.
| | - Mohammad Alizadeh
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
33
|
Han L, Wu KLK, Kwan PY, Chua OWH, Shum DKY, Chan YS. 5-HT 1A receptor-mediated attenuation of synaptic transmission in rat medial vestibular nucleus impacts on vestibular-related motor function. J Physiol 2020; 599:253-267. [PMID: 33006159 DOI: 10.1113/jp280610] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/29/2020] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Chemogenetic activation of medial vestibular nucleus-projecting 5-HT neurons resulted in deficits in vestibular-mediated tasks, including negative geotaxis, balance beam and rota-rod tests. The 5-HT1A receptor mediates the vestibular-related behavioural effects of 5-HT in the vestibular nucleus. 5-HT1A receptor activation attenuated evoked excitatory postsynaptic currents and evoked inhibitory postsynaptic currents via a presynaptic mechanism in the vestibular nucleus. ABSTRACT While the anxiolytic effects of serotonergic neuromodulation are well studied, its role in sensorimotor coordination and postural control is unclear. In this study, we show that an increase of serotonin (5-hydroxytryptamine, 5-HT) at the medial vestibular nucleus (MVN), a brainstem centre for vestibulospinal coordination, by either direct cannula administration or chemogenetic stimulation of MVN-projecting serotonergic neurons, adversely affected performance of rats in vestibular-mediated tasks, including negative geotaxis, balance beam and rota-rod tests. Application of the 5-HT1 and 5-HT7 receptor co-agonist 8-hydroxy-2-(di-n-propylamino) tetralin recapitulated the effect of 5-HT, while co-administration of the specific 5-HT1A receptor antagonist WAY 100135 effectively abolished all 5-HT-induced behavioural deficits. This indicated that 5-HT1A receptors mediated the effects of 5-HT in the rat MVN. Using whole-cell patch-clamp recording, we demonstrated that 5-HT1A receptor activation attenuated both evoked excitatory and evoked inhibitory postsynaptic currents through a presynaptic mechanism in the rat MVN. The results thus highlight the 5-HT1A receptor as the gain controller of vestibular-related brainstem circuits for posture and balance.
Collapse
Affiliation(s)
- Lei Han
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, PR China
| | - Kenneth Lap-Kei Wu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, PR China
| | - Pui-Yi Kwan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, PR China
| | - Oscar Wing-Ho Chua
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, PR China
| | - Daisy Kwok-Yan Shum
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, PR China.,State Key Laboratory of Brain and Cognitive Science, The University of Hong Kong, Hong Kong, PR China
| | - Ying-Shing Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, PR China.,State Key Laboratory of Brain and Cognitive Science, The University of Hong Kong, Hong Kong, PR China
| |
Collapse
|
34
|
Pati S, Saba K, Salvi SS, Tiwari P, Chaudhari PR, Verma V, Mukhopadhyay S, Kapri D, Suryavanshi S, Clement JP, Patel AB, Vaidya VA. Chronic postnatal chemogenetic activation of forebrain excitatory neurons evokes persistent changes in mood behavior. eLife 2020; 9:56171. [PMID: 32955432 PMCID: PMC7652419 DOI: 10.7554/elife.56171] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 09/18/2020] [Indexed: 12/19/2022] Open
Abstract
Early adversity is a risk factor for the development of adult psychopathology. Common across multiple rodent models of early adversity is increased signaling via forebrain Gq-coupled neurotransmitter receptors. We addressed whether enhanced Gq-mediated signaling in forebrain excitatory neurons during postnatal life can evoke persistent mood-related behavioral changes. Excitatory hM3Dq DREADD-mediated chemogenetic activation of forebrain excitatory neurons during postnatal life (P2–14), but not in juvenile or adult windows, increased anxiety-, despair-, and schizophrenia-like behavior in adulthood. This was accompanied by an enhanced metabolic rate of cortical and hippocampal glutamatergic and GABAergic neurons. Furthermore, we observed reduced activity and plasticity-associated marker expression, and perturbed excitatory/inhibitory currents in the hippocampus. These results indicate that Gq-signaling-mediated activation of forebrain excitatory neurons during the critical postnatal window is sufficient to program altered mood-related behavior, as well as functional changes in forebrain glutamate and GABA systems, recapitulating aspects of the consequences of early adversity. Stress and adversity in early childhood can have long-lasting effects, predisposing people to mental illness and mood disorders in adult life. The