1
|
Basińska-Ziobroń A, Danek PJ, Daniel WA. The effect of prolonged treatment with antipsychotic drugs on cytochrome P450 - drug metabolizing enzymes. Mechanisms of action and significance for pharmacotherapy. Expert Opin Drug Metab Toxicol 2025. [PMID: 40491297 DOI: 10.1080/17425255.2025.2517731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/14/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025]
Abstract
INTRODUCTION The treatment of schizophrenia and other psychosis-related mental disorders requires long-term therapy with selected drugs possessing adequate pharmacological receptor spectra, relevant to the patient's clinical state. Antipsychotics can interact with cytochrome P450 (CYP) reciprocally affecting each other in different ways. The enzyme plays an important role in the metabolism of antipsychotics, whereas antipsychotics can affect CYP enzymes in the liver and brain. AREAS COVERED The effects of short and prolonged administration of antipsychotic drugs belonging to different groups (first-, second- and third-generation) on the expression and activity of CYP enzymes in the liver and brain are presented (based on PubMed 3 December 2024). Possible relations between pharmacological receptor spectra of antipsychotics and their influence on the regulation of cytochrome P450 in the liver and brain are considered. The results are discussed in the light of pharmacological and therapeutic significance. EXPERT OPINION During continuous treatment in vivo, the direct mechanisms (drug/metabolite binding to the CYP enzyme) overlap with the effect of antipsychotics on CYP regulation (enzyme induction or inhibition). Clinicians using the information on particular drug-CYP interaction in combination with pharmacogenetic data can make informed decisions about drug selection and dosage, ultimately advancing more effective and safer pharmacotherapy.
Collapse
Affiliation(s)
- Agnieszka Basińska-Ziobroń
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Poland
| | - Przemysław Jan Danek
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Poland
| | - Władysława Anna Daniel
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Poland
| |
Collapse
|
2
|
Robke R, Sansi F, Arbab T, Tunez A, Moore M, Bartsch D, Schönig K, Willuhn I. Optogenetic Stimulation of Novel Tph2-Cre Rats Advances Insight into Serotonin's Role in Locomotion, Reinforcement, and Compulsivity. J Neurosci 2025; 45:e1424242025. [PMID: 40204433 PMCID: PMC12096035 DOI: 10.1523/jneurosci.1424-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
Serotonin critically modulates the activity of many brain networks, including circuits that control motivation and responses to rewarding and aversive stimuli. Furthermore, the serotonin system is targeted by first-line pharmacological treatments for several psychiatric disorders, including obsessive-compulsive disorder. However, understanding the behavioral function of serotonin is hampered by methodological limitations: the (brainstem) location of serotonergic neuron cell-bodies is difficult to access, their innervation of the brain is diffuse, and they release serotonin in relatively low concentrations. Here, we advance this effort by developing novel Tph2-Cre rats, which we utilized to study serotonin in the context of motor, compulsive, and reinforced behaviors using optogenetics in both male and female rats. Specificity and sensitivity of Cre recombinase expression and Cre-dependent processes were validated immunohistochemically, and optogenetic induction of in vivo serotonin release was validated with fast-scan cyclic voltammetry. Optogenetic stimulation of serotonin neurons in the dorsal raphe nucleus did not initiate locomotion or alter aversion-induced locomotion, nor did it elicit (real-time) place preference, and it had no measurable effect on compulsive behavior in the schedule-induced polydipsia task. In contrast, this optogenetic stimulation moderately sustained ongoing spontaneous locomotion and robustly reinforced operant lever pressing for self-stimulation of serotonin neurons, which was exacerbated by food restriction. Together, this work both introduces a novel rat Cre line to study serotonin and advances our understanding of serotonin's behavioral functions. Complementing previous findings, we find that brainwide serotonin release has an overall relatively mild effect on behavior, which manifested only in the absence of natural reinforcers and was modulated by physiological state.
Collapse
Affiliation(s)
- Rhiannon Robke
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105BA, The Netherlands
- Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| | - Francesca Sansi
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105BA, The Netherlands
| | - Tara Arbab
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105BA, The Netherlands
- Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| | - Adria Tunez
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105BA, The Netherlands
- Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| | - Miranda Moore
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105BA, The Netherlands
| | - Dusan Bartsch
- Department of Molecular Biology, Central Institute of Mental Health, Mannheim 68159, Germany
| | - Kai Schönig
- Department of Molecular Biology, Central Institute of Mental Health, Mannheim 68159, Germany
| | - Ingo Willuhn
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105BA, The Netherlands
- Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| |
Collapse
|
3
|
Szebik H, Miskolczi C, Bruzsik B, Balla G, Szabó S, Biró L, Mikics É. Dynamic changes of serotonin transporter expression in the prefrontal cortex evoked by aggressive social interactions. Neurobiol Stress 2025; 36:100722. [PMID: 40230625 PMCID: PMC11994973 DOI: 10.1016/j.ynstr.2025.100722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
Aggression is a complex behavior influenced by developmental experiences, internal state, and social context, yet its neurobiological underpinnings remain insufficiently understood. The serotonergic system, particularly the serotonin transporter (SERT), plays a crucial role in aggression regulation. Here, we investigated region-specific, dynamic changes in SERT expression following aggressive interactions and in mice subjected to early-life social adversity. We found that aggressive encounters (resident-intruder test) triggered a significant, rapid increase in SERT immunoreactivity within 90 min, accompanied by neuronal activation in aggression-related brain regions, including the medial prefrontal cortex (mPFC), lateral septum (LS), medial amygdala (MeA), ventromedial hypothalamus (VMHvl), lateral habenula (LH), and dorsal raphe (DR), but not in the paraventricular thalamus (PVT). Notably, this SERT upregulation occurred across the aggression circuitry but was accompanied by a significant increase in 5-HT levels only in the mPFC, a key region in top-down regulation of social and aggressive behavior. This SERT upregulation was not observed following exposure to a non-social challenge, suggesting that it may be more specifically associated with social contexts. Using super-resolution microscopy, we identified an increased density of SERT localization points within serotonergic mPFC axons after an aggressive encounter. Social isolation during adolescence, a model of early social neglect, impaired this rapid SERT response, particularly in the ventral and medial orbitofrontal regions, and altered the relationship between SERT levels and aggression-related behaviors. These findings demonstrate that SERT expression undergoes rapid, experience-dependent plasticity in response to social aggression, and that early-life adversity disrupts this adaptive mechanism, providing new insights into the serotonergic regulation of aggression and its potential relevance for stress-related social dysfunctions.
