1
|
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
|
2
|
Fang Y, Li Y, Liao X, Deng J, Wang Q, Liang J, Yan B. Corydalis yanhusuo Polysaccharides Ameliorate Chronic Stress-Induced Depression in Mice through Gut Microbiota-Derived Short-Chain Fatty Acid Activation of 5-Hydroxytryptamine Signaling. J Med Food 2023; 26:890-901. [PMID: 38010856 DOI: 10.1089/jmf.2023.k.0050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023] Open
Abstract
Depression, a prevalent psychiatric disorder, presents a serious health risk to humans. Increasing evidence suggested that the gut microbiota and the 5-hydroxytryptamine (5-HT) pathway both contribute significantly to depression. This research aimed to investigate how Corydalis yanhusuo polysaccharides (CYP) could potentially alleviate depression induced by chronic unpredictable mild stress in mice, as well as its underlying mechanism. The sucrose preference test, tail suspension test, and forced swimming test were employed to evaluate the behavior of mice. Enzyme-linked immunosorbent assay and PCR techniques were utilized to measure depression-related factors (dopamine [DA], 5-HT, norepinephrine [NE], brain-derived neurotrophic factor [BDNF], tryptophan hydroxylase 2 [TPH-2], 5-hydroxytryptophan [5-HTP], and tryptophan hydroxylase [TPH-1] levels). Hematoxylin and eosin staining and Nissl staining were conducted to observe histopathological changes in the hippocampus, the differences in the diversity of gut flora between groups were analyzed using 16S rRNA sequencing, and gas chromatography-mass spectrometry metabolomics was utilized to evaluate short-chain fatty acid (SCFA) concentrations. The findings indicated that CYP treatment increased the sucrose preference index, decreased the immobility time, and improved neuropathological injury. In depressed mice, CYP improved the dysregulation of the gut microbiota, and increased the SCFA levels. In addition, CYP enhanced the DA, 5-HT, NE, BDNF, and TPH-2 levels in the brain and the expression of 5-HTP and TPH-1 in the colon, while SCFAs were positively correlated with these levels. In summary, our study suggested that CYP may mitigate depression by ameliorating gut microbiota dysregulation, promoting the generation of SCFAs, and activation of 5-HT signaling expression.
Collapse
Affiliation(s)
- Yu Fang
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanqing Li
- Department of TCM & Western Medicine, Chengdu University of Traditional Chinese Medicine/Affiliated Reproductive Maternity and Child Hospital, Chengdu, China
| | - Xin Liao
- Department of Information, Hospital of Chengdu University of TCM, Chengdu, China
| | - Jie Deng
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiannan Wang
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingtao Liang
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bohua Yan
- Department of GCP, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
3
|
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
|
4
|
Furukawa M, Tada H, Raju R, Wang J, Yokoi H, Yamada M, Shikama Y, Matsushita K. Long-Term Soft-Food Rearing in Young Mice Alters Brain Function and Mood-Related Behavior. Nutrients 2023; 15:2397. [PMID: 37242280 PMCID: PMC10222696 DOI: 10.3390/nu15102397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/15/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
The relationship between caloric and nutrient intake and overall health has been extensively studied. However, little research has focused on the impact of the hardness of staple foods on health. In this study, we investigated the effects of a soft diet on brain function and behavior in mice from an early age. Mice fed a soft diet for six months exhibited increased body weight and total cholesterol levels, along with impaired cognitive and motor function, heightened nocturnal activity, and increased aggression. Interestingly, when these mice were switched back to a solid diet for three months, their weight gain ceased, total cholesterol levels stabilized, cognitive function improved, and aggression decreased, while their nocturnal activity remained high. These findings suggest that long-term consumption of a soft diet during early development can influence various behaviors associated with anxiety and mood regulation, including weight gain, cognitive decline, impaired motor coordination, increased nocturnal activity, and heightened aggression. Therefore, the hardness of food can impact brain function, mental well-being, and motor skills during the developmental stage. Early consumption of hard foods may be crucial for promoting and maintaining healthy brain function.
Collapse
Affiliation(s)
- Masae Furukawa
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan; (R.R.); (J.W.); (H.Y.); (M.Y.); (Y.S.)
| | - Hirobumi Tada
- Department of Nutrition, Faculty of Wellness, Shigakkan University, Obu 474-8651, Japan;
- Department of Integrative Physiology, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Resmi Raju
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan; (R.R.); (J.W.); (H.Y.); (M.Y.); (Y.S.)
| | - Jingshu Wang
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan; (R.R.); (J.W.); (H.Y.); (M.Y.); (Y.S.)
| | - Haruna Yokoi
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan; (R.R.); (J.W.); (H.Y.); (M.Y.); (Y.S.)
| | - Mitsuyoshi Yamada
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan; (R.R.); (J.W.); (H.Y.); (M.Y.); (Y.S.)
- Department of Operative Dentistry, School of Dentistry, Aichi Gakuin University, Nagoya 464-8651, Japan
| | - Yosuke Shikama
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan; (R.R.); (J.W.); (H.Y.); (M.Y.); (Y.S.)
| | - Kenji Matsushita
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan; (R.R.); (J.W.); (H.Y.); (M.Y.); (Y.S.)
| |
Collapse
|
5
|
Kryst J, Majcher-Maślanka I, Chocyk A. Effects of chronic fluoxetine treatment on anxiety- and depressive-like behaviors in adolescent rodents - systematic review and meta-analysis. Pharmacol Rep 2022; 74:920-946. [PMID: 36151445 PMCID: PMC9584991 DOI: 10.1007/s43440-022-00420-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 12/09/2022]
Abstract
Background Drugs prescribed for psychiatric disorders in adolescence should be studied very extensively since they can affect developing and thus highly plastic brain differently than they affect the adult brain. Therefore, we aimed to summarize animal studies reporting the behavioral consequences of chronic exposure to the most widely prescribed antidepressant drug among adolescents i.e., fluoxetine. Methods Electronic databases (Medline via Pubmed, Web of Science Core Collection, ScienceDirect) were systematically searched until April 12, 2022, for published, peer-reviewed, controlled trials concerning the effects of chronic fluoxetine administration vs. vehicle on anxiety and depression measures in naïve and stress-exposed adolescent rodents. All of the relevant studies were selected and critically appraised, and a meta-analysis of eligible studies was performed. Results A total of 18 studies were included in the meta-analysis. In naïve animals, chronic adolescent fluoxetine administration showed dose-related anxiogenic-like effects, measured as a reduction in time spent in the open arms of the elevated plus maze. No significant effects of chronic adolescent fluoxetine on depression-like behavior were reported in naïve animals, while in stress-exposed rodents chronic adolescent fluoxetine significantly decreased immobility time in the forced swim test compared to vehicle. Conclusions These results suggest that although chronic fluoxetine treatment proves positive effects in animal models of depression, it may simultaneously increase anxiety in adolescent animals in a dose-related manner. Although the clinical implications of the data should be interpreted with extreme caution, adolescent patients under fluoxetine treatment should be closely monitored. Supplementary Information The online version contains supplementary material available at 10.1007/s43440-022-00420-w.
