1
|
Xie X, Li W, Xiong Z, Xu J, Liao T, Sun L, Xu H, Zhang M, Zhou J, Xiong W, Fu Z, Li Z, Han Q, Cui D, Anthony DC. Metformin reprograms tryptophan metabolism via gut microbiome-derived bile acid metabolites to ameliorate depression-Like behaviors in mice. Brain Behav Immun 2025; 123:442-455. [PMID: 39303815 DOI: 10.1016/j.bbi.2024.09.014] [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: 10/27/2023] [Revised: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024] Open
Abstract
As an adjunct therapy, metformin enhances the efficacy of conventional antidepressant medications. However, its mode of action remains unclear. Here, metformin was found to ameliorate depression-like behaviors in mice exposed to chronic restraint stress (CRS) by normalizing the dysbiotic gut microbiome. Fecal transplants from metformin-treated mice ameliorated depressive behaviors in stressed mice. Microbiome profiling revealed that Akkermansia muciniphila (A. muciniphila), in particular, was markedly increased in the gut by metformin and that oral administration of this species alone was sufficient to reverse CRS-induced depressive behaviors and normalize aberrant stress-induced 5-hydroxytryptamine (5-HT) metabolism in the brain and gut. Untargeted metabolomic profiling further identified the bile acid metabolites taurocholate and deoxycholic acid as direct A. muciniphila-derived molecules that are, individually, sufficient to rescue the CRS-induced impaired 5-HT metabolism and depression-like behaviors. Thus, we report metformin reprograms 5-HT metabolism via microbiome-brain interactions to mitigate depressive syndromes, providing novel insights into gut microbiota-derived bile acids as potential therapeutic candidates for depressive mood disorders from bench to bedside.
Collapse
Affiliation(s)
- Xiaoxian Xie
- Shanghai Mental Health Center, Shanghai Jiao Tong University, School of Medicine, Shanghai 201109, PR China; Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT Oxford, UK; College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Wenwen Li
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Ze Xiong
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Junyu Xu
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, PR China
| | - Tailin Liao
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, PR China
| | - Lei Sun
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Haoshen Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Mengya Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Jiafeng Zhou
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Wenzheng Xiong
- Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT Oxford, UK
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Zezhi Li
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou 510370, PR China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, PR China.
| | - Qi Han
- Center for Brain Science Shanghai Children s Medical Center, Department of Anatomy and Physiology, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Disease, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China; Shanghai Center for Brain Science and Brain-inspired Technology, Shanghai 200031, PR China.
| | - Donghong Cui
- Shanghai Mental Health Center, Shanghai Jiao Tong University, School of Medicine, Shanghai 201109, PR China.
| | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT Oxford, UK
| |
Collapse
|
2
|
Zhang X, Luo L, Wang C, Lv W, Duan Y, Kong L. Research progress on Chaihu Shugan San in treating perimenopausal syndrome: A review. Medicine (Baltimore) 2024; 103:e41044. [PMID: 39969354 PMCID: PMC11688091 DOI: 10.1097/md.0000000000041044] [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: 08/10/2024] [Accepted: 12/04/2024] [Indexed: 02/20/2025] Open
Abstract
Perimenopausal syndrome (PMS) refers to a series of physical and psychological symptoms caused by fluctuating or decreasing sexual hormone levels during the pre- and postmenopausal periods. With the rapid development of society, more and more women suffer from menstrual disorders and insomnia caused by PMS. Chaihu Shugan San (CSS) is a famous traditional Chinese medicine prescription known for soothing the liver, relieving depression, and regulating qi and blood. Numerous clinical experiments and pharmacological studies have confirmed that CSS has a significant effect on PMS treatment. However, the composition of CSS is complex, its pharmacological effects are diverse, and its therapeutic mechanism for PMS has not been clearly explained. Therefore, this article reviews the classical literature, mechanism, pharmacological effects and clinical research of CSS in the treatment of PMS, so as to provide a reference for clinical application and further research.
Collapse
Affiliation(s)
- Xiaomeng Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Lele Luo
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Chenchen Wang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Wenjing Lv
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Yuanfei Duan
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Lingyuan Kong
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| |
Collapse
|
3
|
Mata-Bermudez A, Diaz-Ruiz A, Silva-García LR, Gines-Francisco EM, Noriega-Navarro R, Rios C, Romero-Sánchez HA, Arroyo D, Landa A, Navarro L. Mucuna pruriens, a Possible Treatment for Depressive Disorders. Neurol Int 2024; 16:1509-1527. [PMID: 39585071 PMCID: PMC11587415 DOI: 10.3390/neurolint16060112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024] Open
Abstract
Depression is a mental disorder that depicts a wide variety of symptoms, including mood and cognitive alterations, as well as recurrent thoughts of death or suicide. It could become the second leading cause of premature death or disability worldwide. Treatments with conventional antidepressants have several limitations in terms of effectiveness, side effects, and high costs. Therefore, medicinal plants such as Mucuna pruriens are potent candidates for treating depressive disorders. This review shows a compendium of evidence supporting the antidepressant effect of the Mucuna pruriens plant in diverse animal models. This includes the mechanisms of action underlying the antidepressant activity of the treatment concerning dopamine, serotonin, norepinephrine, reactive oxygen species, nitric oxide, cortisol, and inflammation. Clinical trials are needed to study the efficacy and safety of Mucuna pruriens for depression.
Collapse
Affiliation(s)
- Alfonso Mata-Bermudez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (A.M.-B.); (R.N.-N.); (D.A.)
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México 04960, Mexico; (L.R.S.-G.); (E.M.G.-F.); (H.A.R.-S.)
| | - Araceli Diaz-Ruiz
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México 14269, Mexico;
| | - Luis Ricardo Silva-García
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México 04960, Mexico; (L.R.S.-G.); (E.M.G.-F.); (H.A.R.-S.)
| | - Eduardo Manuel Gines-Francisco
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México 04960, Mexico; (L.R.S.-G.); (E.M.G.-F.); (H.A.R.-S.)
| | - Roxana Noriega-Navarro
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (A.M.-B.); (R.N.-N.); (D.A.)
| | - Camilo Rios
- Laboratorio de Neurofarmacología Molecular, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México 04960, Mexico;
- Dirección de Investigación, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra, Calzada Mexico-Xochimilco 289, Arenal de Guadalupe, Ciudad de México 14389, Mexico
| | - Héctor Alonso Romero-Sánchez
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México 04960, Mexico; (L.R.S.-G.); (E.M.G.-F.); (H.A.R.-S.)
| | - Diego Arroyo
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (A.M.-B.); (R.N.-N.); (D.A.)
| | - Abraham Landa
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Luz Navarro
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (A.M.-B.); (R.N.-N.); (D.A.)
| |
Collapse
|
4
|
Warner AK, Iskander L, Allen K, Quatela I, Borrelli H, Sachs BD. The effects of brain serotonin deficiency on the behavioral and neurogenesis-promoting effects of voluntary exercise in tryptophan hydroxylase 2 (R439H) knock-in mice. Neuropharmacology 2024; 258:110082. [PMID: 39009217 DOI: 10.1016/j.neuropharm.2024.110082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024]
Abstract
Exercise is known to reduce depression and anxiety symptoms. Although the cellular and molecular mechanisms underlying this effect remain unknown, exercise-induced increases in neurotransmitter release and hippocampal neurogenesis have been hypothesized to play key roles. One neurotransmitter that has been implicated in both antidepressant-like effects and the regulation of hippocampal neurogenesis is serotonin (5-HT). Complete loss of function of the brain 5-HT synthesis enzyme (tryptophan hydroxylase 2, Tph2) has been reported to prevent exercise-induced increases in neurogenesis and to block a subset of antidepressant-like responses to selective serotonin reuptake inhibitors (SSRIs), but whether partial loss of Tph2 function blocks the behavioral and neurogenic effects of exercise has not been established. This study used four tests that are predictive of antidepressant efficacy to determine the impact of 5-HT deficiency on responses to exercise in male and female mice. Our results demonstrate that low 5-HT impairs the behavioral effects of exercise in females in the forced swim and novelty-suppressed feeding tests. However, genetic reductions in 5-HT synthesis did not significantly impact exercise-induced alterations in cellular proliferation or immature neuron production in the hippocampus in either sex. These findings highlight the importance of brain 5-HT in mediating behavioral responses to exercise and suggest that individual differences in brain 5-HT synthesis could influence sensitivity to the mental health benefits of exercise. Furthermore, the observed disconnect between neurogenic and behavioral responses to exercise suggests that increased neurogenesis is unlikely to be the primary driver of the behavioral effects of exercise observed here.
Collapse
Affiliation(s)
- Allison K Warner
- Department of Psychological and Brain Sciences, Villanova University, USA
| | - Lauren Iskander
- Department of Psychological and Brain Sciences, Villanova University, USA
| | - Kristen Allen
- Department of Psychological and Brain Sciences, Villanova University, USA
| | - Isabella Quatela
- Department of Psychological and Brain Sciences, Villanova University, USA
| | - Hannah Borrelli
- Department of Psychological and Brain Sciences, Villanova University, USA
| | - Benjamin D Sachs
- Department of Psychological and Brain Sciences, Villanova University, USA.
| |
Collapse
|
5
|
Zhu HM, Wang B, Wang T, Shao J, Chen HR, Zhang C, Xu LH, Li JJ, Wang M, Xu DX, Meng XH. Prenatal exposure to fenvalerate causes depressive-like behavior in adulthood by inhibiting brain-derived 5-HT synthesis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 352:124137. [PMID: 38740245 DOI: 10.1016/j.envpol.2024.124137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/23/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
The developmental toxicity of fenvalerate, a representative pyrethroid insecticide, is well documented. The present study aimed to explore whether prenatal exposure to fenvalerate causes depression-like behavior in adulthood. Pregnant mice were orally administrated with either corn oil or fenvalerate (2 or 20 mg/kg) during pregnancy. Depressive-like behaviors were assessed by tail suspension test (TST), forced swim test (FST) and sucrose preference test (SPT). Immobility times in TST and FST were increased in offspring whose mothers were exposed to fenvalerate throughout pregnancy. By contrast, sugar preference index, as determined by SPT, was decreased in fenvalerate-exposed offspring. Prefrontal PSD95, a postsynaptic membrane marker, was downregulated in fenvalerate-exposed adulthood offspring. Fenvalerate-induced reduction of prefrontal PSD95 began at GD18 fetal period. Accordingly, prefrontal 5-HT, a neurotransmitter for synaptogenesis, was also reduced in fenvalerate-exposed GD18 fetuses. Tryptophan hydroxylase 2 (TPH2), a key enzyme for 5-HT synthesis, was downregulated in the midbrain of fenvalerate-exposed GD18 fetuses. Additional experiment showed that GRP78 and p-eIF2α, two endoplasmic reticulum stress-related proteins, were increased in the midbrain of fenvalerate-exposed fetal mice. The present results suggest that prenatal exposure to fenvalerate causes depressive-like behavior in adulthood, partially by inhibiting brain-derived 5-HT synthesis.
Collapse
Affiliation(s)
- Hui-Min Zhu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Bo Wang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Tao Wang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jing Shao
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Hui-Ru Chen
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Chi Zhang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Li-Hua Xu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jing-Jing Li
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Min Wang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - De-Xiang Xu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Xiu-Hong Meng
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
6
|
Kitagawa Y, Maloney SK, Pool KR, Webster D, Ohkura S, Blache D, Ding L. Behavioural and physiological responses to stressors in sheep with temperament classified by genotype or phenotype. Sci Rep 2024; 14:8147. [PMID: 38584170 PMCID: PMC10999442 DOI: 10.1038/s41598-024-58959-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/04/2024] [Indexed: 04/09/2024] Open
Abstract
The single nucleotide polymorphism (SNP) rs107856856, located in the tryptophan hydroxylase-2 gene, is associated with the behavioural phenotype for sheep temperament measured at weaning. Here, we tested the association between that SNP and physiological and behavioural responses to stressors in adult sheep. Two groups of adult sheep, one with genotype A/A (calm genotype) and the other with G/G (nervous genotype) in rs107856856, were selected from 160 sheep and were exposed, twice, to an open-field arena and an isolation box test (IBT). During each repeat, the behaviour and physiological responses (cortisol, prolactin, dehydroepiandrosterone [DHEA], brain derived neurotrophic factor [BDNF], characteristics of the response of body temperature, and oxidative stress) were measured. The behavioural and physiological responses of the sheep were compared between genotypes and also between groups classified on their phenotype as assessed by their initial isolation box score ("low responders" and "high responders"). The SNP rs107856856 had some effects on the behavioural phenotype (IBT score) but no effects on the physiological response to stress (cortisol, prolactin, DHEA, BDNF, oxidative stress or changes in body temperature) in the adult sheep, probably because the sheep were exposed, and therefore had adapted, to human contact during their life.
Collapse
Affiliation(s)
- Yuri Kitagawa
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, Japan
- School of Agriculture and Environment, M087, The University of Western Australia, 35 Stirling Highway, Perth, WA, 6009, Australia
| | - Shane K Maloney
- School of Human Sciences, M309, The University of Western Australia, 35 Stirling Highway, Perth, WA, 6009, Australia
| | - Kelsey R Pool
- School of Agriculture and Environment, M087, The University of Western Australia, 35 Stirling Highway, Perth, WA, 6009, Australia
| | - Dane Webster
- School of Human Sciences, M309, The University of Western Australia, 35 Stirling Highway, Perth, WA, 6009, Australia
| | - Satoshi Ohkura
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, Japan
| | - Dominique Blache
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
- School of Agriculture and Environment, M087, The University of Western Australia, 35 Stirling Highway, Perth, WA, 6009, Australia.
| | - Luoyang Ding
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
- School of Agriculture and Environment, M087, The University of Western Australia, 35 Stirling Highway, Perth, WA, 6009, Australia.
| |
Collapse
|
7
|
Zhan Q, Kong F, Shao S, Zhang B, Huang S. Pathogenesis of Depression in Alzheimer's Disease. Neurochem Res 2024; 49:548-556. [PMID: 38015411 DOI: 10.1007/s11064-023-04061-0] [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: 08/31/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 11/29/2023]
Abstract
Depression is a prevalent occurrence among Alzheimer's disease (AD) patients, yet its underlying mechanism remains unclear. Recent investigations have revealed that several pathophysiological changes associated with Alzheimer's disease can lead to mood disorders. These alterations include irregularities in monoamine neurotransmitters, disruptions in glutamatergic synaptic transmission, neuro-inflammation, dysfunction within the hypothalamic-pituitary-adrenocortical (HPA) axis, diminished levels of brain-derived neurotrophic factor (BDNF), and hippocampal atrophy. This review consolidates research findings from pertinent fields to elucidate the mechanisms underlying depression in Alzheimer's disease, aiming to provide valuable insights for the study of its mechanisms and clinical treatment.