Collapse
Affiliation(s)
- Huba Szebik
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
- Semmelweis University, Doctoral School, Budapest, Hungary
| | - Christina Miskolczi
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
- Semmelweis University, Doctoral School, Budapest, Hungary
| | - Bíborka Bruzsik
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Gyula Balla
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Soma Szabó
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - László Biró
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
- Thalamus Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Éva Mikics
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
4
|
Conde KM, Wong H, Fang S, Li Y, Yu M, Deng Y, Liu Q, Fang X, Wang M, Shi Y, Ginnard OZ, Yang Y, Tu L, Liu H, Liu H, Yin N, Bean JC, Han J, Burt ME, Jossy SV, Yang Y, Tong Q, Arenkiel BR, Wang C, He Y, Xu Y. Serotonin neurons integrate GABA and dopamine inputs to regulate meal initiation. Metabolism 2025; 163:156099. [PMID: 39667432 PMCID: PMC11924950 DOI: 10.1016/j.metabol.2024.156099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/04/2024] [Accepted: 12/07/2024] [Indexed: 12/14/2024]
Abstract
Obesity is a growing global health epidemic with limited orally administered therapeutics. Serotonin (5-HT) is one neurotransmitter which remains an excellent target for new weight-loss therapies, but a gap remains in understanding the mechanisms involved in 5-HT produced in the dorsal Raphe nucleus (DRN) and its involvement in meal initiation. Using an optogenetic feeding paradigm, we showed that the 5-HTDRN➔arcuate nucleus (ARH) circuit plays a role in meal initiation. Incorporating electrophysiology and ChannelRhodopsin-2-Assisted Circuit Mapping, we demonstrated that 5-HTDRN neurons receive inhibitory input partially from GABAergic neurons in the DRN, and the 5-HT response can be enhanced by hunger. Additionally, deletion of the GABAA receptor subunit in 5-HT neurons inhibits meal initiation with no effect on the satiation process. Finally, we identified the role of dopaminergic inputs via dopamine receptor D2 in enhancing the response to GABA-induced feeding. Thus, our results indicate that 5-HTDRN neurons are inhibited by synergistic inhibitory actions of GABA and dopamine, for the initiation of a meal.
Collapse
Affiliation(s)
- Kristine M Conde
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - HueyZhong Wong
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Shuzheng Fang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yongxiang Li
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Meng Yu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yue Deng
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Qingzhuo Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Xing Fang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Mengjie Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yuhan Shi
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Olivia Z Ginnard
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yuxue Yang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Longlong Tu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hesong Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hailan Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Na Yin
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jonathan C Bean
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Junying Han
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Megan E Burt
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Sanika V Jossy
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yongjie Yang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chunmei Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yang He
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
5
|
Gärde M, Matheson GJ, Varnäs K, Svenningsson P, Hedman-Lagerlöf E, Lundberg J, Farde L, Tiger M. Altered Serotonin 1B Receptor Binding After Escitalopram for Depression Is Correlated With Treatment Effect. Int J Neuropsychopharmacol 2024; 27:pyae021. [PMID: 38695786 PMCID: PMC11119883 DOI: 10.1093/ijnp/pyae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is commonly treated with selective serotonin reuptake inhibitors (SSRIs). SSRIs inhibit the serotonin transporter (5-HTT), but the downstream antidepressant mechanism of action of these drugs is poorly understood. The serotonin 1B (5-HT1B) receptor is functionally linked to 5-HTT and 5-HT1B receptor binding and 5-HT1B receptor mRNA is reduced in the raphe nuclei after SSRI administration in primates and rodents, respectively. The effect of SSRI treatment on 5-HT1B receptor binding in patients with MDD has not been examined previously. This positron emission tomography (PET) study aimed to quantify brain 5-HT1B receptor binding changes in vivo after SSRI treatment for MDD in relation to treatment effect. METHODS Eight unmedicated patients with moderate to severe MDD underwent PET with the 5-HT1B receptor radioligand [11C]AZ10419369 before and after 3 to 4 weeks of treatment with the SSRI escitalopram 10 mg daily. Depression severity was assessed at time of PET and after 6 to 7 weeks of treatment with the Montgomery-Åsberg Depression Rating Scale. RESULTS We observed a significant reduction in [11C]AZ10419369 binding in a dorsal brainstem (DBS) region containing the median and dorsal raphe nuclei after escitalopram treatment (P = .036). Change in DBS [11C]AZ10419369 binding correlated with Montgomery-Åsberg Depression Rating Scale reduction after 3-4 (r = 0.78, P = .021) and 6-7 (r = 0.94, P < .001) weeks' treatment. CONCLUSIONS Our findings align with the previously reported reduction of 5-HT1B receptor binding in the raphe nuclei after SSRI administration and support future studies testing change in DBS 5-HT1B receptor binding as an SSRI treatment response marker.
Collapse
Affiliation(s)
- M Gärde
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - G J Matheson
- Department of Psychiatry, Columbia University, New York, USA
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - K Varnäs
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - P Svenningsson
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - E Hedman-Lagerlöf
- Division of Psychology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - J Lundberg
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - L Farde
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - M Tiger
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| |
Collapse
|
6
|
Conde KM, Wong H, Fang S, Li Y, Yu M, Deng Y, Liu Q, Fang X, Wang M, Shi Y, Ginnard OZ, Yang Y, Tu L, Liu H, Liu H, Yin N, Bean JC, Han J, Burt ME, Jossy SV, Yang Y, Tong Q, Arenkiel BR, Wang C, He Y, Xu Y. 5-HT Neurons Integrate GABA and Dopamine Inputs to Regulate Meal Initiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.591360. [PMID: 38746314 PMCID: PMC11092489 DOI: 10.1101/2024.04.26.591360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Obesity is a growing global health epidemic with limited effective therapeutics. Serotonin (5-HT) is one major neurotransmitter which remains an excellent target for new weight-loss therapies, but there remains a gap in knowledge on the mechanisms involved in 5-HT produced in the dorsal Raphe nucleus (DRN) and its involvement in meal initiation. Using a closed-loop optogenetic feeding paradigm, we showed that the 5-HTDRN→arcuate nucleus (ARH) circuit plays an important role in regulating meal initiation. Incorporating electrophysiology and ChannelRhodopsin-2-Assisted Circuit Mapping, we demonstrated that 5-HTDRN neurons receive inhibitory input partially from GABAergic neurons in the DRN, and the 5-HT response to GABAergic inputs can be enhanced by hunger. Additionally, deletion of the GABAA receptor subunit in 5-HT neurons inhibits meal initiation with no effect on the satiation process. Finally, we identified the instrumental role of dopaminergic inputs via dopamine receptor D2 in 5-HTDRN neurons in enhancing the response to GABA-induced feeding. Thus, our results indicate that 5-HTDRN neurons are inhibited by synergistic inhibitory actions of GABA and dopamine, which allows for the initiation of a meal.