Collapse
Affiliation(s)
- Joanna Kryst
- Faculty of Physiotherapy, Institute for Basics Sciences, University of Physical Education, Jana Pawła II Av. 78, 31-571, Kraków, Poland
| | - Iwona Majcher-Maślanka
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - Agnieszka Chocyk
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland.
| |
Collapse
|
6
|
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
|
7
|
Maratha S, Sharma V, Walia V. Possible involvement of NO-sGC-cGMP signaling in the antidepressant like effect of pyridoxine in mice. Metab Brain Dis 2022; 37:173-183. [PMID: 34739660 DOI: 10.1007/s11011-021-00858-6] [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: 08/23/2021] [Accepted: 10/23/2021] [Indexed: 11/28/2022]
Abstract
The present study was designed to determine the antidepressant like effect of pyridoxine in mice. Pyridoxine (12.5, 25 and 50 mg/kg, i.p.) was administered to the mice and depression related behavioral and neurochemical alterations were determined. It was observed that pyridoxine (50 mg/kg, i.p.) treatment decreased the immobility period in tail suspension test (TST) and forced swim test (FST) significantly as compared to control. Pyridoxine (50 mg/kg, i.p.) treatment increased the level of serotonin (5-HT) and decreased the level of nitrite in the brain of mice significantly as compared to control. Pyridoxine thus confer antidepressant like effect by increasing the level of 5-HT and by decreasing the level of nitrite in the brain of mice. Further the influence of nitric oxide (NO)/ soluble guanylate cyclase (sGC)/ cyclic guanosine monophosphate (cGMP) in antidepressant-like effect of pyridoxine was studied. It was observed that the pretreatment of NO donor (i.e. L-Arginine) and cGMP modulator (i.e. sildenafil) counteracted while the pretreatment of NO/sGC inhibitor (i.e. methylene blue) potentiated the effect of pyridoxine in TST and FST. Pretreatment of NO donor did not influence, pretreatment of NO/sGC inhibitor decreased while the pretreatment of cGMP modulator increased the level of brain nitrite in pyridoxine treated mice. Further the pretreatment of NO donor and cGMP modulator decreased while the pretreatment of NO/sGC inhibitor increased the level of brain serotonin in pyridoxine treated mice. Pyridoxine thus exerted antidepressant like effect and NO-sGC-cGMP signaling modulated the antidepressant like effect of pyridoxine in mice.
Collapse
Affiliation(s)
- Sushma Maratha
- SGT College of Pharmacy, SGT University, Gurugram, India
| | - Vijay Sharma
- SGT College of Pharmacy, SGT University, Gurugram, India
| | - Vaibhav Walia
- SGT College of Pharmacy, SGT University, Gurugram, India.
| |
Collapse
|
8
|
Maratha S, Sharma V, Walia V. Antidepressant Like Effect of Ascorbic Acid in Mice: Possible Involvement of NO-sGC-cGMP Signaling. Neurochem Res 2021; 47:967-978. [PMID: 34825298 DOI: 10.1007/s11064-021-03496-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 10/19/2022]
Abstract
The present study was designed to determine the antidepressant like activity of ascorbic acid (AA) in mice. Further the influence of NO-sGC-cGMP signaling in the antidepressant like effect of AA in mice was determined. Male swiss albino mice were used in the present study. Mice in the control group received saline and fluoxetine (10 mg/kg, i.p.) was used as the standard antidepressant drug. AA (50, 100 and 150 mg/kg, i.p.) was administered to the mice and depression related behavior were determined using tail suspension test (TST) and forced swim test (FST). Further the whole brain nitrite and serotonin levels were also determined. It was observed that the administration of AA (100 mg/kg, i.p.) reversed the depression like behavior in mice in TST and FST. AA (100 mg/kg, i.p.) treatment decreased the level of nitrite and increased the level of serotonin in the brain of mice significantly as compared to control. Further the behavioral and neurochemical effect of AA (50 mg/kg, i.p) was studied in NO modulator [NO donor: L-Arginine (50 mg/kg, i.p); NO-sGC inhibitor: methylene blue (1 mg/kg, i.p.) and cGMP modulator: sildenafil (1 mg/kg, i.p.)] pretreated mice. It was observed that the pretreatment of NO donor and cGMP modulator counteracted the effect conferred by AA (50 mg/kg, i.p). While the pretreatment of NO-sGC inhibitor potentiated the effect conferred by AA (50 mg/kg, i.p). The present study suggested that the AA confer antidepressant like effect in mice and NO-sGC-cGMP signaling pathway influence the antidepressant like effect of AA in mice.
Collapse
Affiliation(s)
- Sushma Maratha
- SGT College of Pharmacy, SGT University, Gurugram, India
| | - Vijay Sharma
- SGT College of Pharmacy, SGT University, Gurugram, India
| | - Vaibhav Walia
- SGT College of Pharmacy, SGT University, Gurugram, India.
| |
Collapse
|
9
|
Villani C, Carli M, Castaldo AM, Sacchetti G, Invernizzi RW. Fluoxetine increases brain MeCP2 immuno-positive cells in a female Mecp2 heterozygous mouse model of Rett syndrome through endogenous serotonin. Sci Rep 2021; 11:14690. [PMID: 34282222 PMCID: PMC8290043 DOI: 10.1038/s41598-021-94156-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022] Open
Abstract
Motor skill deficit is a common and invalidating symptom of Rett syndrome (RTT), a rare disease almost exclusively affecting girls during the first/second year of life. Loss-of-function mutations of the methyl-CpG-binding protein2 (MECP2; Mecp2 in rodents) gene is the cause in most patients. We recently found that fluoxetine, a selective serotonin (5-HT) reuptake inhibitor and antidepressant drug, fully rescued motor coordination deficits in Mecp2 heterozygous (Mecp2 HET) mice acting through brain 5-HT. Here, we asked whether fluoxetine could increase MeCP2 expression in the brain of Mecp2 HET mice, under the same schedule of treatment improving motor coordination. Fluoxetine increased the number of MeCP2 immuno-positive (MeCP2+) cells in the prefrontal cortex, M1 and M2 motor cortices, and in dorsal, ventral and lateral striatum. Fluoxetine had no effect in the CA3 region of the hippocampus or in any of the brain regions of WT mice. Inhibition of 5-HT synthesis abolished the fluoxetine-induced rise of MeCP2+ cells. These findings suggest that boosting 5-HT transmission is sufficient to enhance the expression of MeCP2 in several brain regions of Mecp2 HET mice. Fluoxetine-induced rise of MeCP2 could potentially rescue motor coordination and other deficits of RTT.