Collapse
Affiliation(s)
- Qingyang Zhan
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Fanyi Kong
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Shuai Shao
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Bo Zhang
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Shuming Huang
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| |
Collapse
|
8
|
Pan YD, Zhang Y, Zheng WY, Zhu MZ, Li HY, Ouyang WJ, Wen QQ, Zhu XH. Intermittent Hypobaric Hypoxia Ameliorates Autistic-Like Phenotypes in Mice. J Neurosci 2024; 44:e1665232023. [PMID: 38124211 PMCID: PMC10869151 DOI: 10.1523/jneurosci.1665-23.2023] [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: 09/04/2023] [Revised: 11/27/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by persistent deficits in social communication and stereotyped behaviors. Although major advances in basic research on autism have been achieved in the past decade, and behavioral interventions can mitigate the difficulties that individuals with autism experience, little is known about the many fundamental issues of the interventions, and no specific medication has demonstrated efficiency for the core symptoms of ASD. Intermittent hypobaric hypoxia (IHH) is characterized by repeated exposure to lowered atmospheric pressure and oxygen levels, which triggers multiple physiological adaptations in the body. Here, using two mouse models of ASD, male Shank3B -/- and Fmr1 -/y mice, we found that IHH training at an altitude of 5,000 m for 4 h per day, for 14 consecutive days, ameliorated autistic-like behaviors. Moreover, IHH training enhanced hypoxia inducible factor (HIF) 1α in the dorsal raphe nucleus (DRN) and activated the DRN serotonergic neurons. Infusion of cobalt chloride into the DRN, to mimic IHH in increasing HIF1α expression or genetically knockdown PHD2 to upregulate HIF1α expression in the DRN serotonergic neurons, alleviated autistic-like behaviors in Shank3B -/- mice. In contrast, downregulation of HIF1α in DRN serotonergic neurons induced compulsive behaviors. Furthermore, upregulating HIF1α in DRN serotonergic neurons increased the firing rates of these neurons, whereas downregulation of HIF1α in DRN serotonergic neurons decreased their firing rates. These findings suggest that IHH activated DRN serotonergic neurons via upregulation of HIF1α, and thus ameliorated autistic-like phenotypes, providing a novel therapeutic option for ASD.
Collapse
Affiliation(s)
- Yi-da Pan
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| | - Yuan Zhang
- School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Wen-Ying Zheng
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
- School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Min-Zhen Zhu
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| | - Huan-Yu Li
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| | - Wen-Jie Ouyang
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
- School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Qin-Qing Wen
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Xin-Hong Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
- School of Psychology, Shenzhen University, Shenzhen 518060, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
9
|
Chen N, Zhao M, Guo Y, Wu N, Cao B, Zhan B, Zhou T, Li Y, Zhu F, Chen W, Li Y, Zhang L. D-mannose is a rapid inducer of ACSS2 to trigger rapid and long-lasting antidepressant responses through augmenting BDNF and TPH2 levels. Transl Psychiatry 2023; 13:338. [PMID: 37914710 PMCID: PMC10620401 DOI: 10.1038/s41398-023-02636-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/03/2023] Open
Abstract
The potentiation of synaptic plasticity and serotonin generation by brain-derived neurotrophic factor (BDNF) and tryptophan hydroxylase 2 (TPH2) is well characterized to facilitate rapid and long-lasting antidepressant actions. Therefore, the identification of the key protein that simultaneously controls both BDNF and TPH2 is important for the treatment of depression. We show here that a lack of acetyl-CoA synthetase short-chain family member 2 (ACSS2) causes impairments in BDNF-dependent synaptic plasticity and tryptophan hydroxylase 2 (TPH2)-mediated serotonin generation, thereby contributing to spontaneous and chronic restraint stress (CRS)-induced depressive-like behavior in mice. Conversely, D-mannose is identified as a rapid ACSS2 inducer and thus mediates rapid and long-lasting antidepressant-like effects. Mechanistically, acute and chronic D-mannose administration inhibits the phosphorylation of EF2 to increase BDNF levels and reverse the reduction of TPH2 histone acetylation and transcription. We reveal that ACSS2 promotes TPH2 histone acetylation and transcription with the requirement of AMPK activation. To elevate nuclear ACSS2 levels, D-mannose can rapidly and persistently activate AMPK via Ca2+-CAMKK2 and the lysosomal AXIN-LKB1 pathway to facilitate its fast-acting and persistent antidepressant responses. Taken together, the results presented here reveal that ACSS2 functions as a novel target to link rapid and persistent antidepressant actions and further suggest that D-mannose is a potential therapeutic agent to resist depression through its augmentation of the ACSS2 dependent BDNF and TPH2 pathways.
Collapse
Affiliation(s)
- Nuo Chen
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ming Zhao
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yaxin Guo
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Nan Wu
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Baihui Cao
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bing Zhan
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tian Zhou
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yubin Li
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Faliang Zhu
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - WanJun Chen
- Mucosal Immunology Section, NIDCR, US National Institutes of Health, Bethesda, MD, USA.
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Lining Zhang
- Department of Immunology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
10
|
Orrico-Sanchez A, Guiard BP, Manta S, Callebert J, Launay JM, Louis F, Paccard A, Gruszczynski C, Betancur C, Vialou V, Gautron S. Organic cation transporter 2 contributes to SSRI antidepressant efficacy by controlling tryptophan availability in the brain. Transl Psychiatry 2023; 13:302. [PMID: 37775532 PMCID: PMC10542329 DOI: 10.1038/s41398-023-02596-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 10/01/2023] Open
Abstract
Selective serotonin reuptake inhibitors (SSRI) are common first-line treatments for major depression. However, a significant number of depressed patients do not respond adequately to these pharmacological treatments. In the present preclinical study, we demonstrate that organic cation transporter 2 (OCT2), an atypical monoamine transporter, contributes to the effects of SSRI by regulating the routing of the essential amino acid tryptophan to the brain. Contrarily to wild-type mice, OCT2-invalidated mice failed to respond to prolonged fluoxetine treatment in a chronic depression model induced by corticosterone exposure recapitulating core symptoms of depression, i.e., anhedonia, social withdrawal, anxiety, and memory impairment. After corticosterone and fluoxetine treatment, the levels of tryptophan and its metabolites serotonin and kynurenine were decreased in the brain of OCT2 mutant mice compared to wild-type mice and reciprocally tryptophan and kynurenine levels were increased in mutants' plasma. OCT2 was detected by immunofluorescence in several structures at the blood-cerebrospinal fluid (CSF) or brain-CSF interface. Tryptophan supplementation during fluoxetine treatment increased brain concentrations of tryptophan and, more discreetly, of 5-HT in wild-type and OCT2 mutant mice. Importantly, tryptophan supplementation improved the sensitivity to fluoxetine treatment of OCT2 mutant mice, impacting chiefly anhedonia and short-term memory. Western blot analysis showed that glycogen synthase kinase-3β (GSK3β) and mammalian/mechanistic target of rapamycin (mTOR) intracellular signaling was impaired in OCT2 mutant mice brain after corticosterone and fluoxetine treatment and, conversely, tryptophan supplementation recruited selectively the mTOR protein complex 2. This study provides the first evidence of the physiological relevance of OCT2-mediated tryptophan transport, and its biological consequences on serotonin homeostasis in the brain and SSRI efficacy.
Collapse
Affiliation(s)
| | - Bruno P Guiard
- Université Paul Sabatier, CNRS, Research Center on Animal Cognition, Toulouse, France
| | - Stella Manta
- Université Paul Sabatier, CNRS, Research Center on Animal Cognition, Toulouse, France
| | - Jacques Callebert
- Sorbonne Paris Cité, Hôpital Lariboisière, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Jean-Marie Launay
- Sorbonne Paris Cité, Hôpital Lariboisière, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Franck Louis
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France
| | - Antoine Paccard
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France
| | | | - Catalina Betancur
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France
| | - Vincent Vialou
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France.
| | - Sophie Gautron
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France.
| |
Collapse
|
11
|
Sun N, Cui WQ, Min XM, Zhang GM, Liu JZ, Wu HY. A new perspective on hippocampal synaptic plasticity and post-stroke depression. Eur J Neurosci 2023; 58:2961-2984. [PMID: 37518943 DOI: 10.1111/ejn.16093] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023]
Abstract
Post-stroke depression, a common complication after stroke, severely affects the recovery and quality of life of patients with stroke. Owing to its complex mechanisms, post-stroke depression treatment remains highly challenging. Hippocampal synaptic plasticity is one of the key factors leading to post-stroke depression; however, the precise molecular mechanisms remain unclear. Numerous studies have found that neurotrophic factors, protein kinases and neurotransmitters influence depressive behaviour by modulating hippocampal synaptic plasticity. This review further elaborates on the role of hippocampal synaptic plasticity in post-stroke depression by summarizing recent research and analysing possible molecular mechanisms. Evidence for the correlation between hippocampal mechanisms and post-stroke depression helps to better understand the pathological process of post-stroke depression and improve its treatment.
Collapse
Affiliation(s)
- Ning Sun
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wen-Qiang Cui
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-Man Min
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guang-Ming Zhang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jia-Zheng Liu
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hong-Yun Wu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
12
|
Jacobsen JPR. Serotonin and depression-an alternative interpretation of the data in Moncrieff et al. Mol Psychiatry 2023; 28:3158-3159. [PMID: 37322060 DOI: 10.1038/s41380-023-02090-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 03/16/2023] [Accepted: 04/21/2023] [Indexed: 06/17/2023]
|
13
|
Komleva PD, Alhalabi G, Izyurov AE, Khotskin NV, Kulikov AV. Effects of the Combination of the C1473G Mutation in the Tph2 Gene and Lethal Yellow Mutations in the Raly-Agouti Locus on Behavior, Brain 5-HT and Melanocortin Systems in Mice. Biomolecules 2023; 13:963. [PMID: 37371543 DOI: 10.3390/biom13060963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/26/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Tryptophan hydroxylase 2 (TPH2) is the key and rate-limited enzyme of serotonin (5-HT) synthesis in the brain. The C1473G mutation in the Tph2 gene results in a two-fold decrease in enzyme activity in the mouse brain. The lethal yellow (AY) mutation in the Raly-Agouti locus results in the overexpression of the Agouti gene in the brain and causes obesity and depressive-like behavior in mice. Herein, the possible influences of these mutations and their combination on body mass, behavior, brain 5-HT and melanocortin systems in mice of the B6-1473CC/aa. B6-1473CC/AYa, B6-1473GG/aa are investigated. B6-1473GG/AYa genotypes were studied. The 1473G and AY alleles increase the activity of TPH2 and the expression of the Agouti gene, respectively, but they do not alter 5-HT and 5-HIAA levels or the expression of the genes Tph2, Maoa, Slc6a4, Htr1a, Htr2a, Mc3r and Mc4r in the brain. The 1473G allele attenuates weight gain and depressive-like immobility in the forced swim test, while the AY allele increases body weight gain and depressive-like immobility. The combination of these alleles results in hind limb dystonia in the B6-1473GG/AYa mice. This is the first evidence for the interaction between the C1473G and AY mutations.
Collapse
Affiliation(s)
- Polyna D Komleva
- Department of Psychoneuropharmacology, Federal Research Center Institute of Cytology and Genetic Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Ghofran Alhalabi
- Department of Genetic Collections of Neural Disorders, Federal Research Center Institute of Cytology and Genetic Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Arseniy E Izyurov
- Department of Genetics of Industrial Microorganisms, Federal Research Center Institute of Cytology and Genetic Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Nikita V Khotskin
- Department of Genetic Collections of Neural Disorders, Federal Research Center Institute of Cytology and Genetic Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Alexander V Kulikov
- Department of Genetic Collections of Neural Disorders, Federal Research Center Institute of Cytology and Genetic Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| |
Collapse
|
14
|
Zhu F, Xue Y, Ji W, Li X, Ma W, Yu P, Jiang Y, Mao L. Galvanic Redox Potentiometry for Fouling-Free and Stable Serotonin Sensing in a Living Animal Brain. Angew Chem Int Ed Engl 2023; 62:e202212458. [PMID: 36688872 DOI: 10.1002/anie.202212458] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/30/2022] [Accepted: 01/23/2023] [Indexed: 01/24/2023]
Abstract
Serotonin (5-HT) is a major neurotransmitter broadly involved in many aspects of feeling and behavior. Although its electro-activity makes it a promising candidate for electrochemical sensing, the persistent generation of fouling layers on the electrode by its oxidation products presents a hurdle for reliable sensing. Here, we present a fouling-free 5-HT sensor based on galvanic redox potentiometry. The sensor efficiently minimizes electrode fouling as revealed by in situ Raman spectroscopy, ensuring a less than 3 % signal change in a 2 hour continuous experiment, whereas amperometric sensors losing 90 % within 30 min. Most importantly, the sensor is highly amenable for in vivo studies, permitting real-time 5-HT monitoring, and supporting the mechanism associated with serotonin release in brain. Our system offers an effective way for sensing different neurochemicals having significant fouling issues, thus facilitating the molecular-level understanding of brain function.
Collapse
Affiliation(s)
- Fenghui Zhu
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Yifei Xue
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Wenliang Ji
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Xin Li
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Wenjie Ma
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing, 100190, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ping Yu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing, 100190, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Jiang
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Lanqun Mao
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| |
Collapse
|
15
|
German-Ponciano LJ, Rosas-Sánchez GU, Cueto-Escobedo J, Fernández-Demeneghi R, Guillén-Ruiz G, Soria-Fregozo C, Herrera-Huerta EV, Rodríguez-Landa JF. Participation of the Serotonergic System and Brain-Derived Neurotrophic Factor in the Antidepressant-like Effect of Flavonoids. Int J Mol Sci 2022; 23:ijms231810896. [PMID: 36142808 PMCID: PMC9505567 DOI: 10.3390/ijms231810896] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/10/2022] [Accepted: 09/11/2022] [Indexed: 11/16/2022] Open
Abstract
Depressive disorders are among the most disabling diseases experienced around the world, and their incidence has significantly increased over the last few decades due to multiple environmental, social, and biological factors. The search for new pharmacological alternatives to treat depression is a global priority. In preclinical research, molecules obtained from plants, such as flavonoids, have shown promising antidepressant-like properties through several mechanisms of action that have not been fully elucidated, including crossing of the blood brain barrier (BBB). This review will focus on discussing the main findings related to the participation of the serotonergic system and brain-derived neurotrophic factor (BDNF) on the antidepressant-like effect of some flavonoids reported by behavioral, neurochemical, and molecular studies. In this sense, evidence shows that depressive individuals have low levels of serotonin and BDNF, while flavonoids can reverse it. Finally, the elucidation of the mechanism used by flavonoids to modulate serotonin and BDNF will contribute to our understanding of the neurobiological bases underlying the antidepressant-like effects produced by these natural compounds.