Collapse
Affiliation(s)
- Kristine M. Conde
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - HueyZhong Wong
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Shuzheng Fang
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yongxiang Li
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Meng Yu
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yue Deng
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Qingzhuo Liu
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Xing Fang
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Mengjie Wang
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yuhan Shi
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Olivia Z. Ginnard
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yuxue Yang
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Longlong Tu
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hesong Liu
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hailan Liu
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Na Yin
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jonathan C. Bean
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Junying Han
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Megan E. Burt
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Sanika V. Jossy
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yongjie Yang
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Benjamin R. Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chunmei Wang
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yang He
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yong Xu
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
7
|
Li X, Feng D, Ma S, Li M, Zhao S, Tang M. Ventral hippocampus is more sensitive to fluoxetine-induced changes in extracellular 5-HT concentration, membrane 5-HT transporter level and immobility times. Neuropharmacology 2024; 242:109766. [PMID: 37858884 DOI: 10.1016/j.neuropharm.2023.109766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/26/2023] [Accepted: 10/15/2023] [Indexed: 10/21/2023]
Abstract
Hippocampal responses to selective 5-HT reuptake inhibitor (SSRI) have long been studied. However, its sub-regional involvements in mediating SSRI's pharmacological effects have not been fully addressed. The current study sought to investigate neurochemical, neurobiological and neurobehavioral changes in response to direct fluoxetine perfusion into the ventral and dorsal sub-regions of the hippocampus in C57BL/6 mice. Following fluoxetine perfusion, time courses of dialysate 5-HT, 5-HT transporter (5-HTT) protein (total, membrane and cytoplasmic fractions), locomotion, and immobility times in the forced swim test (FST) and tail suspension test (TST) were determined. At baseline, 5-HT uptake efficiency assessed by the no-net-flux microdialysis, and 5-HTT protein were measured as well. Results show that fluoxetine dose-dependently increased dialysate 5-HT, lowered membrane 5-HTT protein and increased cytoplasmic fraction without changing the total level, decreased immobility times in both the FST and TST, with greater responses all detected in the ventral sub-region compared to the dorsal sub-region. Fluoxetine didn't affect locomotor activity, ruling out the possibility that fluoxetine's effects on immobility maybe due to alteration in locomotion. Besides, lower 5-HT uptake efficiency and lower membrane 5-HTT protein level were found in the ventral sub-region at baseline. Together, the sub-regional differences at baseline and in responses to fluoxetine added powerful evidence to support the existence of two distinct 5-HT sub-systems in the hippocampus, with greater changes to fluoxetine detected in the ventral sub-system.
Collapse
Affiliation(s)
- Xiang Li
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Dan Feng
- Department of Clinical Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Shenglu Ma
- Department of Clinical Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Mingxing Li
- Department of Clinical Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Shulei Zhao
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Man Tang
- Department of Clinical Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
8
|
Raphe serotonin projections dynamically regulate feeding behavior through targeting inhibitory circuits from rostral zona incerta to paraventricular thalamus. Mol Metab 2022; 66:101634. [PMID: 36351530 PMCID: PMC9672487 DOI: 10.1016/j.molmet.2022.101634] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVE Rostral zona incerta (ZIR) evokes feeding by sending GABA transmission to paraventricular thalamus (PVT). Although central serotonin (5-HT) signaling is known to play critical roles in the regulation of food intake and eating disorders, it remains unknown whether raphe 5-HT neurons functionally innervate ZIR-PVT neural pathway for feeding control. Here, we sought to reveal how raphe 5-HT signaling regulates both ZIR and PVT for feeding control. METHODS We used retrograde neural tracers to map 5-HT projections in Sert-Cre mice and slice electrophysiology to examine the mechanism by which 5-HT modulates ZIR GABA neurons. We also used optogenetics to test the effects of raphe-ZIR and raphe-PVT 5-HT projections on feeding motivation and food intake in mice regularly fed, 24 h fasted, and with intermittent high-fat high-sugar (HFHS) diet. In addition, we applied RNAscope in situ hybridization to identify 5-HT receptor subtype mRNA in ZIR. RESULTS We show raphe 5-HT neurons sent projections to both ZIR and PVT with partial collateral axons. Photostimulation of 5-HT projections inhibited ZIR but excited PVT neurons to decrease motivated food consumption. However, both acute food deprivation and intermittent HFHS diet downregulated 5-HT inhibition on ZIR GABA neurons, abolishing the inhibitory regulation of raphe-ZIR 5-HT projections on feeding motivation and food intake. Furthermore, we found high-level 5-HT1a and 5-HT2c as well as low-level 5-HT7 mRNA expression in ZIR. Intermittent HFHS diet increased 5-HT7 but not 5-HT1a or 5-HT2c mRNA levels in the ZIR. CONCLUSIONS Our results reveal that raphe-ZIR 5-HT projections dynamically regulate ZIR GABA neurons for feeding control, supporting that a dynamic fluctuation of ZIR 5-HT inhibition authorizes daily food intake but a sustained change of ZIR 5-HT signaling leads to overeating induced by HFHS diet.
Collapse
|
9
|
Müller CP. Serotonin and Consciousness-A Reappraisal. Behav Brain Res 2022; 432:113970. [PMID: 35716774 DOI: 10.1016/j.bbr.2022.113970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 11/02/2022]
Abstract
The serotonergic system of the brain is a major modulator of behaviour. Here we describe a re-appraisal of its function for consciousness based on anatomical, functional and pharmacological data. For a better understanding, the current model of consciousness is expanded. Two parallel streams of conscious flow are distinguished. A flow of conscious content and an affective consciousness flow. While conscious content flow has its functional equivalent in the activity of higher cortico-cortical and cortico-thalamic networks, affective conscious flow originates in segregated deeper brain structures for single emotions. It is hypothesized that single emotional networks converge on serotonergic and other modulatory transmitter neurons in the brainstem where a bound percept of an affective conscious flow is formed. This is then dispersed to cortical and thalamic networks, where it is time locked with conscious content flow at the level of these networks. Serotonin acts in concert with other modulatory systems of the brain stem with some possible specialization on single emotions. Together, these systems signal a bound percept of affective conscious flow. Dysfunctions in the serotonergic system may not only give rise to behavioural and somatic symptoms, but also essentially affect the coupling of conscious affective flow with conscious content flow, leading to the affect-stained subjective side of mental disorders like anxiety, depression, or schizophrenia. The present model is an attempt to integrate the growing insights into serotonergic system function. However, it is acknowledged, that several key claims are still at a heuristic level that need further empirical support.
Collapse
Affiliation(s)
- Christian P Müller
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany; Centre for Drug Research, Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia.
| |
Collapse
|
10
|
Cai X, Liu H, Feng B, Yu M, He Y, Liu H, Liang C, Yang Y, Tu L, Zhang N, Wang L, Yin N, Han J, Yan Z, Wang C, Xu P, Wu Q, Tong Q, He Y, Xu Y. A D2 to D1 shift in dopaminergic inputs to midbrain 5-HT neurons causes anorexia in mice. Nat Neurosci 2022; 25:646-658. [PMID: 35501380 PMCID: PMC9926508 DOI: 10.1038/s41593-022-01062-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/24/2022] [Indexed: 12/18/2022]
Abstract
Midbrain dopamine (DA) and serotonin (5-HT) neurons regulate motivated behaviors, including feeding, but less is known about how these circuits may interact. In this study, we found that DA neurons in the mouse ventral tegmental area bidirectionally regulate the activity of 5-HT neurons in the dorsal raphe nucleus (DRN), with weaker stimulation causing DRD2-dependent inhibition and overeating, while stronger stimulation causing DRD1-dependent activation and anorexia. Furthermore, in the activity-based anorexia (ABA) paradigm, which is a mouse model mimicking some clinical features of human anorexia nervosa (AN), we observed a DRD2 to DRD1 shift of DA neurotransmission on 5-HTDRN neurons, which causes constant activation of these neurons and contributes to AN-like behaviors. Finally, we found that systemic administration of a DRD1 antagonist can prevent anorexia and weight loss in ABA. Our results revealed regulation of feeding behavior by stimulation strength-dependent interactions between DA and 5-HT neurons, which may contribute to the pathophysiology of AN.