Collapse
Affiliation(s)
- Claudia Villani
- Laboratory Neurochemistry and Behavior, Neuroscience Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Mirjana Carli
- Laboratory Neurochemistry and Behavior, Neuroscience Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Anna Maria Castaldo
- Laboratory Neurochemistry and Behavior, Neuroscience Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Giuseppina Sacchetti
- Laboratory Neurochemistry and Behavior, Neuroscience Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Roberto William Invernizzi
- Laboratory Neurochemistry and Behavior, Neuroscience Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Kesić M, Mokrović G, Tvrdeić A, Miše B, Štefulj J, Čičin-Šain L. Constitutive Serotonin Tone Modulates Molecular and Behavioral Response to Chronic Fluoxetine Treatment: A Study on Genetic Rat Model. Front Psychiatry 2021; 12:741222. [PMID: 34658974 PMCID: PMC8517234 DOI: 10.3389/fpsyt.2021.741222] [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: 07/14/2021] [Accepted: 09/06/2021] [Indexed: 11/16/2022] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the most commonly prescribed medications for the treatment of mood disorders. Yet, individual response to SSRIs is highly variable, with only a portion of patients showing the desired therapeutic effect. To better understand the molecular basis underlying individual variability in response to SSRIs, here we comparatively studied behavioral and molecular consequences of chronic treatment with fluoxetine, a widely used SSRI, in two sublines of rats with constitutionally different serotonin (5HT) homeostasis: the high-5HT and low-5HT sublines. Platelet 5HT levels, a recognized indicator of SSRI efficacy, were decreased by fluoxetine treatment in both 5HT-sublines. On the other hand, biologically active plasma 5HT levels were reduced only in high-5HT rats. The anxiolytic effect of fluoxetine was also evident only in high-5HT rats, as supported by spatio-temporal and ethological behavioral measures in the elevated plus maze (EPM) test and exploratory behavior measures in the open field (OF) test. None of the behavioral EPM or OF measures were significantly altered by fluoxetine treatment in low-5HT rats. Unexpectedly, 5HT levels in cerebral cortices tended to be reduced only in low-5HT rats. Moreover, the effects of fluoxetine on cortical expression levels of 5HT-related proteins were also present only in low-5HT rats, with serotonin transporter (5HTT) and serotonin receptor type 1a (Htr1a) being down-regulated, while serotonin receptor type 4 (Htr4) was up-regulated by fluoxetine treatment. The obtained results support a role of individual 5HT tone as an important influencing factor on the biological actions of SSRI antidepressants.
Collapse
Affiliation(s)
- Maja Kesić
- Laboratory for Neurochemistry and Molecular Neurobiology, Department of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Gordana Mokrović
- Laboratory for Neurochemistry and Molecular Neurobiology, Department of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Ante Tvrdeić
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Branko Miše
- University Hospital for Infectious Diseases, Zagreb, Croatia
| | - Jasminka Štefulj
- Laboratory for Neurochemistry and Molecular Neurobiology, Department of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Lipa Čičin-Šain
- Laboratory for Neurochemistry and Molecular Neurobiology, Department of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
13
|
Foltran RB, Stefani KM, Höcht C, Diaz SL. Neurochemical, Behavioral, and Neurogenic Validation of a Hyposerotonergic Animal Model by Voluntary Oral Consumption of para-Chlorophenylalanine. ACS Chem Neurosci 2020; 11:952-959. [PMID: 32107912 DOI: 10.1021/acschemneuro.9b00687] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The inhibitor of tryptophan hydroxylase, para-chlorophenylalanine (PCPA), has been classically employed as a pharmacological tool to deplete serotonin (5-HT) in animal models and to evaluate whether this neurotransmitter is involved in the action of pharmacological compounds. PCPA is usually administrated by intraperitoneal (ip) injections, which are stressful and painful. To avoid ip injections, we designed and validated a protocol for PCPA oral administration. C57BL/6 elite male mice received PCPA during 7 days either ip or by giving the drug inside jelly cubes at an estimated dose of 500 mg/kg on days 1 and 2 and 250 mg/kg for the rest of the treatment. 5-HT levels decreased by 85% and 55% in the hippocampus of mice treated with oral or ip PCPA, respectively, whereas in the prefrontal cortex, 5-HT levels decreased by 65% (oral) and 50% (ip). Behavioral tests, like the forced swimming test (FST), the nestlet shredding test (NST), and the marble burying test (MBT), were performed. In the FST, mice received fluoxetine ip 30 min before the test. In mice with oral PCPA treatment, fluoxetine did not induce significant reductions of immobility, indicating that reduction of 5-HT levels was effective. No effect of ip or oral 5-HT depletion was observed in the NST nor in the MBT. In a second experiment, mice received oral PCPA for 8 weeks: again, serotonin levels were significantly decreased in both hippocampus and cortex, and effects on hippocampal neurogenesis replicated previous observations in hyposerotonergic mice. Therefore, neurochemical, behavioral, and neurogenic results allow us to validate this refined protocol for voluntary oral consumption of PCPA.
Collapse
Affiliation(s)
- Rocío B. Foltran
- Instituto de Biología Celular y Neurociencias Prof. E. De Robertis, CONICET-UBA, Paraguay 2155,
3rd floor, C1121ABG Buenos Aires, Argentina
| | - Karen M. Stefani
- Instituto de Biología Celular y Neurociencias Prof. E. De Robertis, CONICET-UBA, Paraguay 2155,
3rd floor, C1121ABG Buenos Aires, Argentina
| | - Christian Höcht
- Cátedra de Farmacología, Fac. de Farmacia y Bioquímica, UBA. Junin 956, 5th
floor, C1113AAD Buenos Aires, Argentina
| | - Silvina L. Diaz
- Instituto de Biología Celular y Neurociencias Prof. E. De Robertis, CONICET-UBA, Paraguay 2155,
3rd floor, C1121ABG Buenos Aires, Argentina
| |
Collapse
|
14
|
Neuron-glia interaction through Serotonin-BDNF-NGFR axis enables regenerative neurogenesis in Alzheimer's model of adult zebrafish brain. PLoS Biol 2020; 18:e3000585. [PMID: 31905199 PMCID: PMC6964913 DOI: 10.1371/journal.pbio.3000585] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 01/16/2020] [Accepted: 12/12/2019] [Indexed: 11/23/2022] Open
Abstract
It was recently suggested that supplying the brain with new neurons could counteract Alzheimer’s disease (AD). This provocative idea requires further testing in experimental models in which the molecular basis of disease-induced neuronal regeneration could be investigated. We previously found that zebrafish stimulates neural stem cell (NSC) plasticity and neurogenesis in AD and could help to understand the mechanisms to be harnessed for developing new neurons in diseased mammalian brains. Here, by performing single-cell transcriptomics, we found that amyloid toxicity-induced interleukin-4 (IL4) promotes NSC proliferation and neurogenesis by suppressing the tryptophan metabolism and reducing the production of serotonin. NSC proliferation was suppressed by serotonin via down-regulation of brain-derived neurotrophic factor (BDNF)-expression in serotonin-responsive periventricular neurons. BDNF enhances NSC plasticity and neurogenesis via nerve growth factor receptor A (NGFRA)/ nuclear factor 'kappa-light-chain-enhancer' of activated B-cells (NFkB) signaling in zebrafish but not in rodents. Collectively, our results suggest a complex neuron-glia interaction that regulates regenerative neurogenesis after AD conditions in zebrafish. Can regeneration of lost neurons counteract neurodegenerative disease? This study shows that serotonergic neurons alter neural stem cell proliferation and neurogenesis via a complex neuron-glia interaction involving interleukin-4, BDNF and NGF receptor in a zebrafish model of Alzheimer's disease.