weeks immediately before and after birth are critical for establishing key networks of neurons in the brain. Therefore, any disruption to these neural circuits during this time can be detrimental to emotional development. However, it is still unclear which cellular mechanisms cause these lasting changes in behavior. Studies in animals suggest that these long-term effects could result from abnormalities in a few signaling pathways in the brain. For example, it has been proposed that overstimulating the cells that activate circuits in the forebrain – also known as excitatory neurons – may contribute to the behavioral changes that persist into adulthood. To test this theory, Pati et al. used genetic engineering to modulate a signaling pathway in male mice, which is known to stimulate excitatory neurons in the forebrain. The experiments showed that prolonged activation of excitatory neurons in the first two weeks after birth resulted in anxious and despair-like behaviors as the animals aged. The mice also displayed discrepancies in how they responded to certain external sensory information, which is a hallmark of schizophrenia-like behavior. However, engineering the same changes in adolescent and adult mice had no effect on their mood-related behaviors. This animal study reinforces just how critical the first few weeks of life are for optimal brain development. It provides an insight into a possible mechanism of how disruption during this time could alter emotional behavior. The findings are also relevant to psychiatrists interested in the underlying causes of mental illness after early childhood adversity.
Collapse
Affiliation(s)
- Sthitapranjya Pati
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Kamal Saba
- Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Sonali S Salvi
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Praachi Tiwari
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Pratik R Chaudhari
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Vijaya Verma
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Sourish Mukhopadhyay
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Darshana Kapri
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Shital Suryavanshi
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - James P Clement
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Anant B Patel
- Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Vidita A Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
35
|
Hanswijk SI, Spoelder M, Shan L, Verheij MMM, Muilwijk OG, Li W, Liu C, Kolk SM, Homberg JR. Gestational Factors throughout Fetal Neurodevelopment: The Serotonin Link. Int J Mol Sci 2020; 21:E5850. [PMID: 32824000 PMCID: PMC7461571 DOI: 10.3390/ijms21165850] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/24/2020] [Accepted: 08/11/2020] [Indexed: 12/21/2022] Open
Abstract
Serotonin (5-HT) is a critical player in brain development and neuropsychiatric disorders. Fetal 5-HT levels can be influenced by several gestational factors, such as maternal genotype, diet, stress, medication, and immune activation. In this review, addressing both human and animal studies, we discuss how these gestational factors affect placental and fetal brain 5-HT levels, leading to changes in brain structure and function and behavior. We conclude that gestational factors are able to interact and thereby amplify or counteract each other's impact on the fetal 5-HT-ergic system. We, therefore, argue that beyond the understanding of how single gestational factors affect 5-HT-ergic brain development and behavior in offspring, it is critical to elucidate the consequences of interacting factors. Moreover, we describe how each gestational factor is able to alter the 5-HT-ergic influence on the thalamocortical- and prefrontal-limbic circuitry and the hypothalamo-pituitary-adrenocortical-axis. These alterations have been associated with risks to develop attention deficit hyperactivity disorder, autism spectrum disorders, depression, and/or anxiety. Consequently, the manipulation of gestational factors may be used to combat pregnancy-related risks for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sabrina I. Hanswijk
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Marcia Spoelder
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Ling Shan
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands;
| | - Michel M. M. Verheij
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Otto G. Muilwijk
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Weizhuo Li
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China; (W.L.); (C.L.)
| | - Chunqing Liu
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China; (W.L.); (C.L.)