Collapse
Affiliation(s)
| | | | - Jonathan Cueto-Escobedo
- Departamento de Investigación Clínica y Traslacional Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa 91190, Mexico
| | | | - Gabriel Guillén-Ruiz
- Programa de Investigadoras e Investigadores por México CONACyT-Instituto de Neuroetología, Universidad Veracruzana, Xalapa 91190, Mexico
| | - César Soria-Fregozo
- Centro Universitario de Los Lagos, Universidad de Guadalajara, Lagos de Moreno 47460, Mexico
| | | | | |
Collapse
|
16
|
Brain serotonin deficiency and fluoxetine lead to sex-specific effects on binge-like food consumption in mice. Psychopharmacology (Berl) 2022; 239:2975-2984. [PMID: 35750862 DOI: 10.1007/s00213-022-06181-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/17/2022] [Indexed: 10/17/2022]
Abstract
RATIONALE Although pharmacotherapies are often effective in reducing binge eating in conditions such as bulimia nervosa and binge eating disorder, subsets of patients do not benefit sufficiently from existing treatments, and the reasons for treatment failure remain unclear. OBJECTIVES This study aimed to evaluate whether genetic reductions in brain serotonin influence binge eating and/or the ability of fluoxetine, a selective serotonin reuptake inhibitor, to reduce binge eating in mice. METHODS This study used a validated model of binge-like consumption of high-fat diet to compare binge-like food intake in control and fluoxetine-treated wild-type and serotonin-deficient mice from the tryptophan hydroxylase 2 (R439H) knock-in line. In addition, real-time PCR was used to evaluate potential genotype and sex differences in the effects of fluoxetine on gene expression in the raphe nucleus. RESULTS The results reveal that brain serotonin deficiency is sufficient to increase binge eating in males, but not females. However, while chronic fluoxetine reduced binge eating in both genotypes of males and in wild-type females, it failed to reduce binge eating in serotonin-deficient females. Transcriptional responses to chronic fluoxetine were also characterized by sex and genotype differences. CONCLUSIONS Overall, this study revealed significant sex differences in the effects of fluoxetine and brain serotonin deficiency on binge-like food intake and suggests that low brain serotonin could impact eating disorders both by promoting binge eating and by limiting the efficacy of fluoxetine to reduce binge eating.
Collapse
|
17
|
Zaniewska M, Mosienko V, Bader M, Alenina N. Tph2 Gene Expression Defines Ethanol Drinking Behavior in Mice. Cells 2022; 11:cells11050874. [PMID: 35269497 PMCID: PMC8909500 DOI: 10.3390/cells11050874] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/17/2022] [Accepted: 02/28/2022] [Indexed: 01/22/2023] Open
Abstract
Indirect evidence supports a link between disrupted serotonin (5-hydroxytryptamine; 5-HT) signaling in the brain and addictive behaviors. However, the effects of hyposerotonergia on ethanol drinking behavior are contradictory. In this study, mice deficient in tryptophan hydroxylase 2 (Tph2−/−), the rate-limiting enzyme of 5-HT synthesis in the brain, were used to assess the role of central 5-HT in alcohol drinking behavior. Life-long 5-HT depletion in these mice led to an increased ethanol consumption in comparison to wild-type animals in a two-bottle choice test. Water consumption was increased in naïve 5-HT-depleted mice. However, exposure of Tph2−/− animals to ethanol resulted in the normalization of water intake to the level of wild-type mice. Tph2 deficiency in mice did not interfere with ethanol-evoked antidepressant response in the forced swim test. Gene expression analysis in wild-type animals revealed no change in Tph2 expression in the brain of mice consuming ethanol compared to control mice drinking water. However, within the alcohol-drinking group, inter-individual differences in chronic ethanol intake correlated with Tph2 transcript levels. Taken together, central 5-HT is an important modulator of drinking behavior in mice but is not required for the antidepressant effects of ethanol.
Collapse
Affiliation(s)
- Magdalena Zaniewska
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (V.M.); (M.B.)
- Laboratory of Pharmacology and Brain Biostructure, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland
- Correspondence: (M.Z.); (N.A.); Tel.: +48-1-2662-3289 (M.Z.); +49-30-9406-3576 (N.A.)
| | - Valentina Mosienko
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (V.M.); (M.B.)
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (V.M.); (M.B.)
- Institute for Biology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
- Charité University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Potsdamer Str. 58, 10785 Berlin, Germany
| | - Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (V.M.); (M.B.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Potsdamer Str. 58, 10785 Berlin, Germany
- Correspondence: (M.Z.); (N.A.); Tel.: +48-1-2662-3289 (M.Z.); +49-30-9406-3576 (N.A.)
| |
Collapse
|
18
|
ZHOU P, XIAO H, LI Y, DONG X. Sustained hyperarousal induced by acute stress in tryptophan-hydroxylase-2 genetic deficient male mice. ACTA PSYCHOLOGICA SINICA 2022. [DOI: 10.3724/sp.j.1041.2022.00604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
19
|
Liu ZL, Wang XQ, Liu MF, Ye BJ. Meta-analysis of association between TPH2 single nucleotide poiymorphism and depression. Neurosci Biobehav Rev 2021; 134:104517. [PMID: 34979191 DOI: 10.1016/j.neubiorev.2021.104517] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/14/2021] [Accepted: 12/29/2021] [Indexed: 10/19/2022]
Abstract
Tryptophan hydroxylase 2 (TPH2) plays a crucial role in the human brain. Although the association between the TPH2 gene and depression has been suggested in previous meta-analyses, studies based on Chinese subjects are often neglected. Therefore, we included some previous studies based on Chinese subjects to explore the relationship between TPH2 polymorphisms and depression via conducting an extensive meta-analysis. We reviewed 40 research papers that included data on TPH2 gene single nucleotide polymorphisms (SNPs) from 5766 patients with depression and 5988 healthy subjects. The analysis showed an association between polymorphisms in the TPH2 gene and depression, and some results were significant in 24 studies that included Chinese Han study participants. The results of our meta-analysis showed that rs4570625, rs17110747, rs120074175, rs4290270, rs120074175, and rs4290270 may be significantly associated with depression, and that rs11178997 (A/A genotype) may be a significant risk factor for depression in the Chinese subjects. Based on the results of this study, biological experiments should be performed in the future to explore how different SNPs affect depression.
Collapse
Affiliation(s)
- Zhang-Lin Liu
- School of Psychology, Center of Mental Health Education and Research, Key Laboratory of Psychology and Cognition Science of Jiangxi, Jiangxi Normal University, China.
| | - Xin-Qiang Wang
- School of Psychology, Center of Mental Health Education and Research, Key Laboratory of Psychology and Cognition Science of Jiangxi, Jiangxi Normal University, China.
| | - Ming-Fan Liu
- School of Psychology, Center of Mental Health Education and Research, Key Laboratory of Psychology and Cognition Science of Jiangxi, Jiangxi Normal University, China.
| | - Bao-Juan Ye
- School of Psychology, Center of Mental Health Education and Research, Key Laboratory of Psychology and Cognition Science of Jiangxi, Jiangxi Normal University, China.
| |
Collapse
|
20
|
von Linstow CU, Waider J, Bergh MSS, Anzalone M, Madsen C, Nicolau AB, Wirenfeldt M, Lesch KP, Finsen B. The Combined Effects of Amyloidosis and Serotonin Deficiency by Tryptophan Hydroxylase-2 Knockout Impacts Viability of the APP/PS1 Mouse Model of Alzheimer’s Disease. J Alzheimers Dis 2021; 85:1283-1300. [DOI: 10.3233/jad-210581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background: A decline of brain serotonin (5-HT) is held responsible for the changes in mood that can be observed in Alzheimer’s disease (AD). However, 5-HT’ergic signaling is also suggested to reduce the production of pathogenic amyloid-4β (Aβ). Objective: To investigate the effect of targeted inactivation of tryptophan hydroxylase-2 (Tph2), which is essential for neuronal 5-HT synthesis, on amyloidosis in amyloid precursor protein (APP)swe/presenilin 1 (PS1) ΔE9 transgenic mice. Methods: Triple-transgenic (3xTg) APP/PS1 mice with partial (+/-) or complete Tph2 knockout (–/–) were allowed to survive until 6 months old with APP/PS1, Tph2–/–, and wildtype mice. Survival and weight were recorded. Levels of Aβ 42/40/38, soluble APPα (sAβPPα) and sAβPPβ, and cytokines were analyzed by mesoscale, neurotransmitters by mass spectrometry, and gene expression by quantitative PCR. Tph2, microglia, and Aβ were visualized histologically. Results: Tph2 inactivation in APP/PS1 mice significantly reduced viability, without impacting soluble and insoluble Aβ 42 and Aβ 40 in neocortex and hippocampus, and with only mild changes of soluble Aβ 42/Aβ 40. However, sAβPPα and sAβPPβ in hippocampus and Aβ 38 and Aβ 40 in cerebrospinal fluid were reduced. 3xTg–/–mice were devoid of Tph2 immunopositive fibers and 5-HT. Cytokines were unaffected by genotype, as were neocortical TNF, HTR2a and HTR2b mRNA levels in Tph2–/– mice. Microglia clustered around Aβ plaques regardless of genotype. Conclusion: The results suggest that Tph2 inactivation influences AβPP processing, at least in the hippocampus, although levels of Aβ are unchanged. The reduced viability of 3xTg–/–mice could indicate that 5-HT protects against the seizures that can impact the viability of APP/PS1 mice.
Collapse
Affiliation(s)
- Christian Ulrich von Linstow
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Jonas Waider
- Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Würzburg, Germany
| | - Marianne Skov-Skov Bergh
- Section for Drug Abuse Research, Department of Forensic Sciences, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
| | - Marco Anzalone
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
- BRIDGE - Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Cecilie Madsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
- BRIDGE - Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Aina Battle Nicolau
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Martin Wirenfeldt
- BRIDGE - Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
- Department of Pathology, Institute of Clinical Science, Odense University Hospital, Denmark
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Würzburg, Germany
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Neuropsychology and Psychiatry, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - Bente Finsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
- BRIDGE - Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|
21
|
Weissbach A, Pauly MG, Herzog R, Hahn L, Halmans S, Hamami F, Bolte C, Camargos S, Jeon B, Kurian MA, Opladen T, Brüggemann N, Huppertz HJ, König IR, Klein C, Lohmann K. Relationship of Genotype, Phenotype, and Treatment in Dopa-Responsive Dystonia: MDSGene Review. Mov Disord 2021; 37:237-252. [PMID: 34908184 DOI: 10.1002/mds.28874] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/26/2021] [Accepted: 11/08/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Pathogenic variants in 5 genes (GCH1, TH, PTS, SPR, and QDPR), involved in dopamine/tetrahydrobiopterin biosynthesis or recycling, have been linked to Dopa-responsive dystonia (DRD). Diagnosis and treatment are often delayed due to high between- and within-group variability. OBJECTIVES Comprehensively analyzed individual genotype, phenotype, treatment response, and biochemistry information. METHODS 734 DRD patients and 151 asymptomatic GCH1 mutation carriers were included using an MDSGene systematic literature review and an automated classification approach to distinguish between different forms of monogenic DRDs. RESULTS Whereas dystonia, L-Dopa responsiveness, early age at onset, and diurnal fluctuations were identified as red flags, parkinsonism without dystonia was rarely reported (11%) and combined with dystonia in only 18% of patients. While sex was equally distributed in autosomal recessive DRD, there was female predominance in autosomal dominant DYT/PARK-GCH1 patients accompanied by a lower median age at onset and more dystonia in females compared to males. Accordingly, the majority of asymptomatic heterozygous GCH1 mutation carriers (>8 years of age) were males. Multiple other subgroup-specific characteristics were identified, showing high accuracy in the automated classification approach: Seizures and microcephaly were mostly seen in DYT/PARK-PTS, autonomic symptoms appeared commonly in DYT/PARK-TH and DYT/PARK-PTS, and sleep disorders and oculogyric crises in DYT/PARK-SPR. Biochemically, homovanillic acid and 5-hydroxyindoleacetic acid in CSF were reduced in most DRDs, but neopterin and biopterin were increased only in DYT/PARK-PTS and DYT/PARK-SPR. Hyperphenylalaninemia was seen in DYT/PARK-PTS, DYT/PARK-QDPR, and rarely reported in autosomal recessive DYT/PARK-GCH1. CONCLUSIONS Our indicators will help to specify diagnosis and accelerate start of treatment. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Anne Weissbach
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany.,Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
| | - Martje G Pauly
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany.,Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany.,Department of Neurology, University Hospital Schleswig Holstein, Lübeck, Germany
| | - Rebecca Herzog
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany.,Department of Neurology, University Hospital Schleswig Holstein, Lübeck, Germany
| | - Lisa Hahn
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany.,Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
| | - Sara Halmans
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
| | - Feline Hamami
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
| | - Christina Bolte
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
| | - Sarah Camargos
- Department of Internal Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Beomseok Jeon
- Department of Neurology, Seoul National University College of Medicine, Seoul, South Korea
| | - Manju A Kurian
- Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Thomas Opladen
- Division of Child Neurology and Metabolic Disorders, University Children's Hospital, Heidelberg, Germany
| | - Norbert Brüggemann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany.,Department of Neurology, University Hospital Schleswig Holstein, Lübeck, Germany
| | | | - Inke R König
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
22
|
Tesoro-Cruz E, Manuel-Apolinar L, Oviedo N, Orozco-Suárez S, Crespo Ramírez M, Bekker-Méndez VC, Aguirre-García MM, Rojas-Osornio SA, Paredes-Cervantes V, Pérez de la Mora M. Increase of 5-HT levels is induced both in mouse brain and HEK-293 cells following their exposure to a non-viral tryptophan hydroxylase construct. Transl Psychiatry 2021; 11:515. [PMID: 34625528 PMCID: PMC8501106 DOI: 10.1038/s41398-021-01634-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 09/03/2021] [Accepted: 09/21/2021] [Indexed: 12/21/2022] Open
Abstract
Tryptophan hydroxylase type 2 (Tph2) is the rate-limiting enzyme for serotonin (5-HT) biosynthesis in the brain. Dysfunctional Tph2 alters 5-HT biosynthesis, leading to a deficiency of 5-HT, which could have repercussions on human behavior. In the last decade, several studies have associated polymorphisms of the TPH2 gene with suicidal behavior. Additionally, a 5-HT deficiency has been implicated in various psychiatric pathologies, including alcoholism, impulsive behavior, anxiety, and depression. Therefore, the TPH2 gene could be an ideal target for analyzing the effects of a 5-HT deficiency on brain function. The aim of this study was to use the construct pIRES-hrGFP-1a-Tph2-FLAG to treat CD1-male mice and to transfect HEK-293-cells and then to evaluate whether this treatment increases 5-HT production. 5-HT levels were enhanced 48 h post-transfection, in HEK-293 cells. Three days after the ocular administration of pIRES-hrGFP-1a-Tph2-FLAG to mice, putative 5-HT production was significantly higher than in the control in both hypothalamus and amygdala, but not in the brainstem. Further research will be needed on the possible application of this treatment for psychiatric diseases involving a Tph2 dysfunction or serotonin deficiency.