Collapse
|
15
|
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
|
16
|
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
|
17
|
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
|
18
|
Foltran RB, Stefani KM, Bonafina A, Resasco A, Diaz SL. Differential Hippocampal Expression of BDNF Isoforms and Their Receptors Under Diverse Configurations of the Serotonergic System in a Mice Model of Increased Neuronal Survival. Front Cell Neurosci 2019; 13:384. [PMID: 31555094 PMCID: PMC6712164 DOI: 10.3389/fncel.2019.00384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 08/06/2019] [Indexed: 12/14/2022] Open
Abstract
Neurotrophic factors are relevant regulators of the neurogenic process at different levels. In particular, the brain-derived neurotrophic factor, BDNF, is highly expressed in the hippocampus (HC) of rodents and participates in the control of neuronal proliferation, and survival in the dentate gyrus (DG). Likewise, serotonin is also involved in the regulation of neurogenesis, though its role is apparently more complex. Indeed, both enhancement of serotonin neurotransmission as well as serotonin depletion, paradoxically increase neuronal survival in the HC of mice. In this study, we analyzed the protein expression of the BDNF isoforms, i.e., pro- and mature-BDNF, and their respective receptors p75 and TrkB, in the HC of mice chronically treated with para-chloro-phenyl-alanine (PCPA), an inhibitor of serotonin synthesis. The same analysis was conducted in hyposerotonergic mice with concomitant administration of the 5-HT1A receptor agonist, 8-Hydroxy-2-(di-n- propylamino) tetralin (8-OH-DPAT). Increased expression of p75 receptor with decreased expression of pro-BDNF was observed after chronic PCPA. Seven-day treatment with 8-OH-DPAT reestablished the expression of pro-BDNF modified by PCPA, and induced an increase in the expression of p75 receptor. It has been demonstrated that PCPA-treated mice have higher number of immature neurons in the HC. Given that immature neurons participate in the pattern separation process, the object pattern separation test was conducted. A better performance of hyposerotonergic mice was not confirmed in this assay. Altogether, our results show that molecules in the BDNF signaling pathway are differentially expressed under diverse configurations of the serotonergic system, allowing for fine-tuning of the neurogenic process.
Collapse
Affiliation(s)
- Rocío Beatriz Foltran
- Instituto de Biología Celular y Neurociencia Prof. E. De Robertis, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Karen Melany Stefani
- Instituto de Biología Celular y Neurociencia Prof. E. De Robertis, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Antonela Bonafina
- Instituto de Biología Celular y Neurociencia Prof. E. De Robertis, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Agustina Resasco
- Instituto de Biología Celular y Neurociencia Prof. E. De Robertis, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Silvina Laura Diaz
- Instituto de Biología Celular y Neurociencia Prof. E. De Robertis, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
19
|
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
|
20
|
Drozd US, Shaburova EV, Dygalo NN. Genetic approaches to the investigation of serotonergic neuron functions in animals. Vavilovskii Zhurnal Genet Selektsii 2019. [DOI: 10.18699/vj19.513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The serotonergic system is one of the most important neurotransmitter systems that take part in the regulation of vital CNS functions. The understanding of its mechanisms will help scientists create new therapeutic approaches to the treatment of mental and neurodegenerative diseases and find out how this neurotransmitter system interacts with other parts of the brain and regulates their activity. Since the serotonergic system anatomy and functionality are heterogeneous and complex, the best tools for studying them are based on manipulation of individual types of neurons without affecting neurons of other neurotransmitter systems. The selective cell control is possible due to the genetic determinism of their functions. Proteins that determine the uniqueness of the cell type are expressed under the regulation of cell-specific promoters. By using promoters that are specific for genes of the serotonin system, one can control the expression of a gene of interest in serotonergic neurons. Here we review approaches based on such promoters. The genetic models to be discussed in the article have already shed the light on the role of the serotonergic system in modulating behavior and processing sensory information. In particular, genetic knockouts of serotonin genes sert, pet1, and tph2 promoted the determination of their contribution to the development and functioning of the brain. In addition, the review describes inducible models that allow gene expression to be controlled at various developmental stages. Finally, the application of these genetic approaches in optogenetics and chemogenetics provided a new resource for studying the functions, discharge activity, and signal transduction of serotonergic neurons. Nevertheless, the advantages and limitations of the discussed genetic approaches should be taken into consideration in the course of creating models of pathological conditions and developing pharmacological treatments for their correction.
Collapse
Affiliation(s)
- U. S. Drozd
- Novosibirsk State University; Institute of Cytology and Genetics, SB RAS
| | - E. V. Shaburova
- Novosibirsk State University; Institute of Cytology and Genetics, SB RAS
| | - N. N. Dygalo
- Novosibirsk State University; Institute of Cytology and Genetics, SB RAS
| |
Collapse
|
21
|
Pratelli M, Pasqualetti M. Serotonergic neurotransmission manipulation for the understanding of brain development and function: Learning from Tph2 genetic models. Biochimie 2019; 161:3-14. [DOI: 10.1016/j.biochi.2018.11.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/24/2018] [Indexed: 01/04/2023]
|
22
|
Abstract
Selective serotonin reuptake inhibitors are among the most prescribed antidepressants. Fluoxetine is the lead molecule which exerts its therapeutic effects, at least in part, by promoting neuroplasticity through increased brain-derived neurotrophic factor (BDNF)/tropomyosin-related receptor kinase B (TrkB) signalling. It is unclear however, to which extent the neuroplastic effects of fluoxetine are solely mediated by the inhibition of the serotonin transporter (5-HTT). To answer this question, the effects of fluoxetine on neuroplasticity were analysed in both wild type (WT) and 5-Htt knock-out (KO) mice. Using Western blotting and RT-qPCR approaches, we showed that fluoxetine 10 µM activated BDNF/TrkB signalling pathways in both CD1 and C57BL/6J mouse primary cortical neurons. Interestingly, effects on BDNF signalling were observed in primary cortical neurons from both 5-Htt WT and KO mice. In addition, a 3-week in vivo fluoxetine treatment (15 mg/kg/d; i.p.) increased the expression of plasticity genes in brains of both 5-Htt WT and KO mice, and tended to equally enhance hippocampal cell proliferation in both genotypes, without reaching significance. Our results further suggest that fluoxetine-induced neuroplasticity does not solely depend on 5-HTT blockade, but might rely, at least in part, on 5-HTT-independent direct activation of TrkB.
Collapse
|
23
|
Sachs BD, Tran HL, Folse E, Caron MG. Brain-region-specific Molecular Responses to Maternal Separation and Social Defeat Stress in Mice. Neuroscience 2018; 373:122-136. [PMID: 29341883 DOI: 10.1016/j.neuroscience.2018.01.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 01/03/2018] [Accepted: 01/04/2018] [Indexed: 01/14/2023]
Abstract
The association between stress and mental illness has been well documented, but the molecular consequences of repeated exposure to stress have not been completely identified. The present study sought to elucidate the combinatorial effects of early-life maternal separation stress and adult social defeat stress on alterations in signal transduction and gene expression that have been previously implicated in susceptibility to psychosocial stress. Molecular analyses were performed in the prelimbic/infralimbic cortex, amygdala, and nucleus accumbens, three brain regions that have been suggested to play critical roles in determining stress responses. The current data reveal that both maternal separation and social defeat significantly impact the expression of genes involved in histone methylation and the β-catenin-, endogenous opioid-, neurotrophin-, and glucocorticoid signaling pathways. Although the effects of maternal separation and social defeat were largely non-overlapping, a subset of genes in each brain region were governed by additive, opposing, or other types of interactions between these stress paradigms, thus highlighting potential molecular mechanisms through which these stressors might coordinately regulate brain function and behavior.