Collapse
Affiliation(s)
- Xing Cai
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Hailan Liu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Bing Feng
- Brain Glycemic and Metabolism Control Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Meng Yu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yang He
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Hesong Liu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Chen Liang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yongjie Yang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Longlong Tu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Nan Zhang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Lina Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Na Yin
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Junying Han
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Zili Yan
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Chunmei Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Pingwen Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Division of Endocrinology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Qi Wu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yanlin He
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Brain Glycemic and Metabolism Control Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA.
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
11
|
Study on the Antifatigue Effect of Compound Amino Acid Capsules. Cell Microbiol 2022. [DOI: 10.1155/2022/6593811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Supplementing amino acids was proven to relieve fatigue caused by exercise. This study explored the antifatigue effects of compound amino acid capsules (CAAC) on rats undergoing the forced swimming test (FST). CAAC augmented the endurance of FST in rats and alleviated the damage of skeletal muscle tissue and reduced the content of biochemical indicators in the serum. Furthermore, CAAC prevented skeletal muscle dysfunction in FST rats by modulating inflammation and oxidation reactions. After the treatment with CAAC, apoptosis and apoptosis-related protein and p-p65 were weakened, while the levels of SIRT1 and SIRT1/PGC-1α/Nrf2 pathway-related proteins were enhanced. The antifatigue properties of CAAC were associated with its antioxidant and anti-inflammatory capabilities, which were realized by activating the SIRT1/PGC-1α/Nrf2 pathway.
Collapse
|
12
|
He Y, Cai X, Liu H, Conde KM, Xu P, Li Y, Wang C, Yu M, He Y, Liu H, Liang C, Yang T, Yang Y, Yu K, Wang J, Zheng R, Liu F, Sun Z, Heisler L, Wu Q, Tong Q, Zhu C, Shu G, Xu Y. 5-HT recruits distinct neurocircuits to inhibit hunger-driven and non-hunger-driven feeding. Mol Psychiatry 2021; 26:7211-7224. [PMID: 34290371 PMCID: PMC8776930 DOI: 10.1038/s41380-021-01220-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 06/14/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023]
Abstract
Obesity is primarily a consequence of consuming calories beyond energetic requirements, but underpinning drivers have not been fully defined. 5-Hydroxytryptamine (5-HT) neurons in the dorsal Raphe nucleus (5-HTDRN) regulate different types of feeding behavior, such as eating to cope with hunger or for pleasure. Here, we observed that activation of 5-HTDRN to hypothalamic arcuate nucleus (5-HTDRN → ARH) projections inhibits food intake driven by hunger via actions at ARH 5-HT2C and 5-HT1B receptors, whereas activation of 5-HTDRN to ventral tegmental area (5-HTDRN → VTA) projections inhibits non-hunger-driven feeding via actions at 5-HT2C receptors. Further, hunger-driven feeding gradually activates ARH-projecting 5-HTDRN neurons via inhibiting their responsiveness to inhibitory GABAergic inputs; non-hunger-driven feeding activates VTA-projecting 5-HTDRN neurons through reducing a potassium outward current. Thus, our results support a model whereby parallel circuits modulate feeding behavior either in response to hunger or to hunger-independent cues.
Collapse
Affiliation(s)
- Yanlin He
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Pennington Biomedical Research Center, Brain Glycemic and Metabolism Control Department, Louisiana State University, Baton Rouge, LA, USA
| | - Xing Cai
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Hailan Liu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Krisitine M Conde
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Pingwen Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, USA
| | - Yongxiang Li
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, College of Animal Science, South China Agricultural University, Guangdong, China
| | - Chunmei Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Meng Yu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yang He
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Hesong Liu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Chen Liang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Tingting Yang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yongjie Yang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Kaifan Yu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Julia Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Rong Zheng
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Feng Liu
- Departments of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Zheng Sun
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Lora Heisler
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, UK
| | - Qi Wu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Canjun Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, College of Animal Science, South China Agricultural University, Guangdong, China
| | - Gang Shu
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, College of Animal Science, South China Agricultural University, Guangdong, China.
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
13
|
Orexin receptors 1 and 2 in serotonergic neurons differentially regulate peripheral glucose metabolism in obesity. Nat Commun 2021; 12:5249. [PMID: 34475397 PMCID: PMC8413382 DOI: 10.1038/s41467-021-25380-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 08/05/2021] [Indexed: 12/02/2022] Open
Abstract
The wake-active orexin system plays a central role in the dynamic regulation of glucose homeostasis. Here we show orexin receptor type 1 and 2 are predominantly expressed in dorsal raphe nucleus-dorsal and -ventral, respectively. Serotonergic neurons in ventral median raphe nucleus and raphe pallidus selectively express orexin receptor type 1. Inactivation of orexin receptor type 1 in serotonin transporter-expressing cells of mice reduced insulin sensitivity in diet-induced obesity, mainly by decreasing glucose utilization in brown adipose tissue and skeletal muscle. Selective inactivation of orexin receptor type 2 improved glucose tolerance and insulin sensitivity in obese mice, mainly through a decrease in hepatic gluconeogenesis. Optogenetic activation of orexin neurons in lateral hypothalamus or orexinergic fibers innervating raphe pallidus impaired or improved glucose tolerance, respectively. Collectively, the present study assigns orexin signaling in serotonergic neurons critical, yet differential orexin receptor type 1- and 2-dependent functions in the regulation of systemic glucose homeostasis. The wake-active orexin system plays a central role in the dynamic regulation of glucose homeostasis. Here the authors report that inactivation of the orexin receptor type 1 or 2 in serotonergic neurons differentially regulate systemic glucose homeostasis in the context of diet induced obesity.
Collapse
|
14
|
Nasrollahi S, Karimi S, Hamidi G, Naderitehrani M, Abed A. Blockade of the orexin 1 receptors in the nucleus accumbens' shell reversed the reduction effect of olanzapine on motivation for positive reinforcers. Neurosci Lett 2021; 762:136137. [PMID: 34311049 DOI: 10.1016/j.neulet.2021.136137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
Abstract
Effort-based choice of high reward requires one to decide how much effort to expend for a certain amount of reward. Orexin is a crucial neuropeptide in the physiological aspect especially a variety of affective and cognitive processes. The nucleus accumbens (NAc) is a region of the neural system that serves effort-related high reward choices andthe Orexin 1 receptor (OX1R) is distributed extensively throughout the nucleus accumbens shell (AcbS). Olanzapine (OLZ), a typical antipsychotic drug, has a high affinity to D2 as an antagonist, and also partial agonistic-like action at D2 receptors has been reported. We examined the interaction of OLZ with the orexinergic receptor 1 in AcbS on effort- related high reward choice when two goal arms were different in the amount of accessible reward. The animals had to pass the barrier for receiving a high reward in one arm (HRA) or obtain a low reward in the other arm without any cost. Before surgery, all animals were selecting the HRA on almost every trial.During test days, the rats received local injections of either DMSO 20% /0.5 µl, as vehicle or SB334867 (30, 100, 300 nM/0.5 µl), as selective OX1R antagonist, within the AcbS. Other group received OLZ (32 µM/0.5 µl DMSO20%) / vehicle alone or 5 min after administration of SB334867 (300 nM/0.5 µl). The results showed that administration of OLZ in the AcbS alters rat's preference for high reward. On the other hand, blocked of the OX1R (300 nM/0.5 µl) in this region could reverse the effect of OLZ, however, administration of the OX1R antagonists alone in the AcbS led to decreasing rat's preference for high reward. This result indicates that the orexin-1 antagonist might affect some effects of antipsychotic drugs.