Collapse
Affiliation(s)
- Emiliano Tesoro-Cruz
- Unidad de Investigación Biomédica en Inmunología e Infectología, Hospital de Infectología, Centro Médico Nacional "La Raza", IMSS, Ciudad de México, México.
| | - Leticia Manuel-Apolinar
- grid.418385.3Unidad de Investigación Médica en Enfermedades Endócrinas, UMAE, Hospital de Especialidades, Centro Médico Nacional “Siglo XXI”, IMSS, Ciudad de México, México
| | - Norma Oviedo
- grid.418382.40000 0004 1759 7317Unidad de Investigación Biomédica en Inmunología e Infectología, Hospital de Infectología, Centro Médico Nacional “La Raza”, IMSS, Ciudad de México, México
| | - Sandra Orozco-Suárez
- grid.418385.3Unidad de Investigación Médica en Enfermedades Neurólogicas, UMAE, Hospital de Especialidades, Centro Médico Nacional “Siglo XXI”, IMSS, Ciudad de México, México
| | - Minerva Crespo Ramírez
- grid.9486.30000 0001 2159 0001División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Vilma Carolina Bekker-Méndez
- grid.418382.40000 0004 1759 7317Unidad de Investigación Biomédica en Inmunología e Infectología, Hospital de Infectología, Centro Médico Nacional “La Raza”, IMSS, Ciudad de México, México
| | - M. Magdalena Aguirre-García
- grid.419172.80000 0001 2292 8289Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, UNAM, Instituto Nacional de Cardiología Ignacio Chávez., Ciudad de México, México
| | - Sandra Angélica Rojas-Osornio
- grid.418275.d0000 0001 2165 8782Sección de Estudios de Posgrado e Investigación de la Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México, México
| | - Vladimir Paredes-Cervantes
- grid.418382.40000 0004 1759 7317Unidad de Investigación Biomédica en Inmunología e Infectología, Hospital de Infectología, Centro Médico Nacional “La Raza”, IMSS, Ciudad de México, México
| | - Miguel Pérez de la Mora
- grid.9486.30000 0001 2159 0001División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
23
|
Bian Y, Ma Y, Ma Q, Yang L, Zhu Q, Li W, Meng L. Prolonged Maternal Separation Induces the Depression-Like Behavior Susceptibility to Chronic Unpredictable Mild Stress Exposure in Mice. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6681397. [PMID: 34368355 PMCID: PMC8342142 DOI: 10.1155/2021/6681397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/07/2021] [Accepted: 07/07/2021] [Indexed: 12/25/2022]
Abstract
Early life stress is an important determinant for developing depression later in life. It is reported that maternal separation (MS) could trigger stress sensitivity in adulthood when exposed to stress again. However, it could also result in resilience to stress-induced depression. The conclusions are contradictory. To address this issue, C57BL/6N newborn pups were exposed to either daily short MS (MS for 15 min per day; MS15) or prolonged MS (MS for 180 min per day; MS180) from the first day postpartum (PD1) to PD21. Adult mice were then subjected to chronic unpredictable mild stress (CUMS) exposure from PD64 to PD105. The behavior tests such as the forced swimming test (FST), tail suspension test (TST), and open-field test were performed once a week during this time. Besides, the hippocampal neurosteroids, serum stress hormones, and hippocampal monoamine neurotransmitters were measured at PD106. We found that mice in the MS180 group displayed the reduced struggling time and the increased latency to immobility in both FST and TST. However, there was no significant difference in the MS15 group. The levels of hippocampal neurosteroids (progesterone and allopregnanolone) were decreased, and the serum levels of corticosterone, corticotropin-releasing hormone, and adrenocorticotropic hormone were overexpressed in the MS180 group. Besides, the expressions of monoamine neurotransmitters such as 5-hydroxytryptamine and 5-hydroxy indole acetic acid significantly decreased in the MS180 group, but not in the MS15 group. All findings revealed that prolonged MS, rather than short MS, could increase the susceptibility to depression-like behavior when reexposed to stress in adulthood. However, future studies are warranted to identify the underlying neuromolecular mechanism of the MS experience on the susceptibility to adult stress reexposure.
Collapse
Affiliation(s)
- Yaoyao Bian
- College of Acupuncture and Massage, College of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing 210023, China
- TCM Nursing Intervention Laboratory of Chronic Diseases, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yanting Ma
- College of Acupuncture and Massage, College of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qian Ma
- Department of Nursing, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215009, China
| | - Lili Yang
- School of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jingwen Library, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qinmei Zhu
- School of Medicine, Yangzhou Polytechnic College, Yangzhou 225009, China
| | - Wenlin Li
- College of Acupuncture and Massage, College of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jingwen Library, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lingdong Meng
- Department of Nephrology, Yangzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Yangzhou 225002, China
| |
Collapse
|
24
|
Huq SN, Warner AK, Buckhaults K, Sachs BD. The Effects of Brain Serotonin Deficiency on Responses to High Fat Diet in Female Mice. Front Neurosci 2021; 15:683103. [PMID: 34276291 PMCID: PMC8282998 DOI: 10.3389/fnins.2021.683103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/13/2021] [Indexed: 12/11/2022] Open
Abstract
Clinical studies have reported an increased risk of depression and anxiety disorders among individuals who are obese, and women are more likely than men to suffer from depression, anxiety, and obesity. However, the effects of obesity-promoting diets on depression- and anxiety-like behavior remain controversial. A recent study from our group used the tryptophan hydroxylase 2 (R439H) knock-in mouse line to evaluate the impact of genetic brain serotonin (5-HT) deficiency on behavioral responses to high fat diet (HFD) in male mice. That study indicated that chronic exposure to HFD induced pro-anxiety-like effects in the open field test and antidepressant-like effects in the forced swim test in wild-type males. Interestingly, the antidepressant-like effect of HFD, but not the anxiogenic effect, was blocked by brain 5-HT deficiency in males. The current work sought to repeat these studies in females. Our new data suggest that females are less susceptible than males to HFD-induced weight gain and HFD-induced alterations in behavior. In addition, the effects of chronic HFD on the expression of inflammation-related genes in the hippocampus were markedly different in females than we had previously reported in males, and HFD was shown to impact the expression of several inflammation-related genes in a genotype-dependent manner. Together, our findings highlight the importance of brain 5-HT and sex in regulating behavioral and molecular responses to HFD. Our results may have important implications for our understanding of the clinically observed sex differences in the consequences of obesity.
Collapse
Affiliation(s)
- Shama N Huq
- Department of Psychological and Brain Sciences, College of Liberal Arts and Sciences, Villanova University, Villanova, PA, United States
| | - Allison K Warner
- Department of Psychological and Brain Sciences, College of Liberal Arts and Sciences, Villanova University, Villanova, PA, United States
| | - Kerry Buckhaults
- Department of Psychological and Brain Sciences, College of Liberal Arts and Sciences, Villanova University, Villanova, PA, United States
| | - Benjamin D Sachs
- Department of Psychological and Brain Sciences, College of Liberal Arts and Sciences, Villanova University, Villanova, PA, United States.,Department of Psychological and Brain Sciences, Villanova University, Villanova, PA, United States
| |
Collapse
|
25
|
Dalvi-Garcia F, Fonseca LL, Vasconcelos ATR, Hedin-Pereira C, Voit EO. A model of dopamine and serotonin-kynurenine metabolism in cortisolemia: Implications for depression. PLoS Comput Biol 2021; 17:e1008956. [PMID: 33970902 PMCID: PMC8136856 DOI: 10.1371/journal.pcbi.1008956] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/20/2021] [Accepted: 04/10/2021] [Indexed: 12/31/2022] Open
Abstract
A major factor contributing to the etiology of depression is a neurochemical imbalance of the dopaminergic and serotonergic systems, which is caused by persistently high levels of circulating stress hormones. Here, a computational model is proposed to investigate the interplay between dopaminergic and serotonergic-kynurenine metabolism under cortisolemia and its consequences for the onset of depression. The model was formulated as a set of nonlinear ordinary differential equations represented with power-law functions. Parameter values were obtained from experimental data reported in the literature, biological databases, and other general information, and subsequently fine-tuned through optimization. Model simulations predict that changes in the kynurenine pathway, caused by elevated levels of cortisol, can increase the risk of neurotoxicity and lead to increased levels of 3,4-dihydroxyphenylaceltahyde (DOPAL) and 5-hydroxyindoleacetaldehyde (5-HIAL). These aldehydes contribute to alpha-synuclein aggregation and may cause mitochondrial fragmentation. Further model analysis demonstrated that the inhibition of both serotonin transport and kynurenine-3-monooxygenase decreased the levels of DOPAL and 5-HIAL and the neurotoxic risk often associated with depression. The mathematical model was also able to predict a novel role of the dopamine and serotonin metabolites DOPAL and 5-HIAL in the ethiology of depression, which is facilitated through increased cortisol levels. Finally, the model analysis suggests treatment with a combination of inhibitors of serotonin transport and kynurenine-3-monooxygenase as a potentially effective pharmacological strategy to revert the slow-down in monoamine neurotransmission that is often triggered by inflammation.
Collapse
Affiliation(s)
- Felipe Dalvi-Garcia
- Bioinformatics Lab, National Laboratory for Scientific Computing, Petrópolis, Rio de Janeiro, Brazil
- School of Medicine and Surgery, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luis L. Fonseca
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Ana Tereza R. Vasconcelos
- Bioinformatics Lab, National Laboratory for Scientific Computing, Petrópolis, Rio de Janeiro, Brazil
| | - Cecilia Hedin-Pereira
- Center of Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eberhard O. Voit
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| |
Collapse
|
26
|
Changes in Gut Microbiota by Chronic Stress Impair the Efficacy of Fluoxetine. Cell Rep 2021; 30:3682-3690.e6. [PMID: 32187541 DOI: 10.1016/j.celrep.2020.02.099] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 11/25/2019] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Major depressive disorders (MDDs) constitute a leading cause of disability worldwide and current pharmacological treatments are partially effective. The gut microbiota (GM) has recently emerged as a target of therapeutic interest for MDDs. In this study, we transfer GM from mice that sustained unpredictable chronic mild stress (UCMS) to healthy recipient mice. The fecal transfer induces despair-like behavior, decreases neurogenesis in the hippocampus (HpC), and impairs the antidepressant and neurogenic effects of a standard selective serotonin (5-HT) reuptake inhibitor, fluoxetine (FLX). These effects are paralleled by deficits in 5-HT bioavailability, biosynthesis, and reuptake in the HpC. Treatment with 5-hydroxytryptophan restores the levels of 5-HT and its precursors in the HpC, improves HpC neurogenesis, and alleviates despair-like symptoms. Our results reveal that stress-induced changes in GM are involved in the pathogenesis of depressive disorders and minimize FLX efficacy via alterations in the serotonergic pathway of Trp metabolism.
Collapse
|
27
|
Abstract
Neuropsychiatric disorders are major causes of the global burden of diseases, frequently co-occurring with multiple co-morbidities, especially obesity, type 2 diabetes mellitus, non-alcoholic fatty liver disease and its various risk factors in the metabolic syndrome. While the determining factors of neuropsychiatric disorders are complex, recent studies have shown that there is a strong link between diet, metabolic state and neuropsychiatric disorders, including anxiety and depression. There is no doubt that rodent models are of great value for preclinical research. Therefore, this article focuses on a rodent model of chronic consumption of high-fat diet (HFD), and/or the addition of a certain amount of cholesterol or sugar, meanwhile, summarising the pattern of diet that induces anxiety/depressive-like behaviour and the underlying mechanism. We highlight how dietary and metabolic risk influence neuropsychiatric behaviour in animals. Changes in dietary patterns, especially HFD, can induce anxiety- or depression-like behaviours, which may vary by diet exposure period, sex, age, species and genetic background of the animals used. Furthermore, dietary patterns significantly aggravate anxiety/depression-like behaviour in animal models of neuropsychiatric disorders. The mechanisms by which diet induces anxiety/depressive-like behaviour may involve neuroinflammation, neurotransmitters/neuromodulators, neurotrophins and the gut-brain axis. Future research should be focused on elucidating the mechanism and identifying the contribution of diet and diet-induced metabolic risk to neuropsychiatric disorders, which can form the basis for future clinical dietary intervention strategies for neuropsychiatric disorders.
Collapse
|
28
|
Li C, Meng F, Garza JC, Liu J, Lei Y, Kirov SA, Guo M, Lu XY. Modulation of depression-related behaviors by adiponectin AdipoR1 receptors in 5-HT neurons. Mol Psychiatry 2021; 26:4205-4220. [PMID: 31980728 PMCID: PMC7377958 DOI: 10.1038/s41380-020-0649-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/20/2019] [Accepted: 01/10/2020] [Indexed: 01/17/2023]
Abstract
The adipocyte-derived hormone adiponectin has a broad spectrum of functions beyond metabolic control. We previously reported that adiponectin acts in the brain to regulate depression-related behaviors. However, its underlying neural substrates have not been identified. Here we show that adiponectin receptor 1 (AdipoR1) is expressed in the dorsal raphe nucleus (DRN) and colocalized with tryptophan hydroxylase 2 (TPH2), a marker of serotonin (5-HT) neurons. Selective deletion of AdipoR1 in 5-HT neurons induced anhedonia in male mice, as indicated by reduced female urine sniffing time and saccharin preference, and behavioral despair in female mice and enhanced stress-induced decrease in sucrose preference in both sexes. The expression levels of TPH2 were downregulated with a concurrent reduction of 5-HT-immunoreactivity in the DRN and its two major projection regions, the hippocampus and medial prefrontal cortex (mPFC), in male but not female mice lacking AdipoR1 in 5-HT neurons. In addition, serotonin transporter (SERT) expression was upregulated in both DRN projection fields of male mice but only in the mPFC of female mice. These changes presumably lead to decreased 5-HT synthesis and/or increased 5-HT reuptake, thereby reducing 5-HT transmission. The augmented behavioral responses to the selective serotonin reuptake inhibitor fluoxetine but not desipramine, a selective norepinephrine reuptake inhibitor, observed in conditional knockout male mice supports deficient 5-HT transmission underlying depression-related phenotypes. Our results indicate that adiponectin acts on 5-HT neurons through AdipoR1 receptors to regulate depression-related behaviors in a sex-dependent manner.