Collapse
Affiliation(s)
- Benjamin D Sachs
- Duke University Medical Center, Department of Cell Biology, Durham, NC 27710, United States; Villanova University, Department of Psychological and Brain Sciences, Villanova, PA 19085, United States.
| | - Ha L Tran
- Duke University Medical Center, Department of Cell Biology, Durham, NC 27710, United States
| | - Emily Folse
- Villanova University, Department of Psychological and Brain Sciences, Villanova, PA 19085, United States
| | - Marc G Caron
- Duke University Medical Center, Department of Cell Biology, Durham, NC 27710, United States; Duke University Medical Center, Department of Neurobiology, Durham, NC 27710, United States; Duke University Medical Center, Department of Medicine, Durham, NC 27710, United States
| |
Collapse
|
24
|
Hong LZ, Huang KF, Hung SW, Kuo LT. Chronic fluoxetine treatment enhances sympathetic activities associated with abnormality of baroreflex function in conscious normal rats. Eur J Pharmacol 2017. [DOI: 10.1016/j.ejphar.2017.06.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
25
|
Hashizume K, Yamanaka M, Ueda S. POU3F2 participates in cognitive function and adult hippocampal neurogenesis via mammalian-characteristic amino acid repeats. GENES BRAIN AND BEHAVIOR 2017; 17:118-125. [PMID: 28782255 DOI: 10.1111/gbb.12408] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 07/02/2017] [Accepted: 08/03/2017] [Indexed: 12/15/2022]
Abstract
POU3F2/BRN-2 is a transcription factor that is mainly expressed in the central nervous system and plays an important role in brain development. The transactivation domain of POU3F2 includes multiple mammalian-characteristic tandem amino acid repeats (homopolymeric amino acid repeats). We previously generated knock-in mice (Pou3f2Δ/Δ mice) in which all three homopolymeric amino acid repeats were deleted from the Pou3f2 transactivation domain and identified phenotypic impairments in maternal behavior and pup recognition. Yet, the exact biological implications of homopolymeric repeats are not completely understood. In this study, we investigated cognitive function and hippocampal neurogenesis in Pou3f2Δ/Δ mice. Pou3f2Δ/Δ mice exhibited cognitive impairment in object recognition and object location tests. Immunohistochemistry for doublecortin, a marker of immature neurons, showed a lower number of newborn neurons in the dentate gyrus of adult Pou3f2Δ/Δ mice compared with wild-type mice. Consistent with this observation, adult Pou3f2Δ/Δ mice had lower numbers of 5-bromo-2'-deoxyuridine (BrdU) and NeuN double-positive cells at 4 weeks after BrdU injection compared with control mice, indicating the decreased generation of mature granule cells in Pou3f2Δ/Δ mice. Taken together, these results suggest that POU3F2 is involved in cognitive function as well as adult hippocampal neurogenesis, and that homopolymeric amino acid repeats in this gene play a functional role.
Collapse
Affiliation(s)
- K Hashizume
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - M Yamanaka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - S Ueda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
26
|
Foltran RB, Diaz SL. BDNF isoforms: a round trip ticket between neurogenesis and serotonin? J Neurochem 2016; 138:204-21. [PMID: 27167299 DOI: 10.1111/jnc.13658] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/08/2016] [Accepted: 05/02/2016] [Indexed: 12/12/2022]
Abstract
The brain-derived neurotrophic factor, BDNF, was discovered more than 30 years ago and, like other members of the neurotrophin family, this neuropeptide is synthetized as a proneurotrophin, the pro-BDNF, which is further cleaved to yield mature BDNF. The myriad of actions of these two BDNF isoforms in the central nervous system is constantly increasing and requires the development of sophisticated tools and animal models to refine our understanding. This review is focused on BDNF isoforms, their participation in the process of neurogenesis taking place in the hippocampus of adult mammals, and the modulation of their expression by serotonergic agents. Interestingly, around this triumvirate of BDNF, serotonin, and neurogenesis, a series of recent research has emerged with apparently counterintuitive results. This calls for an exhaustive analysis of the data published so far and encourages thorough work in the quest for new hypotheses in the field. BDNF is synthetized as a pre-proneurotrophin. After removal of the pre-region, proBDNF can be cleaved by intracellular or extracellular proteases. Mature BDNF can bind TrkB receptors, promoting their homodimerization and intracellular phosphorylation. Phosphorylated-TrkB can activate three different signaling pathways. Whereas G-protein-coupled receptors can transactivate TrkB receptors, truncated forms can inhibit mBDNF signaling. Pro-BDNF binds p75(NTR) by its mature domain, whereas the pro-region binds co-receptors.
Collapse
Affiliation(s)
- Rocío Beatriz Foltran
- Instituto de Biología Celular y Neurociencias Prof. E. De Robertis, CONICET-UBA, Fac. de Medicina - UBA, Buenos Aires, Argentina
| | - Silvina Laura Diaz
- Instituto de Biología Celular y Neurociencias Prof. E. De Robertis, CONICET-UBA, Fac. de Medicina - UBA, Buenos Aires, Argentina
| |
Collapse
|
27
|
Ibeneme SC, Anyachukwu CC, Nwosu A, Ibeneme GC, Bakare M, Fortwengel G. Symptoms of Poststroke Depression among Stroke Survivors: An Appraisal of Psychiatry Needs and Care during Physiotherapy Rehabilitation. SCIENTIFICA 2016; 2016:5646052. [PMID: 27190683 PMCID: PMC4846762 DOI: 10.1155/2016/5646052] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/15/2016] [Accepted: 02/17/2016] [Indexed: 06/05/2023]
Abstract
Purpose. To identify stroke survivors with symptoms of poststroke depression and the extent of psychiatry needs and care they have received while on physiotherapy rehabilitation. Participants. Fifty stroke survivors (22 females and 28 males) at the outpatient unit of Physiotherapy Department, University of Nigeria Teaching Hospital, Enugu, who gave their informed consent, were randomly selected. Their age range and mean age were 26-66 years and 54.76 ± 8.79 years, respectively. Method. A multiple case study of 50 stroke survivors for symptoms of poststroke depression was done with Beck's Depression Inventory, mini mental status examination tool, and Modified Motor Assessment Scale. The tests were performed independently by the participants except otherwise stated and scored on a scale of 0-6. Data were analyzed using Z-test for proportional significance and chi-square test for determining relationship between variables, at p < 0.05. Results. Twenty-one (42.0%) stroke survivors had symptoms of PSD, which was significantly dependent on duration of stroke (χ (2) = 21.680, df = 6, and p = 0.001), yet none of the participants had a psychiatry review. Conclusions. Symptoms of PSD may be common in cold compared to new cases of stroke and may need psychiatry care while on physiotherapy rehabilitation.