Collapse
Affiliation(s)
- Saeedeh Nasrollahi
- Institute for Basic Sciences, Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran; Department of Physiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Sara Karimi
- Institute for Basic Sciences, Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Gholamali Hamidi
- Institute for Basic Sciences, Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran; Department of Physiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| | - Monireh Naderitehrani
- Institute for Basic Sciences, Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran; Department of Physiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Abed
- Institute for Basic Sciences, Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
15
|
Bromek E, Daniel WA. The regulation of liver cytochrome P450 expression and activity by the brain serotonergic system in different experimental models. Expert Opin Drug Metab Toxicol 2021; 17:413-424. [PMID: 33400885 DOI: 10.1080/17425255.2021.1872543] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Introduction: Cytochrome P450 (CYP) metabolizes vital endogenous (steroids, vitamins) and exogenous (drugs, toxins) substrates. Studies of the last decade have revealed that the brain dopaminergic and noradrenergic systems are involved in the regulation of CYP. Recent research indicates that the brain serotonergic system is also engaged in its regulation.Areas covered: This review focuses on the role of the brain serotonergic system in the regulation of liver CYP expression. It shows the effect of lesion and activation of the serotonergic system after peripheral or intracerebral injections of neurotoxins, serotonin precursor, or serotonin (5-HT) receptor agonists. An opposite role of the hypothalamic paraventricular and arcuate nuclei and 5-HT receptors present therein in the regulation of CYP is described. The engagement of those nuclei in the neuroendocrine regulation of CYP by hypothalamic releasing or inhibiting hormones, pituitary hormones, and peripheral gland hormones are shown.Expert opinion: In general, the brain serotonergic system negatively regulates liver cytochrome P450. However, the effects of serotonergic agents on the enzyme expression depend on their mechanism of action, the route of administration (intracerebral/peripheral), as well as on local intracerebral site of injection and 5-HT receptor-subtypes present therein.
Collapse
Affiliation(s)
- Ewa Bromek
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Władysława Anna Daniel
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
16
|
Boukersi H, Lebaili N, Nosjean A, Samson N, Faure A, Granon S. Effects of water restriction on social behavior and 5-HT neurons density in the dorsal and median raphe nuclei in mice. Behav Brain Res 2020; 399:113022. [PMID: 33232678 DOI: 10.1016/j.bbr.2020.113022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 10/22/2022]
Abstract
We explored here the hypothesis that temporary chronic water restriction in mice affects social behavior, via its action on the density of 5-HT neurons in dorsal and median raphe nuclei (DRN and MRN). For that, we submitted adult C57BL/6 J mice to mild and controlled temporary dehydration, i.e., 6 h of water access every 48 h for 15 days. We investigated their social behavior in a social interaction task known to allow free and reciprocal social contact. Results showed that temporary dehydration increases significantly time spent in social contact and social dominance. It also expands 5-HT neuron density within both DRN and MRN and the behavioral and neuronal plasticity were positively correlated. Our findings suggest that disturbance in 5-HT neurotransmission caused by temporary dehydration stress unbalances choice processes of animals in social context.
Collapse
Affiliation(s)
- Houari Boukersi
- Department of Biology, Faculty of Natural Science and Life, Hassiba Benbouali University, Chlef, Algeria; Animal Ecophysiology Laboratory, Higher Normal School Elbachir El-Ibrahimi, Kouba, Algers, Algeria; Paris-Saclay Institute of Neuroscience (NeuroPSI), Paris-Saclay University, CNRS 9197, Orsay, France.
| | - Nemcha Lebaili
- Animal Ecophysiology Laboratory, Higher Normal School Elbachir El-Ibrahimi, Kouba, Algers, Algeria
| | - Anne Nosjean
- Paris-Saclay Institute of Neuroscience (NeuroPSI), Paris-Saclay University, CNRS 9197, Orsay, France
| | - Nathalie Samson
- Paris-Saclay Institute of Neuroscience (NeuroPSI), Paris-Saclay University, CNRS 9197, Orsay, France
| | - Alexis Faure
- Paris-Saclay Institute of Neuroscience (NeuroPSI), Paris-Saclay University, CNRS 9197, Orsay, France
| | - Sylvie Granon
- Paris-Saclay Institute of Neuroscience (NeuroPSI), Paris-Saclay University, CNRS 9197, Orsay, France
| |
Collapse
|
17
|
Li X, Sun X, Sun J, Zu Y, Zhao S, Sun X, Li L, Zhang X, Wang W, Liang Y, Wang W, Liang X, Sun C, Guan X, Tang M. Depressive-like state sensitizes 5-HT 1A and 5-HT 1B auto-receptors in the dorsal raphe nucleus sub-system. Behav Brain Res 2020; 389:112618. [PMID: 32360167 DOI: 10.1016/j.bbr.2020.112618] [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] [Received: 01/20/2020] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 12/14/2022]
Abstract
Dorsal raphe (DR) and median raphe (MR) 5-HT neurons are two distinct sub-systems known to be regulated by 5-HT1A and 5-HT1B auto-receptors. Whether the auto-receptors in each sub-system are functionally altered in depressive-like state remains unknown. The present study is aimed to study a specific circuit (DR-ventral hippocampus and MR-dorsal hippocampus) within each sub-system to investigate changes in receptor sensitivity in the pathogenesis of depression. A mouse model of depression was developed through the social defeat paradigm, and was then treated with fluoxetine (FLX). 5-HT1A auto-receptor in the neuronal cell body (DR or MR) and 5-HT1B auto-receptor in the axonal terminal (ventral or dorsal hippocampus) were directly targeted by local perfusion of antagonists (5-HT1A: WAY100635; 5-HT1B: GR127935) through reverse microdialysis. Time courses of dialysate 5-HT measured at the axonal terminal were subsequently determined for each circuit. At baseline, 5-HT1A and 5-HT1B antagonists dose-dependently increased dialysate 5-HT, with sub-circuit specificity. In the depressive-like state, greater increases in dialysate 5-HT were observed only in the DR-ventral hippocampus circuit following local delivery of both antagonists, which were then fully restored following the FLX treatment. In contrast, no changes were observed in the MR-dorsal hippocampus circuit. Our results demonstrate differential changes in sensitivities of 5-HT1A and 5-HT1B auto-receptors in the DR-ventral hippocampus and MR-dorsal hippocampus circuits. 5-HT1A and 5-HT1B auto-receptors in the DR-ventral hippocampus circuit are sensitized in the depressive-like state. Taken together, these results suggest that the DR sub-system maybe the neural substrate mediating depressive phenotypes.