Collapse
Affiliation(s)
- Chen Li
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China. .,Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| | - Fantao Meng
- grid.452240.5Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong China
| | - Jacob C. Garza
- grid.410427.40000 0001 2284 9329Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA USA ,grid.38142.3c000000041936754XPresent Address: Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Jing Liu
- grid.452240.5Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong China
| | - Yun Lei
- grid.410427.40000 0001 2284 9329Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA USA
| | - Sergei A. Kirov
- grid.410427.40000 0001 2284 9329Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA USA
| | - Ming Guo
- grid.452240.5Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong China ,grid.410427.40000 0001 2284 9329Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA USA
| | - Xin-Yun Lu
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
29
|
Wigner P, Synowiec E, Jóźwiak P, Czarny P, Bijak M, Białek K, Szemraj J, Gruca P, Papp M, Śliwiński T. The Effect of Chronic Mild Stress and Escitalopram on the Expression and Methylation Levels of Genes Involved in the Oxidative and Nitrosative Stresses as Well as Tryptophan Catabolites Pathway in the Blood and Brain Structures. Int J Mol Sci 2020; 22:ijms22010010. [PMID: 33374959 PMCID: PMC7792593 DOI: 10.3390/ijms22010010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/02/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
Previous studies suggest that depression may be associated with reactive oxygen species overproduction and disorders of the tryptophan catabolites pathway. Moreover, one-third of patients do not respond to conventional pharmacotherapy. Therefore, the study investigates the molecular effect of escitalopram on the expression of Cat, Gpx1/4, Nos1/2, Tph1/2, Ido1, Kmo, and Kynu and promoter methylation in the hippocampus, amygdala, cerebral cortex, and blood of rats exposed to CMS (chronic mild stress). The animals were exposed to CMS for two or seven weeks followed by escitalopram treatment for five weeks. The mRNA and protein expression of the genes were analysed using the TaqMan Gene Expression Assay and Western blotting, while the methylation was determined using methylation-sensitive high-resolution melting. The CMS caused an increase of Gpx1 and Nos1 mRNA expression in the hippocampus, which was normalised by escitalopram administration. Moreover, Tph1 and Tph2 mRNA expression in the cerebral cortex was increased in stressed rats after escitalopram therapy. The methylation status of the Cat promoter was decreased in the hippocampus and cerebral cortex of the rats after escitalopram therapy. The Gpx4 protein levels were decreased following escitalopram compared to the stressed/saline group. It appears that CMS and escitalopram influence the expression and methylation of the studied genes.
Collapse
Affiliation(s)
- Paulina Wigner
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland; (P.W.); (E.S.); (K.B.)
| | - Ewelina Synowiec
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland; (P.W.); (E.S.); (K.B.)
| | - Paweł Jóźwiak
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland;
| | - Piotr Czarny
- Department of Medical Biochemistry, Medical University of Lodz, 90-647 Lodz, Poland; (P.C.); (J.S.)
| | - Michał Bijak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland;
| | - Katarzyna Białek
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland; (P.W.); (E.S.); (K.B.)
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 90-647 Lodz, Poland; (P.C.); (J.S.)
| | - Piotr Gruca
- Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland; (P.G.); (M.P.)
| | - Mariusz Papp
- Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland; (P.G.); (M.P.)
| | - Tomasz Śliwiński
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland; (P.W.); (E.S.); (K.B.)
- Correspondence: ; Tel.: +48-42-635-44-86; Fax: +48-42-635-44-84
| |
Collapse
|
30
|
Inflammatory Response, a Key Pathophysiological Mechanism of Obesity-Induced Depression. Mediators Inflamm 2020; 2020:8893892. [PMID: 33299381 PMCID: PMC7707993 DOI: 10.1155/2020/8893892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years, with the acceleration of life rhythm and the increase of social competition, the incidence of obesity and depression has been increasing, which has seriously affected the quality of life and health of people. Obesity and depression, two seemingly unrelated physical and psychological diseases, in fact, are closely related: obese people are more likely to have depression than nonobese ones. We have reviewed and analyzed the relevant research literature and found that the inflammatory response plays a key role in obesity-induced depression. This article will discuss in detail the inflammatory mechanisms by which obesity induces depression.
Collapse
|
31
|
Tesoro-Cruz E, Oviedo N, Manuel-Apolinar L, Orozco-Suárez S, Pérez de la Mora M, Martínez-Pérez G, Guerra-Castillo FX, Aguirre-Alvarado C, Bekker-Méndez VC. Ophthalmic Administration of a DNA Plasmid Harboring the Murine Tph2 Gene: Evidence of Recombinant Tph2-FLAG in Brain Structures. Mol Biotechnol 2020; 62:200-209. [PMID: 32030628 DOI: 10.1007/s12033-020-00239-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tryptophan hydroxylase-type 2 (Tph2) is the first rate-limiting step in the biosynthesis of serotonin (5-HT) in the brain. The ophthalmic administration (Op-Ad) is a non-invasive method that allows delivering genetic vehicles through the eye and reaches the brain. Here, the murine Tph2 gene was cloned in a non-viral vector (pIRES-hrGFP-1a), generating pIRES-hrGFP-1a-Tph2, plus the FLAG-tag. Recombinant Tph2-FLAG was detected and tested in vitro and in vivo, where 25 μg of pIRES-hrGFP-1a-Tph2-FLAG was Op-Ad to mice. The construct was capable of expressing and producing the recombinant Tph2-FLAG in vitro and in vivo. The in vivo assays showed that the construct efficiently crossed the Hemato-Ocular Barrier and the Blood-Brain Barrier, reached brain cells, passed the optical nerves, and transcribed mRNA-Tph2-FLAG in different brain areas. The recombinant Tph2-FLAG was observed in amygdala and brainstem, mainly in raphe dorsal and medial. Relative Tph2 expression of threefold over basal level was recorded three days after Op-Ad. These results demonstrated that pIRES-hrGFP-Tph2-FLAG, administrated through the eyes was capable of reaching the brain, transcribing, and translating Tph2. In conclusion, this study showed the feasibility of delivering therapeutic genes, such as the Tph2, the first enzyme, rate-limiting step in the 5-HT biosynthesis.
Collapse
Affiliation(s)
- Emiliano Tesoro-Cruz
- Unidad de Investigación Biomédica en Infectología e Inmunología, Hospital de Infectología, Centro Médico Nacional "La Raza", IMSS, Paseo de las Jacarandas s/n esquina Calzada Vallejo, Col. La Raza, C.P. 02990, Mexico City, Mexico.
| | - Norma Oviedo
- Unidad de Investigación Biomédica en Infectología e Inmunología, Hospital de Infectología, Centro Médico Nacional "La Raza", IMSS, Paseo de las Jacarandas s/n esquina Calzada Vallejo, Col. La Raza, C.P. 02990, Mexico City, Mexico.
| | - Leticia Manuel-Apolinar
- Unidad de Investigación Médica en Enfermedades Endocrinas, UMAE, Hospital de Especialidades, Centro Médico Nacional "Siglo XXI", IMSS, Mexico City, Mexico
| | - Sandra Orozco-Suárez
- Unidad de Investigación Médica en Enfermedades Neurológicas, UMAE, Hospital de Especialidades, Centro Médico Nacional "Siglo XXI", IMSS, Mexico City, Mexico
| | - Miguel Pérez de la Mora
- Division of Neuroscience, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, UNAM, Mexico City, Mexico
| | - Gloria Martínez-Pérez
- Unidad de Investigación Biomédica en Infectología e Inmunología, Hospital de Infectología, Centro Médico Nacional "La Raza", IMSS, Paseo de las Jacarandas s/n esquina Calzada Vallejo, Col. La Raza, C.P. 02990, Mexico City, Mexico
| | - Francisco Xavier Guerra-Castillo
- Unidad de Investigación Biomédica en Infectología e Inmunología, Hospital de Infectología, Centro Médico Nacional "La Raza", IMSS, Paseo de las Jacarandas s/n esquina Calzada Vallejo, Col. La Raza, C.P. 02990, Mexico City, Mexico
| | - Charmina Aguirre-Alvarado
- Unidad de Investigación Biomédica en Infectología e Inmunología, Hospital de Infectología, Centro Médico Nacional "La Raza", IMSS, Paseo de las Jacarandas s/n esquina Calzada Vallejo, Col. La Raza, C.P. 02990, Mexico City, Mexico
| | - Vilma Carolina Bekker-Méndez
- Unidad de Investigación Biomédica en Infectología e Inmunología, Hospital de Infectología, Centro Médico Nacional "La Raza", IMSS, Paseo de las Jacarandas s/n esquina Calzada Vallejo, Col. La Raza, C.P. 02990, Mexico City, Mexico
| |
Collapse
|
32
|
The Impact of Chronic Mild Stress and Agomelatine Treatment on the Expression Level and Methylation Status of Genes Involved in Tryptophan Catabolic Pathway in PBMCs and Brain Structures. Genes (Basel) 2020; 11:genes11091093. [PMID: 32962062 PMCID: PMC7563711 DOI: 10.3390/genes11091093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 01/11/2023] Open
Abstract
Depression is the serious mental disorder. Previous studies suggest that the development mechanism of depression may be associated with disorders of the tryptophan catabolic pathway (TRYCAT). Thus, this study investigates the effect of agomelatine treatment on the expression and methylation status of genes involved in TRYCAT in the brain and blood of rats exposed to a chronic mild stress (CMS). Separate groups of rats were exposed to CMS for two or seven weeks; the second group received vehicle or agomelatine for five weeks. After completion of both stress conditions and treatment, the expression levels of messenger RNA (mRNA) and protein, as well as the methylation status of promoters, were measured in peripheral blood mononuclear cells (PBMCs) and in brain structures with the use of TaqMan Gene Expression Assay, Western blot, and methylation-sensitive high-resolution melting techniques. In PBMCs, Kmo mRNA expression increased in the group after CMS, while this effect was normalized by agomelatine therapy. In brain, KatI and KatII expression changed following CMS exposure. Moreover, CMS decreased the methylation status of the second Tdo2 promoter in the amygdala. Protein expression of Tph1, Tph2, Ido1, and KatII changed in the group after CMS and agomelatine administration, most prominently in the basal ganglia, cerebral cortex, hippocampus, and amygdala. The results indicate that CMS and agomelatine affect the mRNA and protein expression, as well as the methylation of promoters of genes involved in the tryptophan catabolic pathway.
Collapse
|
33
|
Sgambato V. Breathing new life into neurotoxic-based monkey models of Parkinson's disease to study the complex biological interplay between serotonin and dopamine. PROGRESS IN BRAIN RESEARCH 2020; 261:265-285. [PMID: 33785131 DOI: 10.1016/bs.pbr.2020.07.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Numerous clinical studies have shown that the serotonergic system also degenerates in patients with Parkinson's disease. The causal role of this impairment in Parkinson's symptomatology and the response to treatment remains to be refined, in particular thanks to approaches allowing the two components DA and 5-HT to be isolated if possible. We have developed a macaque monkey model of Parkinson's disease exhibiting a double lesion (dopaminergic and serotonergic) thanks to the sequential use of MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) and MDMA (3,4-methylenedioxy-N-methamphetamine) (or MDMA prior MPTP). We characterized this monkey model by multimodal imaging (PET, positron emission tomography with several radiotracers; DTI, diffusion tensor imaging), behavioral assessments (parkinsonism, dyskinesia, neuropsychiatric-like behavior) and post-mortem analysis (with DA and 5-HT markers). When administrated after MPTP, MDMA damaged the 5-HT presynaptic system without affecting the remaining DA neurons. The lesion of 5-HT fibers induced by MDMA altered rigidity and prevented dyskinesia and neuropsychiatric-like symptoms induced by levodopa therapy in MPTP-treated animals. Interestingly also, prior MDMA administration aggravates the parkinsonian deficits and associated DA injury. Dystonic postures, action tremor and global spontaneous activities were significantly affected. All together, these data clearly indicate that late or early lesions of the 5-HT system have a differential impact on parkinsonian symptoms in the macaque model of Parkinson's disease. Whether MDMA has an impact on neuropsychiatric-like symptoms such as apathy, anxiety, depression remains to be addressed. Despite its limitations, this toxin-based double-lesioned monkey model takes on its full meaning and provides material for the experimental study of the heterogeneity of patients.
Collapse
Affiliation(s)
- Véronique Sgambato
- Université de Lyon, CNRS UMR 5229, Institut des Sciences Cognitives Marc Jeannerod, Bron, France.
| |
Collapse
|
34
|
Meng F, Liu J, Dai J, Wu M, Wang W, Liu C, Zhao D, Wang H, Zhang J, Li M, Li C. Brain-derived neurotrophic factor in 5-HT neurons regulates susceptibility to depression-related behaviors induced by subchronic unpredictable stress. J Psychiatr Res 2020; 126:55-66. [PMID: 32416387 DOI: 10.1016/j.jpsychires.2020.05.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/12/2020] [Accepted: 05/04/2020] [Indexed: 12/15/2022]
Abstract
Chronic stress is a major risk factor for the development of depression. Brain-derived neurotrophic factor (BDNF) plays an important role in neural functions and exhibits antidepressant effects. However, studies on depression-related behavioral response to BDNF have mainly focused on the limbic system, whereas other regions of the brain still require further exploration. Here, we report that exposure to chronic unpredictable stress (CUS) can induce depression-associated behaviors in mice. CUS could decrease total Bdnf mRNA and protein levels in the dorsal raphe nucleus (DRN), which correlated with depression-related behaviors. A corresponding reduction in exon-specific Bdnf mRNA was observed in the DRN of CUS mice. Bdnf was highly expressed in 5- Hydroxytryptamine (5-HT) neurons from the DRN. Selective deletion of Bdnf in 5-HT neurons alone could not induce anhedonia and behavioral despair in male or female mice, as indicated by the unchanged female urine sniffing time and preference for sucrose/saccharin. However, it could increase the latency to food in female mice, but not in male mice as shown by novelty-suppressed food test. Nevertheless, enhanced stress-induced susceptibility is observed in these male mice as suggested by the decrease in female urine sniffing time, and for female mice by the reduced sucrose preference and increased immobility in forced swim test. Furtherly, total Bdnf mRNA levels in DRN were correlated with depression-related behaviors of female, but not male 5-HT neurons specific Bdnf knockout mice. Our results indicate that BDNF might act on 5-HT neurons to regulate depression-related behaviors and stress vulnerability in a sex-dependent manner.