Collapse
Affiliation(s)
- Sam Chidi Ibeneme
- Department of Medical Rehabilitation, Faculty of Health Sciences & Technology, College of Medicine, University of Nigeria, Enugu Campus, Enugu 40024, Nigeria
- German UNESCO Unit on Bioethics, Fakultat III-Medien, Information und Design, Hochschule Hannover-University of Applied Sciences and Arts, 30539 Hannover, Germany
| | - Canice Chukwudi Anyachukwu
- Department of Medical Rehabilitation, Faculty of Health Sciences & Technology, College of Medicine, University of Nigeria, Enugu Campus, Enugu 40024, Nigeria
| | - Akachukwu Nwosu
- National Institute for Sports, Surulere, Lagos 101283, Nigeria
| | - Georgian Chiaka Ibeneme
- Department of Nursing Sciences, Faculty of Health Sciences & Technology, College of Medicine, Ebonyi State University, Abakaliki 5480214, Nigeria
| | - Muideen Bakare
- Federal Neuro-Psychiatric Hospital, New Haven, Enugu 400221, Nigeria
| | - Gerhard Fortwengel
- German UNESCO Unit on Bioethics, Fakultat III-Medien, Information und Design, Hochschule Hannover-University of Applied Sciences and Arts, 30539 Hannover, Germany
| |
Collapse
|
28
|
Reducing central serotonin in adulthood promotes hippocampal neurogenesis. Sci Rep 2016; 6:20338. [PMID: 26839004 PMCID: PMC4738271 DOI: 10.1038/srep20338] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/30/2015] [Indexed: 12/20/2022] Open
Abstract
Chronic administration of selective serotonin reuptake inhibitors (SSRIs), which up-regulates central serotonin (5-HT) system function, enhances adult hippocampal neurogenesis. However, the relationship between central 5-HT system and adult neurogenesis has not fully been understood. Here, we report that lowering 5-HT level in adulthood is also able to enhance adult hippocampal neurogenesis. We used tamoxifen (TM)-induced Cre in Pet1-CreERT2 mice to either deplete central serotonergic (5-HTergic) neurons or inactivate 5-HT synthesis in adulthood and explore the role of central 5-HT in adult hippocampal neurogenesis. A dramatic increase in hippocampal neurogenesis is present in these two central 5-HT-deficient mice and it is largely prevented by administration of agonist for 5-HTR2c receptor. In addition, the survival of new-born neurons in the hippocampus is enhanced. Furthermore, the adult 5-HT-deficient mice showed reduced depression-like behaviors but enhanced contextual fear memory. These findings demonstrate that lowering central 5-HT function in adulthood can also enhance adult hippocampal neurogenesis, thus revealing a new aspect of central 5-HT in regulating adult neurogenesis.
Collapse
|
29
|
Sachs BD, Rodriguiz RM, Tran HL, Iyer A, Wetsel WC, Caron MG. Serotonin deficiency alters susceptibility to the long-term consequences of adverse early life experience. Psychoneuroendocrinology 2015; 53:69-81. [PMID: 25602134 PMCID: PMC4344834 DOI: 10.1016/j.psyneuen.2014.12.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 11/26/2014] [Accepted: 12/29/2014] [Indexed: 12/22/2022]
Abstract
Brain 5-HT deficiency has long been implicated in psychiatric disease, but the effects of 5-HT deficiency on stress susceptibility remain largely unknown. Early life stress (ELS) has been suggested to contribute to adult psychopathology, but efforts to study the long-term consequences of ELS have been limited by a lack of appropriate preclinical models. Here, we evaluated the effects of 5-HT deficiency on several long-term cellular, molecular, and behavioral responses of mice to a new model of ELS that combines early-life maternal separation (MS) of pups and postpartum learned helplessness (LH) training in dams. Our data demonstrate that this paradigm (LH/MS) induces depressive-like behavior and impairs pup retrieval in dams. In addition, we show that brain 5-HT deficiency exacerbates anxiety-like behavior induced by LH/MS and blunts the effects of LH/MS on acoustic startle responses in adult offspring. Although the mechanisms underlying these effects remain unclear, following LH/MS, 5-HT-deficient animals had significantly less mRNA expression of the mineralocorticoid receptor in the amygdala than wild-type animals. In addition, 5-HT-deficient mice exhibited reduced mRNA levels of the 5-HT2a receptor and p11 in the hippocampus regardless of stress. LH/MS decreased the number of doublecortin+ immature neurons in the hippocampus in both wild-type (WT) and 5-HT-deficient animals. Our data emphasize the importance of complex interactions between genetic factors and early life experience in mediating long-term changes in emotional behavior. These findings may have important implications for our understanding of the combinatorial roles of 5-HT deficiency, ELS, and postpartum depression in the development of neuropsychiatric disorders.
Collapse
MESH Headings
- Amygdala/metabolism
- Animals
- Animals, Newborn
- Behavior, Animal
- Depression, Postpartum/metabolism
- Depression, Postpartum/psychology
- Disease Models, Animal
- Disease Susceptibility
- Female
- Gene Knock-In Techniques
- Helplessness, Learned
- Hippocampus/metabolism
- Maternal Deprivation
- Mice
- Mutation
- Neurogenesis/genetics
- RNA, Messenger/metabolism
- Receptor, Serotonin, 5-HT2A/genetics
- Receptor, Serotonin, 5-HT2A/metabolism
- Receptors, Mineralocorticoid/genetics
- Receptors, Mineralocorticoid/metabolism
- Reflex, Abnormal
- Reflex, Startle
- Serotonin/deficiency
- Stress, Psychological/metabolism
- Stress, Psychological/psychology
- Tryptophan Hydroxylase/genetics
Collapse
Affiliation(s)
- Benjamin D Sachs
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, United States
| | - Ramona M Rodriguiz
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, United States; Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, NC 27710, United States
| | - Ha L Tran
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, United States
| | - Akshita Iyer
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, United States
| | - William C Wetsel
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, United States; Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, United States; Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, NC 27710, United States; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, United States
| | - Marc G Caron
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, United States; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, United States.
| |
Collapse
|
30
|
Brain 5-HT deficiency increases stress vulnerability and impairs antidepressant responses following psychosocial stress. Proc Natl Acad Sci U S A 2015; 112:2557-62. [PMID: 25675490 DOI: 10.1073/pnas.1416866112] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Brain serotonin (5-HT) deficiency and exposure to psychosocial stress have both been implicated in the etiology of depression and anxiety disorders, but whether 5-HT deficiency influences susceptibility to depression- and anxiety-like phenotypes induced by psychosocial stress has not been formally established. Most clinically effective antidepressants increase the extracellular levels of 5-HT, and thus it has been hypothesized that antidepressant responses result from the reversal of endogenous 5-HT deficiency, but this hypothesis remains highly controversial. Here we evaluated the impact of brain 5-HT deficiency on stress susceptibility and antidepressant-like responses using tryptophan hydroxylase 2 knockin (Tph2KI) mice, which display 60-80% reductions in brain 5-HT. Our results demonstrate that 5-HT deficiency leads to increased susceptibility to social defeat stress (SDS), a model of psychosocial stress, and prevents the fluoxetine (FLX)-induced reversal of SDS-induced social avoidance, suggesting that 5-HT deficiency may impair antidepressant responses. In light of recent clinical and preclinical studies highlighting the potential of inhibiting the lateral habenula (LHb) to achieve antidepressant and antidepressant-like responses, we also examined whether LHb inhibition could achieve antidepressant-like responses in FLX-insensitive Tph2KI mice subjected to SDS. Our data reveal that using designer receptors exclusively activated by designer drugs (DREADDs) to inhibit LHb activity leads to reduced SDS-induced social avoidance behavior in both WT and Tph2KI mice. This observation provides additional preclinical evidence that inhibiting the LHb might represent a promising alternative therapeutic approach under conditions in which selective 5-HT reuptake inhibitors are ineffective.
Collapse
|
31
|
Abstract
BACKGROUND Chronic treatment with antidepressants has been shown to enhance neurogenesis in the adult mammalian brain. Although this effect was initially reported to be restricted to the hippocampus, recent work has suggested that fluoxetine, a selective serotonin reuptake inhibitor, also promotes neurogenesis in the cortex. However, whether antidepressants target neural progenitor cells in other brain regions has not been examined. METHODS Here, we used BrdU labeling and immunohistochemistry with a transgenic mouse line in which nestin+ neural progenitor cells can be inducibly labeled with the fluorescent protein, Tomato, following tamoxifen administration. We investigated the effects of chronic fluoxetine on cell proliferation and nestin+ progenitor cells in periventricular areas in the medial hypothalamus and medial habenula, two brain areas involved in stress and anxiety responses. RESULTS Our data provide the first in vivo evidence that fluoxetine promotes cell proliferation and neurogenesis and increases the mRNA levels of BDNF in the hypothalamus and habenula. CONCLUSIONS By identifying novel cellular targets of fluoxetine, our results may provide new insight into the mechanisms underlying antidepressant responses.