Collapse
Affiliation(s)
- Xiang Li
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Xianan Sun
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Jing Sun
- Department of Outpatient, Rocket Force University of Engineering Clinic Affiliated to 986 Hospital of Air Force, Xi'an, 710043, China
| | - Yi Zu
- Department of Academic Quality Assurance, China Medical University, Shenyang, 110122, China
| | - Shulei Zhao
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Xiao Sun
- Department of Internal Medicine, Shenyang Women's and Children's Hospital, Shenyang, 110011, China
| | - Lu Li
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Xinjing Zhang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Wei Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Yuezhu Liang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Wenyao Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Xuankai Liang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Chi Sun
- Department of Academic Quality Assurance, China Medical University, Shenyang, 110122, China
| | - Xue Guan
- Department of Academic Quality Assurance, China Medical University, Shenyang, 110122, China
| | - Man Tang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
18
|
Maia GH, Soares JI, Almeida SG, Leite JM, Baptista HX, Lukoyanova AN, Brazete CS, Lukoyanov NV. Altered serotonin innervation in the rat epileptic brain. Brain Res Bull 2019; 152:95-106. [DOI: 10.1016/j.brainresbull.2019.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 06/28/2019] [Accepted: 07/08/2019] [Indexed: 01/19/2023]
|
19
|
Bueno D, Lima LB, Souza R, Gonçalves L, Leite F, Souza S, Furigo IC, Donato J, Metzger M. Connections of the laterodorsal tegmental nucleus with the habenular‐interpeduncular‐raphe system. J Comp Neurol 2019; 527:3046-3072. [DOI: 10.1002/cne.24729] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Debora Bueno
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo São Paulo Brazil
| | - Leandro B. Lima
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo São Paulo Brazil
| | - Rudieri Souza
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo São Paulo Brazil
| | - Luciano Gonçalves
- Department of Human AnatomyFederal University of the Triângulo Mineiro Uberaba Brazil
| | - Fernanda Leite
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo São Paulo Brazil
| | - Stefani Souza
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo São Paulo Brazil
| | - Isadora C. Furigo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo São Paulo Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo São Paulo Brazil
| | - Martin Metzger
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo São Paulo Brazil
| |
Collapse
|
20
|
Moriya S, Yamashita A, Nishi R, Ikoma Y, Yamanaka A, Kuwaki T. Acute nociceptive stimuli rapidly induce the activity of serotonin and noradrenalin neurons in the brain stem of awake mice. IBRO Rep 2019; 7:1-9. [PMID: 31194165 PMCID: PMC6554543 DOI: 10.1016/j.ibror.2019.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 05/25/2019] [Indexed: 12/12/2022] Open
Abstract
Nociception is an important type of perception that has major influence on daily human life. There are some descending pathways related to pain management and modulation, which are collectively known as the descending antinociceptive system (DAS). Noradrenalin (NA) in the locus coeruleus (LC) and serotonin (5-HT) in the rostral ventromedial medulla (RVM) are components of the DAS. Most 5-HT neurons in the dorsal raphe (DR) have ascending projections rather than descending projections, and they project to the thalamus that modulates nociception. Both the DAS and the DR are believed to be involved in pain-emotion symptoms. In this study, we utilized a fiber photometry system to specifically examine the activity of LC NA neurons and RVM/DR 5-HT neurons using mice carrying tetracycline-controlled transactivator transgene (tTA) under the control of either a dopamine β-hydroxylase promoter or a tryptophan hydroxylase-2 promoter and site-specific infection of an adeno-associated virus carrying a TetO G-CaMP6 gene. After confirmation of specific expression of G-CaMP6 in the target populations, changes in green fluorescent signal intensity were recorded in awake mice upon exposure to acute nociceptive stimulation consisting of a pinch and application of heat (55 °C) to the tail. Both stimuli resulted in rapid and transient (<15 s) increases in the activity of LC NA neurons and RVM/DR 5-HT neurons while the control stimuli did not induce any changes. The present results clearly indicate that acute nociceptive stimuli increase the activity of LC NA neurons and RVM/DR 5 H T neurons and suggest a possible therapeutic target for pain treatment.
Collapse
Key Words
- 5-HT, serotonin
- AAV, adeno associated virus
- CaM, calmodulin
- DAS, descending antinociceptive system
- DBH, Dopamine beta hydroxylase
- DR, dorsal raphe
- Dorsal raphe (DR)
- Fiber photometry
- G-CaMP6
- LC, locus coeruleus
- Locus coeruleus (LC)
- NA, noradrenalin
- PAG, periaqueductal gray
- PBS, phosphate-buffered saline
- PFA, paraformaldehyde
- PMT, photomultiplier tube
- RVM, rostral ventromedial medulla
- Rostral ventromedial medulla (RVM)
- SEM, standard error of the mean
- SNRI, serotonin noradrenalin reuptake inhibitor
- TPH, tryptophan hydroxylase
Collapse
Affiliation(s)
- Shunpei Moriya
- Department of Physiology, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, 890-8544, Japan
| | - Akira Yamashita
- Department of Physiology, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, 890-8544, Japan
| | - Ryusei Nishi
- Department of Physiology, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, 890-8544, Japan
| | - Yoko Ikoma
- Department of Physiology, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, 890-8544, Japan
| | - Akihiro Yamanaka
- Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| | - Tomoyuki Kuwaki
- Department of Physiology, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, 890-8544, Japan
| |
Collapse
|
21
|
Wang LS, Zhang MD, Tao X, Zhou YF, Liu XM, Pan RL, Liao YH, Chang Q. LC-MS/MS-based quantification of tryptophan metabolites and neurotransmitters in the serum and brain of mice. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1112:24-32. [PMID: 30836315 DOI: 10.1016/j.jchromb.2019.02.021] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/31/2019] [Accepted: 02/21/2019] [Indexed: 10/27/2022]
Abstract
l-Tryptophan (Trp) metabolites and related neurotransmitters play crucial roles in physiological functions, and their imbalances are implicated in the pathology of depression, Alzheimer's disease and other diseases. Measurement of Trp metabolites and related neurotransmitters possesses a great potential to elucidate the disease mechanisms and evaluate the outcomes of therapeutic interventions. A simple, rapid, sensitive and specific liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed for simultaneous determination of Trp, l-kynurenine (Kyn), kynurenic acid (Kyna), 3-hydroxykynurenine (3-HK), 5-hydroxytryptamine (5-HT), 5-hydroxyindoleacetic acid (5-HIAA), norepinephrine (NE), l-glutamic acid (Glu), γ-aminobutyric acid (GABA) and acetylcholine (ACh) in mice serum and the brain tissues in a single chromatographic run. Samples were spiked with the internal standard, mixed with trifluoroacetic acid to precipitate protein and analyzed by LC-MS/MS. Chromatographic separation was achieved using a Restek Ultra Aqueous C18 column in combination with a gradient elution within 8 min. Mass spectrometric detection was performed using multiple reaction monitoring with electrospray ionization source in positive mode. The method exhibited good selectivity and correlation coefficient values for the calibration curves of each analyte were >0.99. The limit of detection and quantification ranged from 0.96 to 24.48 nmol/L and 3.42 to 244.82 nmol/L, respectively. The intra- and inter-day precision were ≤13.92%. All analytes were stable in prepared samples at room temperature in the autosampler for 24 h. This method was successfully applied to the analysis of biological samples from control and chronic mild stress (CMS) induced depression mice. It was found that Kyn and 3-HK pathways were enhanced by CMS, while the levels of Trp, Kyna, 5-HIAA, Glu, GABA and ACh were significantly reduced. The changes in 5-HT and NE levels were not uniform in the periphery and the brain. This method can therefore be applied to analyze Trp metabolites and related neurotransmitters levels to monitor disease states, study the mechanisms and outcomes of therapeutic interventions.