Collapse
Affiliation(s)
- Fantao Meng
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jing Liu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Juanjuan Dai
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Min Wu
- Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Wentao Wang
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Cuilan Liu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Di Zhao
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Hongcai Wang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jingyan Zhang
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Min Li
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chen Li
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
35
|
Wigner P, Synowiec E, Jóźwiak P, Czarny P, Bijak M, Białek K, Szemraj J, Gruca P, Papp M, Śliwiński T. The Effect of Chronic Mild Stress and Venlafaxine on the Expression and Methylation Levels of Genes Involved in the Tryptophan Catabolites Pathway in the Blood and Brain Structures of Rats. J Mol Neurosci 2020; 70:1425-1436. [PMID: 32406039 PMCID: PMC7399689 DOI: 10.1007/s12031-020-01563-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/22/2020] [Indexed: 12/21/2022]
Abstract
A growing body of evidence suggests that depression may be associated with impairment of the tryptophan catabolites (TRYCATs) pathway. The present study investigated the effects of the chronic administration of venlafaxine on the expression and methylation status of Katl, Tph1/2, Ido1, Kmo and Kynu in the brain and blood of rats exposed to the CMS model of depression. The rats were subjected to the CMS procedure for 2 or 7 weeks and administered venlafaxine (10 mg/kg/day, IP) for 5 weeks. mRNA and protein expression and the methylation status of gene promoters in PBMCs and six brain structures were evaluated and analysed using the TaqMan Gene Expression Assay and Western blotting, and methylation-sensitive high-resolution melting (MS-HRM), respectively. We found that the CMS procedure increased KatI expression in the midbrain and KatII expression in the midbrain and the amygdala, while venlafaxine administration decreased KatII expression in the hypothalamus and the cerebral cortex. The methylation status of the Tph1 and Kmo promoters in peripheral blood mononuclear cells (PBMCs) was significantly increased in the stressed group after antidepressant therapy. The protein levels of Tph1 and Ido1 were decreased following venlafaxine administration. Our results confirmed that CMS and venlafaxine modulate the expression levels and methylation status of genes involved in the TRYCATs pathway.
Collapse
Affiliation(s)
- Paulina Wigner
- Faculty of Biology and Environmental Protection, Laboratory of Medical Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Ewelina Synowiec
- Faculty of Biology and Environmental Protection, Laboratory of Medical Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Paweł Jóźwiak
- Faculty of Biology and Environmental Protection, Department of Cytobiochemistry, University of Lodz, Lodz, Poland
| | - Piotr Czarny
- Department of Medical Biochemistry, Medical University of Lodz, Lodz, Poland
| | - Michał Bijak
- Faculty of Biology and Environmental Protection, Department of General Biochemistry, University of Lodz, Lodz, Poland
| | - Katarzyna Białek
- Faculty of Biology and Environmental Protection, Laboratory of Medical Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, Lodz, Poland
| | - Piotr Gruca
- Polish Academy of Sciences, Institute of Pharmacology, Krakow, Poland
| | - Mariusz Papp
- Polish Academy of Sciences, Institute of Pharmacology, Krakow, Poland
| | - Tomasz Śliwiński
- Faculty of Biology and Environmental Protection, Laboratory of Medical Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland.
| |
Collapse
|
36
|
Del Colle A, Israelyan N, Gross Margolis K. Novel aspects of enteric serotonergic signaling in health and brain-gut disease. Am J Physiol Gastrointest Liver Physiol 2020; 318:G130-G143. [PMID: 31682158 PMCID: PMC6985840 DOI: 10.1152/ajpgi.00173.2019] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/17/2019] [Accepted: 10/17/2019] [Indexed: 02/08/2023]
Abstract
Gastrointestinal (GI) comorbidities are common in individuals with mood and behavioral dysfunction. Similarly, patients with GI problems more commonly suffer from co-morbid psychiatric diagnoses. Although the central and enteric nervous systems (CNS and ENS, respectively) have largely been studied separately, there is emerging interest in factors that may contribute to disease states involving both systems. There is strong evidence to suggest that serotonin may be an important contributor to these brain-gut conditions. Serotonin has long been recognized for its critical functions in CNS development and function. The majority of the body's serotonin, however, is produced in the GI tract, where it plays key roles in ENS development and function. Further understanding of the specific impact that enteric serotonin has on brain-gut disease may lay the foundation for the creation of novel therapeutic targets. This review summarizes the current data focusing on the important roles that serotonin plays in ENS development and motility, with a focus on novel aspects of serotonergic signaling in medical conditions in which CNS and ENS co-morbidities are common, including autism spectrum disorders and depression.
Collapse
Affiliation(s)
- Andrew Del Colle
- Morgan Stanley Children's Hospital, Department of Pediatrics, Columbia University Medical Center, New York, New York
| | - Narek Israelyan
- Morgan Stanley Children's Hospital, Department of Pediatrics, Columbia University Medical Center, New York, New York
- Vagelos College of Physicians and Surgeons, Columbia University Medical Center, New York, New York
| | - Kara Gross Margolis
- Morgan Stanley Children's Hospital, Department of Pediatrics, Columbia University Medical Center, New York, New York
| |
Collapse
|
37
|
Karth MM, Baugher BJ, Daly N, Karth MD, Gironda SC, Sachs BD. Brain 5-HT Deficiency Prevents Antidepressant-Like Effects of High-Fat-Diet and Blocks High-Fat-Diet-Induced GSK3β Phosphorylation in the Hippocampus. Front Mol Neurosci 2019; 12:298. [PMID: 31920532 PMCID: PMC6917648 DOI: 10.3389/fnmol.2019.00298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/20/2019] [Indexed: 02/06/2023] Open
Abstract
Obesity is associated with an increased risk of depression and anxiety disorders, but the nature of the relationship(s) between obesity and mental illness remains highly controversial. Some argue that depression and anxiety lead to increased consumption of "comfort foods," the intake of which reduces negative affect and promotes obesity. In contrast, others have theorized that negative affect results from chronic excessive consumption of highly palatable foods. The brain serotonin (5-HT) system has long been implicated in both the development and treatment of mental illness. Preclinical studies have shown that low brain 5-HT exacerbates depression- and anxiety-like behaviors induced by stress and blocks reductions in depression-like behavior induced by antidepressants, but the effects of brain 5-HT deficiency on responses to high-fat diet (HFD) have not been explored. The current work used genetically modified mice to evaluate the effects of low 5-HT on behavioral and molecular alterations induced by chronic exposure to HFD. Our results reveal that HFD decreases depression-like behavior and increases some anxiety-like behaviors in wild-type (WT) mice. However, genetic brain 5-HT deficiency blocks HFD-induced reductions in forced swim immobility and prevents HFD-induced increases in hippocampal GSK3β phosphorylation despite having no significant effects on HFD-induced changes in body weight or anxiety-like behavior. Together, our results suggest that brain 5-HT deficiency significantly impacts a subset of behavioral and molecular responses to HFD, a finding that could help explain the complex relationships between obesity and mental illness.
Collapse
Affiliation(s)
- Michelle M Karth
- Department of Psychological and Brain Sciences, College of Liberal Arts and Sciences, Villanova University, Villanova, PA, United States
| | - Brittany J Baugher
- Department of Psychological and Brain Sciences, College of Liberal Arts and Sciences, Villanova University, Villanova, PA, United States
| | - Nicole Daly
- Department of Psychological and Brain Sciences, College of Liberal Arts and Sciences, Villanova University, Villanova, PA, United States
| | - Melinda D Karth
- Department of Psychological and Brain Sciences, College of Liberal Arts and Sciences, Villanova University, Villanova, PA, United States
| | - Stephen C Gironda
- Department of Psychological and Brain Sciences, College of Liberal Arts and Sciences, Villanova University, Villanova, PA, United States
| | - Benjamin D Sachs
- Department of Psychological and Brain Sciences, College of Liberal Arts and Sciences, Villanova University, Villanova, PA, United States
| |
Collapse
|
38
|
Slow-release delivery enhances the pharmacological properties of oral 5-hydroxytryptophan: mouse proof-of-concept. Neuropsychopharmacology 2019; 44:2082-2090. [PMID: 31035282 PMCID: PMC6898594 DOI: 10.1038/s41386-019-0400-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/10/2019] [Accepted: 04/16/2019] [Indexed: 11/09/2022]
Abstract
5-hydroxytryptophan (5-HTP) has shown therapeutic promise in a range of human CNS disorders. But native 5-HTP immediate release (IR) is poorly druggable, as rapid absorption causes rapid onset of adverse events, and rapid elimination causes fluctuating exposure. Recently, we reported that 5-HTP delivered as slow-release (SR) in mice augmented the brain pro-serotonergic effect of selective serotonin reuptake inhibitors (SSRIs), without the usual adverse events associated with 5-HTP IR. However, our previous study entailed translational limitations, in terms of route, dose, and duration. Here we modeled oral 5-HTP SR in mice by administering 5-HTP via the food. We modeled oral SSRI treatment via fluoxetine in the water, in a regimen recapitulating clinical pharmacokinetics and pharmacodynamics. 5-HTP SR produced plasma 5-HTP levels well within the range enhancing brain 5-HT function in humans. 5-HTP SR robustly increased brain 5-HT synthesis and levels. When administered with an SSRI, 5-HTP SR enhanced 5-HT-sensitive behaviors and neurotrophic mRNA expression. 5-HTP SR's pro-serotonergic effects were stronger in mice with endogenous brain 5-HT deficiency. In a comprehensive screen, 5-HTP SR was devoid of overt toxicological effects. The present preclinical data, appreciated in the context of published 5-HTP clinical data, suggest that 5-HTP SR could represent a new therapeutic approach to the plethora of CNS disorders potentially treatable with a pro-serotonergic drug. 5-HTP SR might in particular be therapeutically relevant when brain 5-HT deficiency is pathogenic and as an adjunctive augmentation therapy to SSRI therapy.
Collapse
|
39
|
Xing H, Zhang X, Xing N, Qu H, Zhang K. Uncovering pharmacological mechanisms of Zhi-Zi-Hou-Po decoction in chronic unpredictable mild stress induced rats through pharmacokinetics, monoamine neurotransmitter and neurogenesis. JOURNAL OF ETHNOPHARMACOLOGY 2019; 243:112079. [PMID: 31302206 DOI: 10.1016/j.jep.2019.112079] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/10/2019] [Accepted: 07/10/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zhi-Zi-Hou-Po decoction (ZZHPD), a classical Chinese prescription, has been reported to improve depressive behaviors in clinic. However, definite pharmacological effects and mechanisms of ZZHPD on monoaminergic system and hippocampal neurogenesis are ambiguous. It need to be further illuminated. AIM OF THE STUDY Our study is designed to reveal pharmacological mechanisms of ZZHPD on depression through pharmacokinetics, monoamine neurotransmitters and neurogenesis. MATERIALS AND METHODS Chronic unpredictable mild stress (CUMS) is used to establish rats model of depression. Then, the antidepressant effects of ZZHPD are evaluated by detecting body weight, sucrose preference and forced swimming test. The regulatory functions of ZZHPD on monoaminergic system are assessed by measuring monoamine neurotransmitters, neurotransmitter precursor substances, synthesized rate-limiting enzymes and transporters. Finally, potential molecular mechanism of ZZHPD on hippocampal neurogenesis is evaluated by investigating newborn immature neuron and newborn mature neuron. RESULTS Our results show that ZZHPD remarkably normalizes CUMS-induced decline in weight gain, decrease of sucrose consumption rate in sucrose preference test and increase of immobility time in forced swimming test. Moreover, ZZHPD significantly reverses CUMS-induced reduction of 5-hydroxytryptamine (5-HT), dopamine (DA), tryptophan (Trp), tyrosine (Tyr), tryptophan hydroxylase2 (TPH2) and tyrosine hydroxylase (TH), whereas decreases level of serotonin transporter (SERT) in CUMS-induced rats. Finally, ZZHPD obviously improves CUMS-induced decrease of newborn immature neuron and newborn mature neuron in dentate gyrus of hippocampus. CONCLUSION This study demonstrates that ZZHPD can alleviate CUMS-induced depression-like behaviors. It is probably attributed to the fact that ZZHPD could enhance monoaminergic system and hippocampal neurogenesis. Our findings provide the new perspectives on molecular targets of ZZHPD, and it will facilitate its clinical application.
Collapse
Affiliation(s)
- Hang Xing
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaoxu Zhang
- 13 Ward of General Surgery Department, Da Qing Long Nan Hospital, Daqing, China
| | - Nannan Xing
- Department of Pharmacy, Harbin Traditional Chinese Medicine Hospital, Harbin, China
| | | | - Kuo Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
40
|
Israelyan N, Colle AD, Li Z, Park Y, Xing A, Jacobsen JP, Luna RA, Jensen DD, Madra M, Saurman V, Rahim R, Latorre R, Law K, Carson W, Bunnett NW, Caron MG, Margolis KG. Effects of Serotonin and Slow-Release 5-Hydroxytryptophan on Gastrointestinal Motility in a Mouse Model of Depression. Gastroenterology 2019; 157:507-521.e4. [PMID: 31071306 PMCID: PMC6650329 DOI: 10.1053/j.gastro.2019.04.022] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Mood disorders and constipation are often comorbid, yet their shared etiologies have rarely been explored. The neurotransmitter serotonin (5-HT) regulates central nervous system and enteric nervous system (ENS) development and long-term functions, including gastrointestinal (GI) motility and mood. Therefore, defects in neuron production of 5-HT might result in brain and intestinal dysfunction. Tryptophan hydroxylase 2 (TPH2) is the rate-limiting enzyme in 5-HT biosynthesis. A variant of TPH2 that encodes the R441H substitution (TPH2-R441H) was identified in individuals with severe depression. We studied mice with an analogous mutation (TPH2-R439H), which results in a 60%-80% decrease in levels of 5-HT in the central nervous system and behaviors associated with depression in humans. Feeding chow that contains 5-HTP slow release (5-HTP SR) to TPH2-R439H mice restores levels of 5-HT in the central nervous system and reduces depressive-like behaviors. METHODS We compared the effects of feeding chow, with or without 5-HTP SR, to mice with the TPH2-R439H mutation and without this mutation (control mice). Myenteric and submucosal plexuses were isolated from all 4 groups of mice, and immunocytochemistry was used to quantify total enteric neurons, serotonergic neurons, and 5-HT-dependent subsets of neurons. We performed calcium imaging experiments to evaluate responses of enteric neurons to tryptamine-evoked release of endogenous 5-HT. In live mice, we measured total GI transit, gastric emptying, small intestinal transit, and propulsive colorectal motility. To measure colonic migrating motor complexes (CMMCs), we isolated colons and constructed spatiotemporal maps along the proximodistal length to quantify the frequency, velocity, and length of CMMCs. We measured villus height, crypt perimeter, and relative densities of enterochromaffin and enteroendocrine cells in small intestinal tissue. RESULTS Levels of 5-HT were significantly lower in enteric neurons from TPH2-R439H mice than from control mice. TPH2-R439H mice had abnormalities in ENS development and ENS-mediated GI functions, including reduced motility and intestinal epithelial growth. Total GI transit and propulsive colorectal motility were slower in TPH2-R439H mice than controls, and CMMCs were slower and less frequent. Villus height and crypt perimeter were significantly decreased in colon tissues from TPH2-R439H mice compared with controls. Administration of 5-HTP SR to adult TPH2-R439H mice restored 5-HT to enteric neurons and reversed these abnormalities. Adult TPH2-R439H mice given oral 5-HTP SR had normalized numbers of enteric neurons, total GI transit, and colonic motility. Intestinal tissue from these mice had normal measures of CMMCs and enteric epithelial growth CONCLUSIONS: In studies of TPH2-R439H mice, we found evidence for reduced release of 5-HT from enteric neurons that results in defects in ENS development and GI motility. Our findings indicate that neuron production of 5-HT links constipation with mood dysfunction. Administration of 5-HTP SR to mice restored 5-HT to the ENS and normalized GI motility and growth of the enteric epithelium. 5-HTP SR might be used to treat patients with intestinal dysfunction associated with low levels of 5-HT.