Collapse
Affiliation(s)
- Benjamin D Sachs
- Department of Cell Biology, Duke University Medical Center, Durham, NC (Drs Sachs and Caron); Department of Neurobiology, Duke University Medical Center, Durham, NC (Dr Caron)
| | - Marc G Caron
- Department of Cell Biology, Duke University Medical Center, Durham, NC (Drs Sachs and Caron); Department of Neurobiology, Duke University Medical Center, Durham, NC (Dr Caron).
| |
Collapse
|
32
|
Angoa-Pérez M, Kane MJ, Briggs DI, Herrera-Mundo N, Sykes CE, Francescutti DM, Kuhn DM. Mice genetically depleted of brain serotonin do not display a depression-like behavioral phenotype. ACS Chem Neurosci 2014; 5:908-19. [PMID: 25089765 DOI: 10.1021/cn500096g] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Reductions in function within the serotonin (5HT) neuronal system have long been proposed as etiological factors in depression. Selective serotonin reuptake inhibitors (SSRIs) are the most common treatment for depression, and their therapeutic effect is generally attributed to their ability to increase the synaptic levels of 5HT. Tryptophan hydroxylase 2 (TPH2) is the initial and rate-limiting enzyme in the biosynthetic pathway of 5HT in the CNS, and losses in its catalytic activity lead to reductions in 5HT production and release. The time differential between the onset of 5HT reuptake inhibition by SSRIs (minutes) and onset of their antidepressant efficacy (weeks to months), when considered with their overall poor therapeutic effectiveness, has cast some doubt on the role of 5HT in depression. Mice lacking the gene for TPH2 are genetically depleted of brain 5HT and were tested for a depression-like behavioral phenotype using a battery of valid tests for affective-like disorders in animals. The behavior of TPH2(-/-) mice on the sucrose preference test, tail suspension test, and forced swim test and their responses in the unpredictable chronic mild stress and learned helplessness paradigms was the same as wild-type controls. While TPH2(-/-) mice as a group were not responsive to SSRIs, a subset responded to treatment with SSRIs in the same manner as wild-type controls with significant reductions in immobility time on the tail suspension test, indicative of antidepressant drug effects. The behavioral phenotype of the TPH2(-/-) mouse questions the role of 5HT in depression. Furthermore, the TPH2(-/-) mouse may serve as a useful model in the search for new medications that have therapeutic targets for depression that are outside of the 5HT neuronal system.
Collapse
Affiliation(s)
- Mariana Angoa-Pérez
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan 48201, United States
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Michael J. Kane
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan 48201, United States
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Denise I. Briggs
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan 48201, United States
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Nieves Herrera-Mundo
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan 48201, United States
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Catherine E. Sykes
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan 48201, United States
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Dina M. Francescutti
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan 48201, United States
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Donald M. Kuhn
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan 48201, United States
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| |
Collapse
|
33
|
The role of serotonin in adult hippocampal neurogenesis. Behav Brain Res 2014; 277:49-57. [PMID: 25125239 DOI: 10.1016/j.bbr.2014.07.038] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/22/2014] [Accepted: 07/23/2014] [Indexed: 12/17/2022]
Abstract
Serotonin is probably best known for its role in conveying a sense of contentedness and happiness. It is one of the most unique and pharmacologically complex monoamines in both the peripheral and central nervous system (CNS). Serotonin has become in focus of interest for the treatment of depression with multiple serotonin-mimetic and modulators of adult neurogenesis used clinically. Here we will take a broad view of serotonin from development to its physiological role as a neurotransmitter and its contribution to homeostasis of the adult rodent hippocampus. This chapter reflects the most significant findings on cellular and molecular mechanisms from neuroscientists in the field over the last two decades. We illustrate the action of serotonin by highlighting basic receptor targeting studies, and how receptors impact brain function. We give an overview of recent genetically modified mouse models that differ in serotonin availability and focus on the role of the monoamine in antidepressant response. We conclude with a synthesis of the most recent data surrounding the role of serotonin in activity and hippocampal neurogenesis. This synopsis sheds light on the mechanisms and potential therapeutic model by which serotonin plays a critical role in the maintenance of mood.
Collapse
|
34
|
Albert PR, Vahid-Ansari F, Luckhart C. Serotonin-prefrontal cortical circuitry in anxiety and depression phenotypes: pivotal role of pre- and post-synaptic 5-HT1A receptor expression. Front Behav Neurosci 2014; 8:199. [PMID: 24936175 PMCID: PMC4047678 DOI: 10.3389/fnbeh.2014.00199] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/16/2014] [Indexed: 01/03/2023] Open
Abstract
Decreased serotonergic activity has been implicated in anxiety and major depression, and antidepressants directly or indirectly increase the long-term activity of the serotonin system. A key component of serotonin circuitry is the 5-HT1A autoreceptor, which functions as the major somatodendritic autoreceptor to negatively regulate the "gain" of the serotonin system. In addition, 5-HT1A heteroreceptors are abundantly expressed post-synaptically in the prefrontal cortex (PFC), amygdala, and hippocampus to mediate serotonin actions on fear, anxiety, stress, and cognition. Importantly, in the PFC 5-HT1A heteroreceptors are expressed on at least two antagonist neuronal populations: excitatory pyramidal neurons and inhibitory interneurons. Rodent models implicate the 5-HT1A receptor in anxiety- and depression-like phenotypes with distinct roles for pre- and post-synaptic 5-HT1A receptors. In this review, we present a model of serotonin-PFC circuitry that integrates evidence from mouse genetic models of anxiety and depression involving knockout, suppression, over-expression, or mutation of genes of the serotonin system including 5-HT1A receptors. The model postulates that behavioral phenotype shifts as serotonin activity increases from none (depressed/aggressive not anxious) to low (anxious/depressed) to high (anxious, not depressed). We identify a set of conserved transcription factors including Deaf1, Freud-1/CC2D1A, Freud-2/CC2D1B and glucocorticoid receptors that may confer deleterious regional changes in 5-HT1A receptors in depression, and how future treatments could target these mechanisms. Further studies to specifically test the roles and regulation of pyramidal vs. interneuronal populations of 5-HT receptors are needed better understand the role of serotonin in anxiety and depression and to devise more effective targeted therapeutic approaches.