Collapse
Affiliation(s)
- Li-Sha Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Meng-Di Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Xue Tao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Yun-Feng Zhou
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Xin-Min Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Rui-Le Pan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Yong-Hong Liao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Qi Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| |
Collapse
|
22
|
Wen S, Wang C, Gong M, Zhou L. An overview of energy and metabolic regulation. SCIENCE CHINA-LIFE SCIENCES 2018; 62:771-790. [PMID: 30367342 DOI: 10.1007/s11427-018-9371-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/23/2018] [Indexed: 12/21/2022]
Abstract
The physiology and behaviors related to energy balance are monitored by the nervous and humoral systems. Because of the difficulty in treating diabetes and obesity, elucidating the energy balance mechanism and identifying critical targets for treatment are important research goals. Therefore, the purpose of this article is to describe energy regulation by the central nervous system (CNS) and peripheral humoral pathway. Homeostasis and rewarding are the basis of CNS regulation. Anorexigenic or orexigenic effects reflect the activities of the POMC/CART or NPY/AgRP neurons within the hypothalamus. Neurotransmitters have roles in food intake, and responsive brain nuclei have different functions related to food intake, glucose monitoring, reward processing. Peripheral gut- or adipose-derived hormones are the major source of peripheral humoral regulation systems. Nutrients or metabolites and gut microbiota affect metabolism via a discrete pathway. We also review the role of peripheral organs, the liver, adipose tissue, and skeletal muscle in peripheral regulation. We discuss these topics and how the body regulates metabolism.
Collapse
Affiliation(s)
- Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Chaoxun Wang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Min Gong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China.
| |
Collapse
|
23
|
Bambico FR, Comai S, Diwan M, Hasan SN, Conway JD, Darvish-Ghane S, Hamani C, Gobbi G, Nobrega JN. High frequency stimulation of the anterior vermis modulates behavioural response to chronic stress: involvement of the prefrontal cortex and dorsal raphe? Neurobiol Dis 2018; 116:166-178. [DOI: 10.1016/j.nbd.2018.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 02/17/2018] [Accepted: 03/24/2018] [Indexed: 12/25/2022] Open
|
24
|
Shishkina GT, Bulygina VV, Agarina NP, Dygalo NN. The Expression of Brain-Derived Neurotrophic Factor and Tryptophan Hydroxylase in the Dorsal Raphe Nucleus during Repeated Stress. NEUROCHEM J+ 2018. [DOI: 10.1134/s1819712418020101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
25
|
Nesfatin-1 in the dorsal raphe nucleus influences visceral sensitivity via 5-HT neurons in male maternally separated rats. Sci Rep 2018; 8:9334. [PMID: 29921870 PMCID: PMC6008476 DOI: 10.1038/s41598-018-27592-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 06/04/2018] [Indexed: 12/19/2022] Open
Abstract
Nesfatin-1, a satiety molecule processed from nucleobindin2 (NUCB2), is implicated in visceral hypersensitivity in rats and colocalized with 5-hydroxytryptamine (5-HT) in the dorsal raphe nucleus (DRN). Maternal separation (MS) in rats contributes to visceral hypersensitivity via elevated expression of 5-HT in the DRN. Intracerebroventricular injection of nesfatin-1 activates DRN 5-HT neurons. In this study, A model of visceral hypersensitivity was developed by subjecting rats to MS. Colorectal distension was used to detect visceral sensitivity, which was evaluated by abdominal withdrawal reflex (AWR) scores and electromyogram (EMG) magnitude. MS rats exhibited higher AWR scores and EMG magnitude compared with controls. The numbers of nesfatin-1- and tryptophan hydroxylase (TPH, the rate-limiting enzyme for 5-HT synthesis)-positive cells in the DRN were significantly elevated accordingly. Visceral hypersensitivity was significantly alleviated in MS rats treated with intra-DRN administration of anti-nesfatin-1/NUCB2, accompanied by decreased expression of 5-HT and TPH in the DRN, compared with the vehicle-treated group. In contrast, intra-DRN administration of nesfatin-1 into normal adult rats induced visceral hypersensitivity, which correlated with elevated expression of 5-HT and TPH in the DRN. In conclusion, Nesfatin-1 has critical effects on visceral hypersensitivity; the underlying mechanisms might be related to the activation of DRN 5-HT neurons.
Collapse
|
26
|
Verheij MMM, Contet C, Karel P, Latour J, van der Doelen RHA, Geenen B, van Hulten JA, Meyer F, Kozicz T, George O, Koob GF, Homberg JR. Median and Dorsal Raphe Serotonergic Neurons Control Moderate Versus Compulsive Cocaine Intake. Biol Psychiatry 2018; 83:1024-1035. [PMID: 29357981 PMCID: PMC5960600 DOI: 10.1016/j.biopsych.2017.10.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 10/12/2017] [Accepted: 10/18/2017] [Indexed: 01/30/2023]
Abstract
BACKGROUND Reduced expression of the serotonin transporter (SERT) promotes anxiety and cocaine intake in both humans and rats. We tested the hypothesis that median raphe nucleus (MRN) and dorsal raphe nucleus (DRN) serotonergic projections differentially mediate these phenotypes. METHODS We used virally mediated RNA interference to locally downregulate SERT expression and compared the results with those of constitutive SERT knockout. Rats were allowed either short access (ShA) (1 hour) or long access (LgA) (6 hours) to cocaine self-administration to model moderate versus compulsive-like cocaine taking. RESULTS SERT knockdown in the MRN increased cocaine intake selectively under ShA conditions and, like ShA cocaine self-administration, reduced corticotropin-releasing factor (CRF) immunodensity in the paraventricular nucleus of the hypothalamus. In contrast, SERT knockdown in the DRN increased cocaine intake selectively under LgA conditions and, like LgA cocaine self-administration, reduced CRF immunodensity in the central nucleus of the amygdala. SERT knockdown in the MRN or DRN produced anxiety-like behavior, as did withdrawal from ShA or LgA cocaine self-administration. The phenotype of SERT knockout rats was a summation of the phenotypes generated by MRN- and DRN-specific SERT knockdown. CONCLUSIONS Our results highlight a differential role of serotonergic projections arising from the MRN and DRN in the regulation of cocaine intake. We propose that a cocaine-induced shift from MRN-driven serotonergic control of CRF levels in the hypothalamus to DRN-driven serotonergic control of CRF levels in the amygdala may contribute to the transition from moderate to compulsive intake of cocaine.