Collapse
Affiliation(s)
- Narek Israelyan
- Morgan Stanley Children’s Hospital, Department of Pediatrics, Columbia University Medical Center; New York, NY
| | - Andrew Del Colle
- Morgan Stanley Children’s Hospital, Department of Pediatrics, Columbia University Medical Center; New York, NY,Institute of Human Nutrition, Columbia University Medical Center; New York, NY
| | - Zhishan Li
- Morgan Stanley Children’s Hospital, Department of Pediatrics, Columbia University Medical Center; New York, NY,Department of Pathology and Cell Biology, Columbia University Medical Center; New York, NY
| | - Yeji Park
- Morgan Stanley Children’s Hospital, Department of Pediatrics, Columbia University Medical Center; New York, NY,Institute of Human Nutrition, Columbia University Medical Center; New York, NY
| | - Albert Xing
- Morgan Stanley Children’s Hospital, Department of Pediatrics, Columbia University Medical Center; New York, NY
| | | | - Ruth Ann Luna
- Texas Children’s Hospital Microbiome Center, Baylor College of Medicine; Houston, TX
| | - Dane D. Jensen
- Departments of Surgery and Pharmacology, Columbia University Medical Center; New York, NY
| | - Moneek Madra
- Morgan Stanley Children’s Hospital, Department of Pediatrics, Columbia University Medical Center; New York, NY,Institute of Human Nutrition, Columbia University Medical Center; New York, NY
| | - Virginia Saurman
- Morgan Stanley Children’s Hospital, Department of Pediatrics, Columbia University Medical Center; New York, NY
| | - Ray Rahim
- Morgan Stanley Children’s Hospital, Department of Pediatrics, Columbia University Medical Center; New York, NY,Department of Pathology and Cell Biology, Columbia University Medical Center; New York, NY
| | - Rocco Latorre
- Departments of Surgery and Pharmacology, Columbia University Medical Center; New York, NY
| | - Kimberly Law
- Morgan Stanley Children’s Hospital, Department of Pediatrics, Columbia University Medical Center; New York, NY
| | - William Carson
- Department of Cell Biology, Duke University School of Medicine; Durham, NC
| | - Nigel W. Bunnett
- Departments of Surgery and Pharmacology, Columbia University Medical Center; New York, NY
| | - Marc G. Caron
- Department of Cell Biology, Duke University School of Medicine; Durham, NC
| | - Kara G. Margolis
- Morgan Stanley Children’s Hospital, Department of Pediatrics, Columbia University Medical Center; New York, NY
| |
Collapse
|
41
|
Kulikova EA, Kulikov AV. Tryptophan hydroxylase 2 as a therapeutic target for psychiatric disorders: focus on animal models. Expert Opin Ther Targets 2019; 23:655-667. [PMID: 31216212 DOI: 10.1080/14728222.2019.1634691] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Tryptophan hydroxylase 2 (TPH2) is the key, rate-limiting enzyme of serotonin (5-HT) synthesis in the brain. Some polymorphic variants of the human Tph2 gene are associated with psychiatric disorders. Area covered: This review focuses on the mechanisms underlying the association between the TPH2 activity and behavioral disturbances in models of psychiatric disorders. Specifically, it discusses: 1) genetic and posttranslational mechanisms defining the TPH2 activity, 2) behavioral effects of knockout and loss-of-function mutations in the mouse Tph2 gene, 3) pharmacological inhibition and the activation of the TPH2 activity and 4) alterations in the brain TPH2 activity in animal models of psychiatric disorders. We show the dual role of the TPH2 activity: both deficit and excess of the TPH2 activity cause significant behavioral disturbances in animal models of depression, anxiety, aggression, obsessive-compulsive disorders, schizophrenia, and catalepsy. Expert opinion: Pharmacological chaperones correcting the structure of the TPH2 molecule are promising tools for treatment of some hereditary psychiatric disorders caused by loss-of-function mutations in the human Tph2 gene; while some stress-induced affective disorders, associated with the elevated TPH2 activity, may be effectively treated by TPH2 inhibitors. This dual role of TPH2 should be taken into consideration during therapy of psychiatric disorders.
Collapse
Affiliation(s)
- Elizabeth A Kulikova
- a Federal Research Center Institute of Cytology and Genetics , Siberian Division of the Russian Academy of Science , Novosibirsk , Russia
| | - Alexander V Kulikov
- a Federal Research Center Institute of Cytology and Genetics , Siberian Division of the Russian Academy of Science , Novosibirsk , Russia
| |
Collapse
|
42
|
Edwards III GA, Gamez N, Escobedo Jr. G, Calderon O, Moreno-Gonzalez I. Modifiable Risk Factors for Alzheimer's Disease. Front Aging Neurosci 2019; 11:146. [PMID: 31293412 PMCID: PMC6601685 DOI: 10.3389/fnagi.2019.00146] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 05/31/2019] [Indexed: 01/03/2023] Open
Abstract
Since first described in the early 1900s, Alzheimer's disease (AD) has risen exponentially in prevalence and concern. Research still drives to understand the etiology and pathogenesis of this disease and what risk factors can attribute to AD. With a majority of AD cases being of sporadic origin, the increasing exponential growth of an aged population and a lack of treatment, it is imperative to discover an easy accessible preventative method for AD. Some risk factors can increase the propensity of AD such as aging, sex, and genetics. Moreover, there are also modifiable risk factors-in terms of treatable medical conditions and lifestyle choices-that play a role in developing AD. These risk factors have their own biological mechanisms that may contribute to AD etiology and pathological consequences. In this review article, we will discuss modifiable risk factors and discuss the current literature of how each of these factors interplay into AD development and progression and if strategically analyzed and treated, could aid in protection against this neurodegenerative disease.
Collapse
Affiliation(s)
- George A. Edwards III
- The Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, The University of Texas Houston Health Science Center at Houston, Houston, TX, United States
| | - Nazaret Gamez
- The Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, The University of Texas Houston Health Science Center at Houston, Houston, TX, United States
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Department of Cell Biology, Facultad Ciencias, Universidad de Malaga, Malaga, Spain
| | - Gabriel Escobedo Jr.
- The Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, The University of Texas Houston Health Science Center at Houston, Houston, TX, United States
| | - Olivia Calderon
- The Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, The University of Texas Houston Health Science Center at Houston, Houston, TX, United States
| | - Ines Moreno-Gonzalez
- The Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, The University of Texas Houston Health Science Center at Houston, Houston, TX, United States
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Department of Cell Biology, Facultad Ciencias, Universidad de Malaga, Malaga, Spain
| |
Collapse
|
43
|
van Gaalen MM, Schlumbohm C, Folgering JH, Adhikari S, Bhattacharya C, Steinbach D, Stratford RE. Development of a Semimechanistic Pharmacokinetic-Pharmacodynamic Model Describing Dextroamphetamine Exposure and Striatal Dopamine Response in Rats and Nonhuman Primates following a Single Dose of Dextroamphetamine. J Pharmacol Exp Ther 2019; 369:107-120. [PMID: 30733244 DOI: 10.1124/jpet.118.254508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/23/2019] [Indexed: 11/22/2022] Open
Abstract
Acute central nervous system exposure to dextroamphetamine (d-amphetamine) elicits a multitude of effects, including dual action on the dopamine transporter (DAT) to increase extracellular dopamine, and induction of a negative feedback response to limit the dopamine increase. A semimechanistic pharmacokinetic and pharmacodynamic (PK/PD) model with consideration of these multiple effects as a basis was developed. Integrated pharmacokinetics of d-amphetamine in plasma, brain extracellular fluid (ECF) via microdialysis, and cerebrospinal fluid were characterized using a population approach. This PK model was then linked to an indirect-response pharmacodynamic model using as a basis the measurement of extracellular striatal dopamine, also via microdialysis. In both rats and nonhuman primates (NHPs), d-amphetamine stimulation of dopamine outflow (reverse transport) through DAT was primarily responsible for the dose-linear increase in dopamine. As well, in both species a moderator function was needed to account for loss of the dopamine response in the presence of a relatively sustained d-amphetamine ECF exposure, presumptive of an acute tolerance response. PK/PD model structure was consistent between species; however, there was a 10-fold faster return to baseline dopamine in NHPs in response to an acute d-amphetamine challenge. These results suggest preservation from rodents to NHPs regarding the mechanism by which amphetamine increases extracellular dopamine, but a faster system response in NHPs to tolerate this increase. This microdialysis-based PK/PD model suggests greater value in directing preclinical discovery of novel approaches that modify reverse transport stimulation to treat amphetamine abuse. General value regarding insertion of an NHP model in paradigm rodent-to-human translational research is also suggested.
Collapse
Affiliation(s)
- Marcel M van Gaalen
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Christina Schlumbohm
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Joost H Folgering
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Saugat Adhikari
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Chandrali Bhattacharya
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Douglas Steinbach
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Robert E Stratford
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| |
Collapse
|
44
|
Development of a non-human primate model to support CNS translational research: Demonstration with D-amphetamine exposure and dopamine response. J Neurosci Methods 2019; 317:71-81. [PMID: 30768951 DOI: 10.1016/j.jneumeth.2019.02.005] [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: 02/06/2019] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 11/21/2022]
Abstract
BACKGROUND Challenges specific to the discovery and development of candidate CNS drugs have led to implementation of various in silico, in vitro and in vivo approaches to improve the odds for commercialization of novel treatments. NEW METHOD Advances in analytical methodology and microdialysis probe design have enabled development of a non-human primate model capable of measuring concentrations of drugs or endogenous chemicals in brain extracellular fluid (ECF) and cerebrospinal fluid (CSF). Linking these to population modeling reduces animal numbers to support predictive translational sciences in primates. Application to measure D-amphetamine exposure and dopamine response in ECF and CSF demonstrate the approach. RESULTS Following a 0.1 mg/kg intravenous bolus dose of D-amphetamine, a population approach was used to build a plasma compartmental-based and brain physiologic-based pharmacokinetic (PK) model linking drug concentrations in plasma to brain ECF and CSF concentrations. Dopamine was also measured in brain ECF. The PK model was used to simulate the relationship between D-amphetamine exposure and dopamine response in ECF over a wide dose range. COMPARISONS WITH EXISTING METHODS Ability to co-sample and measure drug and endogenous substances in blood, brain ECF and/or CSF, coupled with population modeling, provides an in vivo approach to evaluate CNS drug penetration and effect in non-human primates. CONCLUSIONS A method to measure drug and endogenous neurochemicals in non-human primate brain fluids is demonstrated. Its basis in non-human primates merits improved confidence regarding predictions of drug exposure and target engagement in human CNS.
Collapse
|
45
|
Start AR, Allard Y, Adler A, Toblin R. Predicting Suicide Ideation in the Military: The Independent Role of Aggression. Suicide Life Threat Behav 2019; 49:444-454. [PMID: 29498089 DOI: 10.1111/sltb.12445] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/30/2017] [Indexed: 11/29/2022]
Abstract
The purpose of this study was to examine the longitudinal relationship between aggression and suicide ideation when controlling for other externalizing (i.e., alcohol misuse and risk-taking) and internalizing (i.e., depression and sleep problems) risk factors in an active duty, military sample. Preexisting data from a longitudinal study were analyzed to assess the wellness of service members across the deployment cycle. Participants were 944 active duty service members (95% male, 48% between 18 and 24 years old) who completed surveys upon initial return from deployment and approximately 3 months later. After controlling for other externalizing (alcohol misuse, risk-taking) and internalizing (depression, sleep problems) risk factors, service members reporting aggression were significantly more likely to report suicide ideation than those reporting no aggression (OR = 3.19; OR 95% CI: 1.16-8.80). The independent nature of the relationship between anger and suicidality suggests aggression may be an important indicator of suicidality for service members. Understanding the role of aggression in suicidality may improve the ability to identify at-risk service members and to develop effective interventions to reduce suicide risk.