Collapse
Affiliation(s)
- Paul R Albert
- Neuroscience, Ottawa Hospital Research Institute, University of Ottawa Ottawa, ON, Canada
| | - Faranak Vahid-Ansari
- Neuroscience, Ottawa Hospital Research Institute, University of Ottawa Ottawa, ON, Canada ; Department of Cellular and Molecular Medicine, University of Ottawa Ottawa ON, Canada
| | - Christine Luckhart
- Neuroscience, Ottawa Hospital Research Institute, University of Ottawa Ottawa, ON, Canada ; Department of Cellular and Molecular Medicine, University of Ottawa Ottawa ON, Canada
| |
Collapse
|
35
|
Mosienko V, Matthes S, Hirth N, Beis D, Flinders M, Bader M, Hansson AC, Alenina N. Adaptive changes in serotonin metabolism preserve normal behavior in mice with reduced TPH2 activity. Neuropharmacology 2014; 85:73-80. [PMID: 24863038 DOI: 10.1016/j.neuropharm.2014.05.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 05/02/2014] [Accepted: 05/10/2014] [Indexed: 01/29/2023]
Abstract
Polymorphisms in the TPH2 gene coding for the serotonin synthesizing enzyme in the brain are considered as risk factors associated with depression and anxiety in humans. However, whether a certain variation in the TPH2 gene leads to decreased brain serotonin production and development of psychological abnormalities remains unresolved. We generated a new mouse model, carrying one Tph2-null allele and one Tph21473G-allele, coding for a hypoactive form of the enzyme. We tested these mice along with C57BL/6 mice (Tph2C/C), congenic C57BL/6 mice homozygous for the Tph21473G-allele (Tph2G/G), and heterozygous Tph2-deficient mice (Tph2C/-) for anxiety- and depression-like behavior, and evaluated brain serotonin metabolism and 5-HT1AR signaling by high-performance liquid chromatography and quantitative autoradiography, respectively. Progressive reduction in TPH2 activity had no effect on emotional behavior, and only slightly affected brain serotonin levels. However, serotonin degradation rate was drastically decreased in mice with reduced TPH2 activity, thereby compensating for the lowered rate of serotonin production in these mice. In addition, the hypothermic response to the 5-HT1AR agonist, 8-OH-DPAT, was attenuated in mice with reduced serotonin production. In contrast, 5-HT1A autoreceptor density and G-protein coupling were not changed in mice with gradual decrease in central serotonin. Taken together, these data suggest that in conditions of reduced serotonin production lowered serotonin degradation rate contributes to the maintenance of brain serotonin at levels sufficient for adequate behavior responses. These findings reveal that decreased TPH2 activity cannot be considered a reliable predisposition factor for impaired emotional behavior.
Collapse
Affiliation(s)
| | - Susann Matthes
- Max-Delbrueck-Center for Molecular Medicine, Berlin-Buch, Germany; Humboldt-Universität Berlin, Berlin, Germany
| | - Natalie Hirth
- Institute of Psychopharmacology at the Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Daniel Beis
- Max-Delbrueck-Center for Molecular Medicine, Berlin-Buch, Germany; Humboldt-Universität Berlin, Berlin, Germany
| | - Michael Flinders
- Max-Delbrueck-Center for Molecular Medicine, Berlin-Buch, Germany
| | - Michael Bader
- Max-Delbrueck-Center for Molecular Medicine, Berlin-Buch, Germany
| | - Anita C Hansson
- Institute of Psychopharmacology at the Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Natalia Alenina
- Max-Delbrueck-Center for Molecular Medicine, Berlin-Buch, Germany.
| |
Collapse
|
36
|
Sachs BD, Ni JR, Caron M. Sex differences in response to chronic mild stress and congenital serotonin deficiency. Psychoneuroendocrinology 2014; 40:123-9. [PMID: 24485484 PMCID: PMC3918518 DOI: 10.1016/j.psyneuen.2013.11.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 10/25/2013] [Accepted: 11/08/2013] [Indexed: 12/29/2022]
Abstract
Women exhibit a nearly twofold increased risk of developing depression and anxiety disorders when compared to men, a fact that has been hypothesized to result in part from increased stress susceptibility. Here, we used the tryptophan hydroxylase-2 R439H knock-in mouse (Tph2KI) and the chronic unpredictable mild stress (CMS) model to examine sex differences in response to congenital 5-HT deficiency and chronic stress. Our results demonstrate that female mice, but not 5-HT-deficient animals, exhibit significantly increased susceptibility to CMS-induced despair-like behavior in the forced swim test. In addition, female 5-HT-deficient mice exhibit anhedonia-like behavior in the sucrose preference test, whereas male 5-HT-deficient animals do not, suggesting that females exhibit increased sensitivity to at least some of the effects of congenital 5-HT deficiency. Although CMS did not reduce cell proliferation in the hippocampus, low levels of brain 5-HT were associated with increased hippocampal cell proliferation, an effect that was predominantly observed in females. Overall, these results highlight the importance of interactions between psychiatric disease risk factors such as sex, chronic stress and congenital 5-HT deficiency in the development of aberrant emotional behavior.
Collapse
Affiliation(s)
| | - Jason R. Ni
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Marc Caron
- Department of Cell Biology, Duke University, Durham, NC 27710, USA, Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
37
|
Role of microRNAs in stroke and poststroke depression. ScientificWorldJournal 2013; 2013:459692. [PMID: 24363618 PMCID: PMC3865697 DOI: 10.1155/2013/459692] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 11/07/2013] [Indexed: 12/22/2022] Open
Abstract
microRNAs (miRNA), a sort of noncoding RNAs widely distributed in eukaryotic cells, could regulate gene expression by inhibiting transcription or translation. They were involved in important physiological and pathological processes including growth, development, and occurrence and progression of diseases. miRNAs are crucial for the development of the nervous system. Recent studies have demonstrated that some miRNAs play important roles in the occurrence and development of ischemic cerebrovascular diseases such as stroke and were also involved in the occurrence and development of poststroke depression (PSD). Herein, studies on the role of miRNAs in the cerebral ischemia and PSD were reviewed, and results may be helpful for the diagnosis and prognosis of cerebral ischemia and PSD with miRNAs in clinical practice.
Collapse
|
38
|
Sachs BD, Salahi AA, Caron MG. Congenital brain serotonin deficiency leads to reduced ethanol sensitivity and increased ethanol consumption in mice. Neuropharmacology 2013; 77:177-84. [PMID: 24067926 DOI: 10.1016/j.neuropharm.2013.09.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 08/07/2013] [Accepted: 09/09/2013] [Indexed: 02/01/2023]
Abstract
Serotonergic dysfunction has been hypothesized to play an important role in the pathophysiology of alcoholism. However, whether congenital serotonin (5-HT) deficiency leads to increased alcohol consumption or affects ethanol-related behaviors has not been established. Here, we use a transgenic mouse line that expresses a hypofunctional variant of the 5-HT synthesis enzyme, tryptophan hydroxylase 2, to examine the impact of 5-HT deficiency on responses to alcohol. We demonstrate that these 5-HT-deficient transgenic animals (Tph2KI mice) recover their righting reflex more rapidly than wild-type controls following a high dose of ethanol and exhibit blunted locomotor retardation in response to repeated ethanol administration. In addition, compared to WT controls, 5-HT-deficient animals consume significantly more ethanol and exhibit increased preference for ethanol in two-bottle choice tests. Our data also suggest that 5-HT plays a critical role in mediating the effects of ethanol on Akt/GSK3β signaling in the nucleus accumbens. Overall, our results corroborate previous theories regarding the importance of brain 5-HT levels in mediating responsiveness to alcohol and demonstrate, for the first time, that congenital 5-HT deficiency leads to increased ethanol consumption and decreased sensitivity to the sedative-like effects of ethanol, perhaps in part through modulating Akt/GSK3β signaling.
Collapse
Affiliation(s)
- Benjamin D Sachs
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| | - A Ayten Salahi
- Department of Cell Biology, Duke University, Durham, NC 27710, USA; Research Scholars Program, Duke University, Durham, NC 27710, USA.
| | - Marc G Caron
- Department of Cell Biology, Duke University, Durham, NC 27710, USA; Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|