| | - Sharon M. Kolk
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behavior, Radboud University, 6525 AJ Nijmegen, The Netherlands;
| | - Judith R. Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| |
Collapse
|
36
|
González-Pardo H, Arias JL, Gómez-Lázaro E, López Taboada I, Conejo NM. Sex-Specific Effects of Early Life Stress on Brain Mitochondrial Function, Monoamine Levels and Neuroinflammation. Brain Sci 2020; 10:brainsci10070447. [PMID: 32674298 PMCID: PMC7408325 DOI: 10.3390/brainsci10070447] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/05/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022] Open
Abstract
Sex differences have been reported in the susceptibility to early life stress and its neurobiological correlates in humans and experimental animals. However, most of the current research with animal models of early stress has been performed mainly in males. In the present study, prolonged maternal separation (MS) paradigm was applied as an animal model to resemble the effects of adverse early experiences in male and female rats. Regional brain mitochondrial function, monoaminergic activity, and neuroinflammation were evaluated as adults. Mitochondrial energy metabolism was greatly decreased in MS females as compared with MS males in the prefrontal cortex, dorsal hippocampus, and the nucleus accumbens shell. In addition, MS males had lower serotonin levels and increased serotonin turnover in the prefrontal cortex and the hippocampus. However, MS females showed increased dopamine turnover in the prefrontal cortex and increased norepinephrine turnover in the striatum, but decreased dopamine turnover in the hippocampus. Sex differences were also found for pro-inflammatory cytokine levels, with increased levels of TNF-α and IL-6 in the prefrontal cortex and hippocampus of MS males, and increased IL-6 levels in the striatum of MS females. These results evidence the complex sex- and brain region-specific long-term consequences of early life stress.
Collapse
Affiliation(s)
- Héctor González-Pardo
- Laboratory of Neuroscience, Department of Psychology, Institute of Neuroscience of the Principality of Asturias (INEUROPA), University of Oviedo, Plaza Feijóo, s/n E-33003 Oviedo, Spain; (H.G.-P.); (J.L.A.); (I.L.T.)
| | - Jorge L. Arias
- Laboratory of Neuroscience, Department of Psychology, Institute of Neuroscience of the Principality of Asturias (INEUROPA), University of Oviedo, Plaza Feijóo, s/n E-33003 Oviedo, Spain; (H.G.-P.); (J.L.A.); (I.L.T.)
| | - Eneritz Gómez-Lázaro
- Department of Basic Psychological Processes and their Development, Basque Country University, Avda. Tolosa 70, s/n E-20018 San Sebastian, Spain;
| | - Isabel López Taboada
- Laboratory of Neuroscience, Department of Psychology, Institute of Neuroscience of the Principality of Asturias (INEUROPA), University of Oviedo, Plaza Feijóo, s/n E-33003 Oviedo, Spain; (H.G.-P.); (J.L.A.); (I.L.T.)
| | - Nélida M. Conejo
- Laboratory of Neuroscience, Department of Psychology, Institute of Neuroscience of the Principality of Asturias (INEUROPA), University of Oviedo, Plaza Feijóo, s/n E-33003 Oviedo, Spain; (H.G.-P.); (J.L.A.); (I.L.T.)
- Correspondence:
| |
Collapse
|
37
|
Das S, Ooi FK, Cruz Corchado J, Fuller LC, Weiner JA, Prahlad V. Serotonin signaling by maternal neurons upon stress ensures progeny survival. eLife 2020; 9:e55246. [PMID: 32324136 PMCID: PMC7237211 DOI: 10.7554/elife.55246] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/22/2020] [Indexed: 01/03/2023] Open
Abstract
Germ cells are vulnerable to stress. Therefore, how organisms protect their future progeny from damage in a fluctuating environment is a fundamental question in biology. We show that in Caenorhabditis elegans, serotonin released by maternal neurons during stress ensures the viability and stress resilience of future offspring. Serotonin acts through a signal transduction pathway conserved between C. elegans and mammalian cells to enable the transcription factor HSF1 to alter chromatin in soon-to-be fertilized germ cells by recruiting the histone chaperone FACT, displacing histones, and initiating protective gene expression. Without serotonin release by maternal neurons, FACT is not recruited by HSF1 in germ cells, transcription occurs but is delayed, and progeny of stressed C. elegans mothers fail to complete development. These studies uncover a novel mechanism by which stress sensing by neurons is coupled to transcription response times of germ cells to protect future offspring.