Collapse
Affiliation(s)
- Amanda R Start
- Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Yvonne Allard
- Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Amy Adler
- Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Robin Toblin
- Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
46
|
Yang J, Zhao X, Ma J, Qiao Z, Yang X, Zhao E, Ban B, Zhu X, Cao D, Yang Y, Qiu X. The Interaction of TPH2 and 5-HT2A Polymorphisms on Major Depressive Disorder Susceptibility in a Chinese Han Population: A Case-Control Study. Front Psychiatry 2019; 10:172. [PMID: 31019472 PMCID: PMC6458236 DOI: 10.3389/fpsyt.2019.00172] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 03/08/2019] [Indexed: 12/31/2022] Open
Abstract
Purpose: TPH2 and 5-HT2A appear to play vital roles in the homeostatic regulation of serotonin levels in the brain, their genetic variations may lead to impaired homeostatic regulation of serotonin resulting in abnormal levels of serotonin in the brain, thus predisposing individuals to MDD. However, research studies have yet to confirm which gene-gene interaction effect between TPH2 and 5-HT2A polymorphisms results in increased susceptibility to MDD. Methods: A total of 565 participants, consisting of 278 MDD patients and 287 healthy controls from the Chinese Han population, were recruited for the present study. Six single nucleotide polymorphisms (SNPs) of TPH2/5-HT2A were selected to assess their interaction by use of a generalized multifactor dimensionality reduction method. Results: A-allele carriers of rs11178997 and rs120074175 were more likely to suffer from MDD than T-allele carriers of rs11178997, or G-allele carriers of rs120074175. The interaction between TPH2 (rs120074175, rs11178997) and 5-HT2A (rs7997012) was considered as the best multi-locus model upon the MDD susceptibility. Conclusions: Our data identified an important effect of TPH2 genetic variants (rs11178997 and rs120074175) upon the risk of MDD, and suggested that the interaction of TPH2/5-HT2A polymorphism variants confer a greater susceptibility to MDD in Chinese Han population.
Collapse
Affiliation(s)
- Jiarun Yang
- Department of Medical Psychology, Institute of Public Health, Harbin Medical University, Harbin, China
| | - Xueyan Zhao
- Department of Medical Psychology, Institute of Public Health, Harbin Medical University, Harbin, China
| | - Jingsong Ma
- Department of Medical Psychology, Institute of Public Health, Harbin Medical University, Harbin, China
| | - Zhengxue Qiao
- Department of Medical Psychology, Institute of Public Health, Harbin Medical University, Harbin, China
| | - Xiuxian Yang
- Department of Medical Psychology, Institute of Public Health, Harbin Medical University, Harbin, China
| | - Erying Zhao
- Department of Medical Psychology, Institute of Public Health, Harbin Medical University, Harbin, China
| | - Bo Ban
- Department of Endocrinology, Affiliated Hosptial of Jining Medical University, Jining, China
| | - Xiongzhao Zhu
- Medical Psychological, Institute of the Second Xiangya Hospital of Central South University, Changsha, China
| | - Depin Cao
- Department of Medical Education Management, Harbin Medical University, Harbin, China
| | - Yanjie Yang
- Department of Medical Psychology, Institute of Public Health, Harbin Medical University, Harbin, China
| | - Xiaohui Qiu
- Department of Medical Psychology, Institute of Public Health, Harbin Medical University, Harbin, China
| |
Collapse
|
47
|
Tao S, Chattun MR, Yan R, Geng J, Zhu R, Shao J, Lu Q, Yao Z. TPH-2 Gene Polymorphism in Major Depressive Disorder Patients With Early-Wakening Symptom. Front Neurosci 2018; 12:827. [PMID: 30519155 PMCID: PMC6251472 DOI: 10.3389/fnins.2018.00827] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 10/23/2018] [Indexed: 12/13/2022] Open
Abstract
Background: Sleep disturbances, such as early wakening, are frequently observed in patients with major depressive disorder (MDD). The suprachiasmatic nuclei (SCN), which controls circadian rhythm, is innervated by the raphe nucleus, a region where Tryptophan hydroxylase-2 (TPH-2) gene is primarily expressed. Although TPH-2 is often implicated in the pathophysiology of depression, few studies have applied a genetic and imaging technique to investigate the mechanism of early wakening symptom in MDD. We hypothesized that TPH-2 variants could influence the function of SCN in MDD patients with early wakening symptom. Methods: One hundred and eighty five MDD patients (62 patients without early wakening and 123 patients with early wakening) and 64 healthy controls participated in this study. Blood samples were collected and genotyping of rs4290270, rs4570625, rs11178998, rs7305115, rs41317118, and rs17110747 were performed by next-generation sequencing (NGS) technology. Logistic regression model was employed for genetic data analysis using the PLINK software. Based on the allele type, rs4290270, which was significant in the early wakening MDD group, participants were categorized into two groups (A allele and T carrier). All patients underwent whole brain resting-state functional magnetic resonance imaging (rs-fMRI) scanning and a voxel-wise functional connectivity comparison was performed between the groups. Results: rs4290270 was significantly linked to MDD patients who exhibited early wakening symptom. The functional connectivities of the right SCN with the right fusiform gyrus and right middle frontal gyrus were increased in the T carrier group compared to the A allele group. In addition, the functional connectivities of the left SCN with the right lingual gyrus and left calcarine sulcus were decreased in the T carrier group compared to the A allele group. Conclusion: These findings suggested that the TPH-2 gene variant, rs4290270, affected the circadian regulating function of SCN. The altered functional connectivities, observed between the SCN and right fusiform gyrus, right middle frontal gyrus, the right lingual gyrus and left calcarine sulcus, could highlight the neural mechanism by which SCN induces sleep-related circadian disruption in T carrier MDD patients. Hence, rs4290270 could potentially serve as a reliable biomarker to identify MDD patients with early wakening symptom.
Collapse
Affiliation(s)
- Shiwan Tao
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Mohammad Ridwan Chattun
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Yan
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jiting Geng
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Rongxin Zhu
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Junneng Shao
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China.,Key Laboratory of Child Development and Learning Science, Southeast University, Nanjing, China
| | - Qing Lu
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China.,Key Laboratory of Child Development and Learning Science, Southeast University, Nanjing, China
| | - Zhijian Yao
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.,Medical School of Nanjing University, Nanjing Brain Hospital, Nanjing, China
| |
Collapse
|
48
|
de Abreu MS, Friend AJ, Demin KA, Amstislavskaya TG, Bao W, Kalueff AV. Zebrafish models: do we have valid paradigms for depression? J Pharmacol Toxicol Methods 2018; 94:16-22. [DOI: 10.1016/j.vascn.2018.07.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 06/12/2018] [Accepted: 07/16/2018] [Indexed: 11/26/2022]
|
49
|
Belmer A, Quentin E, Diaz SL, Guiard BP, Fernandez SP, Doly S, Banas SM, Pitychoutis PM, Moutkine I, Muzerelle A, Tchenio A, Roumier A, Mameli M, Maroteaux L. Positive regulation of raphe serotonin neurons by serotonin 2B receptors. Neuropsychopharmacology 2018; 43:1623-1632. [PMID: 29453444 PMCID: PMC5983540 DOI: 10.1038/s41386-018-0013-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 01/10/2018] [Accepted: 01/17/2018] [Indexed: 01/03/2023]
Abstract
Serotonin is a neurotransmitter involved in many psychiatric diseases. In humans, a lack of 5-HT2B receptors is associated with serotonin-dependent phenotypes, including impulsivity and suicidality. A lack of 5-HT2B receptors in mice eliminates the effects of molecules that directly target serotonergic neurons including amphetamine derivative serotonin releasers, and selective serotonin reuptake inhibitor antidepressants. In this work, we tested the hypothesis that 5-HT2B receptors directly and positively regulate raphe serotonin neuron activity. By ex vivo electrophysiological recordings, we report that stimulation by the 5-HT2B receptor agonist, BW723C86, increased the firing frequency of serotonin Pet1-positive neurons. Viral overexpression of 5-HT2B receptors in these neurons increased their excitability. Furthermore, in vivo 5-HT2B-receptor stimulation by BW723C86 counteracted 5-HT1A autoreceptor-dependent reduction in firing rate and hypothermic response in wild-type mice. By a conditional genetic ablation that eliminates 5-HT2B receptor expression specifically and exclusively from Pet1-positive serotonin neurons (Htr2b 5-HTKO mice), we demonstrated that behavioral and sensitizing effects of MDMA (3,4-methylenedioxy-methamphetamine), as well as acute behavioral and chronic neurogenic effects of the antidepressant fluoxetine, require 5-HT2B receptor expression in serotonergic neurons. In Htr2b 5-HTKO mice, dorsal raphe serotonin neurons displayed a lower firing frequency compared to control Htr2b lox/lox mice as assessed by in vivo extracellular recordings and a stronger hypothermic effect of 5-HT1A-autoreceptor stimulation was observed. The increase in head-twitch response to DOI (2,5-dimethoxy-4-iodoamphetamine) further confirmed the lower serotonergic tone resulting from the absence of 5-HT2B receptors in serotonin neurons. Together, these observations indicate that the 5-HT2B receptor acts as a direct positive modulator of serotonin Pet1-positive neurons in an opposite way as the known 5-HT1A-negative autoreceptor.
Collapse
Affiliation(s)
- Arnauld Belmer
- 0000000121866389grid.7429.8INSERM UMR-S 839, 75005 Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, UPMC Univ Paris 6, 75005 Paris, France ,0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, 75005 Paris, France ,0000000089150953grid.1024.7Translational Research Institute, Queensland University of Technology, Brisbane, QLD 4059 Australia
| | - Emily Quentin
- 0000000121866389grid.7429.8INSERM UMR-S 839, 75005 Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, UPMC Univ Paris 6, 75005 Paris, France ,0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, 75005 Paris, France
| | - Silvina L. Diaz
- 0000000121866389grid.7429.8INSERM UMR-S 839, 75005 Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, UPMC Univ Paris 6, 75005 Paris, France ,0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, 75005 Paris, France ,grid.441705.3Instituto de Biología Celular y Neurociencia, Fac. de Cs. Exactas, Químicas y Naturales, Universidad de Morón, UBA-CONICET – Paraguay 2155, 3° piso, C1121ABG Buenos Aires, Argentina
| | - Bruno P. Guiard
- Research Center on Animal Cognition, Center for Integrative Biology, 31062 Toulouse, France ,0000 0001 0723 035Xgrid.15781.3aUniversité Paul Sabatier, 31062 Toulouse, France ,UMR5169 CNRS, 31062 Toulouse, France
| | - Sebastian P. Fernandez
- 0000000121866389grid.7429.8INSERM UMR-S 839, 75005 Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, UPMC Univ Paris 6, 75005 Paris, France ,0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, 75005 Paris, France ,0000 0004 0638 0649grid.429194.3IPMC – CNRS UMR7275 660 Route des Lucioles Sophia-Antipolis, 06560 Valbonne, France
| | - Stéphane Doly
- 0000000121866389grid.7429.8INSERM UMR-S 839, 75005 Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, UPMC Univ Paris 6, 75005 Paris, France ,0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, 75005 Paris, France ,grid.503334.2Université Clermont Auvergne, INSERM, NEURO-DOL, 63000 Clermont-Ferrand, France
| | - Sophie M. Banas
- 0000000121866389grid.7429.8INSERM UMR-S 839, 75005 Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, UPMC Univ Paris 6, 75005 Paris, France ,0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, 75005 Paris, France
| | - Pothitos M. Pitychoutis
- 0000000121866389grid.7429.8INSERM UMR-S 839, 75005 Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, UPMC Univ Paris 6, 75005 Paris, France ,0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, 75005 Paris, France ,0000 0001 2175 167Xgrid.266231.2Department of Biology and Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH USA
| | - Imane Moutkine
- 0000000121866389grid.7429.8INSERM UMR-S 839, 75005 Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, UPMC Univ Paris 6, 75005 Paris, France ,0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, 75005 Paris, France
| | - Aude Muzerelle
- 0000000121866389grid.7429.8INSERM UMR-S 839, 75005 Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, UPMC Univ Paris 6, 75005 Paris, France ,0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, 75005 Paris, France
| | - Anna Tchenio
- 0000000121866389grid.7429.8INSERM UMR-S 839, 75005 Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, UPMC Univ Paris 6, 75005 Paris, France ,0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, 75005 Paris, France ,0000 0001 2165 4204grid.9851.5Dept. Fundamental Neurosciences (DNF) The University of Lausanne, Lausanne, Switzerland
| | - Anne Roumier
- 0000000121866389grid.7429.8INSERM UMR-S 839, 75005 Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, UPMC Univ Paris 6, 75005 Paris, France ,0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, 75005 Paris, France
| | - Manuel Mameli
- 0000000121866389grid.7429.8INSERM UMR-S 839, 75005 Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Universités, UPMC Univ Paris 6, 75005 Paris, France ,0000 0004 0520 8345grid.462192.aInstitut du Fer à Moulin, 75005 Paris, France ,0000 0001 2165 4204grid.9851.5Dept. Fundamental Neurosciences (DNF) The University of Lausanne, Lausanne, Switzerland
| | - Luc Maroteaux
- INSERM UMR-S 839, 75005, Paris, France. .,Sorbonne Universités, UPMC Univ Paris 6, 75005, Paris, France. .,Institut du Fer à Moulin, 75005, Paris, France.
| |
Collapse
|
50
|
Willemze RA, Welting O, van Hamersveld HP, Meijer SL, Folgering JHA, Darwinkel H, Witherington J, Sridhar A, Vervoordeldonk MJ, Seppen J, de Jonge WJ. Neuronal control of experimental colitis occurs via sympathetic intestinal innervation. Neurogastroenterol Motil 2018; 30. [PMID: 28745812 DOI: 10.1111/nmo.13163] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/20/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Vagus nerve stimulation is currently clinically evaluated as a treatment for inflammatory bowel disease. However, the mechanism by which this therapeutic intervention can have an immune-regulatory effect in colitis remains unclear. We determined the effect of intestine-specific vagotomy or intestine-specific sympathectomy of the superior mesenteric nerve (SMN) on dextran sodium sulfate (DSS)-induced colitis in mice. Furthermore, we tested the efficacy of therapeutic SMN stimulation to treat DSS-induced colitis in rats. METHODS Vagal and SMN fibers were surgically dissected to achieve intestine-specific vagotomy and sympathectomy. Chronic SMN stimulation was achieved by implantation of a cuff electrode. Stimulation was done twice daily for 5 minutes using a biphasic pulse (10 Hz, 200 μA, 2 ms). Disease activity index (DAI) was used as a clinical parameter for colitis severity. Colonic cytokine expression was measured by quantitative PCR and ELISA. KEY RESULTS Intestine-specific vagotomy had no effect on DSS-induced colitis in mice. However, SMN sympathectomy caused a significantly higher DAI compared to sham-operated mice. Conversely, SMN stimulation led to a significantly improved DAI compared to sham stimulation, although no other parameters of colitis were affected significantly. CONCLUSIONS & INFERENCES Our results indicate that sympathetic innervation regulates the intestinal immune system as SMN denervation augments, and SMN stimulation ameliorates DSS-induced colitis. Surprisingly, intestine-specific vagal nerve denervation had no effect in DSS-induced colitis.
Collapse
Affiliation(s)
- R A Willemze
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - O Welting
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - H P van Hamersveld
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - S L Meijer
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | | | - H Darwinkel
- Brains On-Line B.V., Groningen, The Netherlands
| | | | - A Sridhar
- Galvani Bioelectronics, Stevenage, UK
| | | | - J Seppen
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - W J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|