Collapse
Affiliation(s)
- Srijit Das
- Department of Biology, Aging Mind and Brain InitiativeIowa CityUnited States
| | - Felicia K Ooi
- Department of Biology, Aging Mind and Brain InitiativeIowa CityUnited States
| | | | | | - Joshua A Weiner
- Department of BiologyIowa CityUnited States
- Iowa Neuroscience InstituteIowa CityUnited States
| | - Veena Prahlad
- Department of Biology, Aging Mind and Brain InitiativeIowa CityUnited States
- Department of BiologyIowa CityUnited States
- Iowa Neuroscience InstituteIowa CityUnited States
| |
Collapse
|
38
|
Hernández-Carballo G, Ruíz-Luna EA, López-López G, Manjarrez E, Flores-Hernández J. Changes in Serotonin Modulation of Glutamate Currents in Pyramidal Offspring Cells of Rats Treated With 5-MT during Gestation. Brain Sci 2020; 10:E221. [PMID: 32276365 PMCID: PMC7225987 DOI: 10.3390/brainsci10040221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 11/16/2022] Open
Abstract
Changes in stimuli and feeding in pregnant mothers alter the behavior of offspring. Since behavior is mediated by brain activity, it is expected that postnatal changes occur at the level of currents, receptors or soma and dendrites structure and modulation. In this work, we explore at the mechanism level the effects on Sprague-Dawley rat offspring following the administration of serotonin (5-HT) agonist 5-methoxytryptamine (5-MT). We analyzed whether 5-HT affects the glutamate-activated (IGlut) and N-methyl-D-aspartate (NMDA)-activated currents (IGlut, INMDA) in dissociated pyramidal neurons from the prefrontal cortex (PFC). For this purpose, we performed voltage-clamp experiments on pyramidal neurons from layers V-VI of the PFC of 40-day-old offspring born from 5-MT-treated mothers at the gestational days (GD) 11 to 21. We found that the pyramidal-neurons from the PFC of offspring of mothers treated with 5-MT exhibit a significant increased reduction in both the IGlut and INMDA when 5-HT was administered. Our results suggest that the concentration increase of a neuromodulator during the gestation induces changes in its modulatory action over the offspring ionic currents during the adulthood thus contributing to possible psychiatric disorders.
Collapse
Affiliation(s)
- Gustavo Hernández-Carballo
- Instituto de Fisiología Benemérita Universidad Autónoma de Puebla, Puebla C.P.72570, Mexico; (G.H.-C.); (E.A.R.-L.); (E.M.)
| | - Evelyn A. Ruíz-Luna
- Instituto de Fisiología Benemérita Universidad Autónoma de Puebla, Puebla C.P.72570, Mexico; (G.H.-C.); (E.A.R.-L.); (E.M.)
| | - Gustavo López-López
- Facultad de Ciencias Químicas Benemérita Universidad Autónoma de Puebla, Puebla C.P.72570, Mexico;
| | - Elias Manjarrez
- Instituto de Fisiología Benemérita Universidad Autónoma de Puebla, Puebla C.P.72570, Mexico; (G.H.-C.); (E.A.R.-L.); (E.M.)
| | - Jorge Flores-Hernández
- Instituto de Fisiología Benemérita Universidad Autónoma de Puebla, Puebla C.P.72570, Mexico; (G.H.-C.); (E.A.R.-L.); (E.M.)
| |
Collapse
|
39
|
Kulikova EA, Fursenko DV, Bazhenova EY, Kulikov AV. Pargyline and р-Chlorophenylalanine Decrease Expression of Ptpn5 Encoding Striatal-Enriched Protein Tyrosine Phosphatase (STEP) in the Mouse Striatum. Mol Biol 2020. [DOI: 10.1134/s0026893320020090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
40
|
Role of the Serotonin Receptor 7 in Brain Plasticity: From Development to Disease. Int J Mol Sci 2020; 21:ijms21020505. [PMID: 31941109 PMCID: PMC7013427 DOI: 10.3390/ijms21020505] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 12/18/2022] Open
Abstract
Our knowledge on the plastic functions of the serotonin (5-HT) receptor subtype 7 (5-HT7R) in the brain physiology and pathology have advanced considerably in recent years. A wealth of data show that 5-HT7R is a key player in the establishment and remodeling of neuronal cytoarchitecture during development and in the mature brain, and its dysfunction is linked to neuropsychiatric and neurodevelopmental diseases. The involvement of this receptor in synaptic plasticity is further demonstrated by data showing that its activation allows the rescue of long-term potentiation (LTP) and long-term depression (LTD) deficits in various animal models of neurodevelopmental diseases. In addition, it is becoming clear that the 5-HT7R is involved in inflammatory intestinal diseases, modulates the function of immune cells, and is likely to play a role in the gut-brain axis. In this review, we will mainly focus on recent findings on this receptor’s role in the structural and synaptic plasticity of the mammalian brain, although we will also illustrate novel aspects highlighted in gastrointestinal (GI) tract and immune system.
Collapse
|
41
|
Barenys M, Reverte I, Masjosthusmann S, Gómez-Catalán J, Fritsche E. Developmental neurotoxicity of MDMA. A systematic literature review summarized in a putative adverse outcome pathway. Neurotoxicology 2019; 78:209-241. [PMID: 31812710 DOI: 10.1016/j.neuro.2019.12.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 11/11/2019] [Accepted: 12/04/2019] [Indexed: 12/14/2022]
Abstract
The increasing use of illegal drugs by pregnant women causes a public health concern because it is associated with health risks for mothers and their developing children. One of such drugs is MDMA (3,4-methylenedioxymethamphetamine) or ecstasy due to its high consumption in relevant age and sex groups and its adverse effects on human and rodent developing brains. To thoroughly review the current knowledge on the developmentally neurotoxic potential of MDMA we systematically collected and summarized articles investigating developmental neurotoxicity (DNT) of MDMA in humans and animals in vivo and in vitro. In addition, we summarized the findings in a putative adverse outcome pathway (AOP). From an initial 299 articles retrieved from the bibliographic databases Web of Science, PubMed and DART, we selected 39 articles according to inclusion/exclusion criteria for data collection after title/abstract and full text screening. Of these 3 where epidemiological studies, 34 where in vivo studies in mice and rats and 2 were in vitro studies. The three epidemiological studies reported from the same longitudinal study and suggested that MDMA exposure during pregnancy impairs neuromotor function in infants. In rat, postnatal exposure towards MDMA also caused locomotor deficits as well as impaired spatial learning that might be associated with decreased serotonin levels in the hippocampus. In vitro MDMA caused cytotoxicity at high concentrations and effects on the serotonergic and neuritogenic alterations at lower concentrations which are in line with some of the in vivo alterations observed. Considering the adverse outcomes of developmental MDMA described in humans and in rodents we summarized the first putative AOP on developmental compound exposure leading to impaired neuromotor function in children. For generation of this AOP, MDMA exposure was taken as a model compound. In addition, we hypothesized a second AOP involving developmental disturbance of the dopaminergic system. However, further in vitro mechanistic studies are needed to understand the molecular initiating event(s) (MIE) triggering the downstream cascades and obtain consistent evidences causally linking the adverse outcome to effects at the cellular, organ and organism level.
Collapse
Affiliation(s)
- Marta Barenys
- GRET, INSA-UB and Toxicology Unit, Pharmacology, Toxicology and Therapeutical Chemistry Department, Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain.
| | - Ingrid Reverte
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
| | - Stefan Masjosthusmann
- IUF - Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Jesús Gómez-Catalán
- GRET, INSA-UB and Toxicology Unit, Pharmacology, Toxicology and Therapeutical Chemistry Department, Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain
| | - Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany; Heinrich-Heine University, 40225 Düsseldorf, Germany
| |
Collapse
|
42
|
Sargin D, Jeoung HS, Goodfellow NM, Lambe EK. Serotonin Regulation of the Prefrontal Cortex: Cognitive Relevance and the Impact of Developmental Perturbation. ACS Chem Neurosci 2019; 10:3078-3093. [PMID: 31259523 DOI: 10.1021/acschemneuro.9b00073] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The prefrontal cortex is essential for both executive function and emotional regulation. The interrelationships among these behavioral domains are increasingly recognized, as well as their sensitivity to serotonin (5-hydroxytryptamine, 5-HT). Prefrontal cortex receives serotonergic inputs from the dorsal and median raphe nuclei and is modulated by multiple subtypes of 5-HT receptor across its layers and cell types. Extremes of serotonergic modulation alter mood regulation in vulnerable individuals, yet the impact of serotonin under more typical physiological parameters remains unclear. In this regard, new tools are permitting a closer examination of the behavioral impact of the serotonin system. Optogenetic and chemogenetic manipulations of dorsal raphe 5-HT neurons reveal that serotonin has a greater impact on executive function than previously appreciated. Domains that appear sensitive to fluctuations in 5-HT neuronal excitability include patience and cognitive flexibility. This work is broadly consistent with ex vivo research investigating how 5-HT regulates prefrontal cortex and its output projections. A growing literature suggests 5-HT modulation of these prefrontal circuits is unexpectedly flexible to alteration during development by genetic, behavioral, environmental or pharmacological manipulations, with lasting repercussions for cognition and emotional regulation. Here, we review the cellular and circuit mechanisms of prefrontal serotonergic modulation, investigate recent research into the cognitive consequences of the serotonergic system, and probe the lasting consequences of developmental perturbations. Understanding both the complexity of the prefrontal serotonin system and its sensitivity during development are essential to learn more about the vulnerabilities of this system in mood and anxiety disorders and the underappreciated cognitive consequences of these disorders and their treatment.
Collapse
Affiliation(s)
- Derya Sargin
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary AB T2N 1N4, Canada
| | - Ha-Seul Jeoung
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | - Evelyn K. Lambe
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of OBGYN, University of Toronto, Toronto, ON M5G 1E2, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
| |
Collapse
|
43
|
Fatima Z, Zahra A, Ghouse M, Wang X, Yuan Z. Maternal SSRIs experience and risk of ASD in offspring: a review. Toxicol Res (Camb) 2018; 7:1020-1028. [PMID: 30510676 PMCID: PMC6220718 DOI: 10.1039/c8tx00102b] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/09/2018] [Indexed: 12/20/2022] Open
Abstract
Antidepressants are extensively used during pregnancy and associated with severe outcomes, including innate malformations, prematurity, and low birth weight, etc. A recent study suggested that prenatal exposure to antidepressants may impair child neurodevelopment process. Thus, the aim of this review is to investigate the potential association between prenatal use of selective 5-HT reuptake inhibitors (SSRIs) and the risk of autism spectrum disorders (ASDs). Twelve studies related to the linkage between SSRI exposure during pregnancy and ASD in children were explored and compiled. However, there is a knowledge gap concerning the potential link between gestational exposure to antidepressants and the risk of ASDs. Despite such limitations, the available data show that some signal exists and signifies that antenatal exposure to SSRIs may increase the risk of ASDs. Thus, there is a vital need for further, large and well-designed research to definitively evaluate the existence and the magnitude of this severe risk.
Collapse
Affiliation(s)
- Zainab Fatima
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues , Huazhong Agricultural University (HZAU) , Wuhan , China . ; ; ; Fax: +86-27-87672232 ; Tel: +86-27-87287186 ; Tel: +86-27-87287186
| | - Aqeela Zahra
- School of Life Sciences , South China Normal University , China
- School of Psychology and Brain Science Institute , South China Normal University , Guangzhou , 510631
| | - Maria Ghouse
- School of Life Sciences , South China Normal University , China
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues , Huazhong Agricultural University (HZAU) , Wuhan , China . ; ; ; Fax: +86-27-87672232 ; Tel: +86-27-87287186 ; Tel: +86-27-87287186
| | - Zonghui Yuan
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues , Huazhong Agricultural University (HZAU) , Wuhan , China . ; ; ; Fax: +86-27-87672232 ; Tel: +86-27-87287186 ; Tel: +86-27-87287186
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products , Wuhan , China
| |
Collapse
|