Collapse
Affiliation(s)
- Michel M M Verheij
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands; Department of Molecular and Animal Physiology, Nijmegen Center for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands.
| | - Candice Contet
- Department of Molecular and Animal Physiology, Nijmegen Center for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Peter Karel
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands
| | - Judith Latour
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands
| | - Rick H A van der Doelen
- Department of Anatomy, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands
| | - Bram Geenen
- Department of Anatomy, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands
| | | | - Francisca Meyer
- Department of Neuroscience, Scripps Research Institute, La Jolla, California
| | - Tamas Kozicz
- Department of Anatomy, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands
| | - Olivier George
- Department of Molecular and Animal Physiology, Nijmegen Center for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - George F Koob
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands
| |
Collapse
|
27
|
Xu Y, López M. Central regulation of energy metabolism by estrogens. Mol Metab 2018; 15:104-115. [PMID: 29886181 PMCID: PMC6066788 DOI: 10.1016/j.molmet.2018.05.012] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 05/09/2018] [Accepted: 05/15/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Estrogenic actions in the brain prevent obesity. Better understanding of the underlying mechanisms may facilitate development of new obesity therapies. SCOPE OF REVIEW This review focuses on the critical brain regions that mediate effects of estrogens on food intake and/or energy expenditure, the molecular signals that are involved, and the functional interactions between brain estrogens and other signals modulating metabolism. Body weight regulation by estrogens in male brains will also be discussed. MAJOR CONCLUSIONS 17β-estradiol acts in the brain to regulate energy homeostasis in both sexes. It can inhibit feeding and stimulate brown adipose tissue thermogenesis. A better understanding of the central actions of 17β-estradiol on energy balance would provide new insight for the development of therapies against obesity in both sexes.
Collapse
Affiliation(s)
- Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| | - Miguel López
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain.
| |
Collapse
|
28
|
Winter C, Greene DM, Mavrogiorgou P, Schaper H, Sohr R, Bult-Ito A, Juckel G. Altered serotonergic and GABAergic neurotransmission in a mice model of obsessive-compulsive disorder. Behav Brain Res 2018; 337:240-245. [DOI: 10.1016/j.bbr.2017.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/07/2017] [Accepted: 09/07/2017] [Indexed: 01/04/2023]
|
29
|
Evaluation of food intake and Fos expression in serotonergic neurons of raphe nuclei after intracerebroventricular injection of adrenaline in free-feeding rats. Brain Res 2018; 1678:153-163. [DOI: 10.1016/j.brainres.2017.10.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 09/21/2017] [Accepted: 10/22/2017] [Indexed: 02/05/2023]
|
30
|
Zhang J, Fan Y, Raza MU, Zhan Y, Du XD, Patel PD, Zhu MY. The regulation of corticosteroid receptors in response to chronic social defeat. Neurochem Int 2017; 108:397-409. [DOI: 10.1016/j.neuint.2017.05.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 05/27/2017] [Accepted: 05/30/2017] [Indexed: 12/24/2022]
|
31
|
The protective effects of resveratrol on social stress-induced cytokine release and depressive-like behavior. Brain Behav Immun 2017; 59:147-157. [PMID: 27592314 PMCID: PMC5154920 DOI: 10.1016/j.bbi.2016.08.019] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/19/2016] [Accepted: 08/30/2016] [Indexed: 12/12/2022] Open
Abstract
Social stress is a risk factor for psychiatric disorders, however only a subset of the population is susceptible while others remain resilient. Inflammation has been linked to the pathogenesis of psychosocial disorders in humans and may underlie these individual differences. Using a resident-intruder paradigm capable of revealing individual differences in coping behavior and inflammatory responses, the present study determined if resveratrol (RSV; 0, 10, 30mg/kg/day) protected against persistent stress-induced inflammation in socially defeated rats. Furthermore, the antidepressant efficacy of RSV was evaluated using the sucrose preference test. Active coping rats were characterized by more time spent in upright postures and increased defeat latencies versus passive coping rats. Five days after defeat, flow cytometry revealed enhanced stimulation of proinflammatory proteins (IL-β, TNF-α) in spleen cells of passive rats as compared to active coping and controls, an effect that was blocked by both doses of RSV. Furthermore, only passive coping rats exhibited increased proinflammatory proteins (IL-1β, TNF-α, GM-CSF) in the locus coeruleus (LC), a noradrenergic brain region implicated in depression. Notably, only 30mg/kg RSV blocked LC neuroinflammation and importantly, was the only dose that blocked anhedonia. Alternatively, while stress had minimal impact on resting cytokines in the dorsal raphe (DR), RSV dose-dependently reduced DR cytokine expression. However, this did not result in changes in indoleamine 2,3-dioxygenase activity or serotonin levels. Taken together, these data suggest that social stress-induced depressive-like behavior evident in passive coping rats may be driven by stress-induced neuroinflammation and highlight natural anti-inflammatory agents to protect against social stress-related consequences.
Collapse
|
32
|
Xu Y. Brain Estrogens and Feeding Behavior. SEX AND GENDER FACTORS AFFECTING METABOLIC HOMEOSTASIS, DIABETES AND OBESITY 2017; 1043:337-357. [DOI: 10.1007/978-3-319-70178-3_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
33
|
Luo M, Li Y, Zhong W. Do dorsal raphe 5-HT neurons encode “beneficialness”? Neurobiol Learn Mem 2016; 135:40-49. [DOI: 10.1016/j.nlm.2016.08.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 08/15/2016] [Accepted: 08/17/2016] [Indexed: 10/21/2022]
|
34
|
Rysz M, Bromek E, Haduch A, Liskova B, Wójcikowski J, Daniel WA. The reverse role of the hypothalamic paraventricular (PVN) and arcuate (ARC) nuclei in the central serotonergic regulation of the liver cytochrome P450 isoform CYP2C11. Biochem Pharmacol 2016; 112:82-9. [DOI: 10.1016/j.bcp.2016.04.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 04/28/2016] [Indexed: 02/05/2023]
|
35
|
Commons KG. Ascending serotonin neuron diversity under two umbrellas. Brain Struct Funct 2016; 221:3347-60. [PMID: 26740230 DOI: 10.1007/s00429-015-1176-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 12/19/2015] [Indexed: 12/30/2022]
Abstract
Forebrain serotonin relevant for many psychological disorders arises in the hindbrain, primarily within the dorsal and median raphe nuclei (DR and MR). These nuclei are heterogeneous, containing several distinct groups of serotonin neurons. Here, new insight into the afferent and efferent connectivity of these areas is reviewed in correlation with their developmental origin. These data suggest that the caudal third of the DR, the area originally designated B6, may be misidentified as part of the DR as it shares many features of connectivity with the MR. By considering the rostral DR independently and affiliating the B6 to the MR, the diverse subgroups of serotonin neurons can be arranged with more coherence into two umbrella groups, each with distinctive domains of influence. Serotonin neurons within the rostral DR are uniquely interconnected with brain areas associated with emotion and motivation such as the amygdala, accumbens and ventral pallidum. In contrast serotonin neurons in the B6 and MR are characterized by their dominion over the septum and hippocampus. This distinction between the DR and B6/MR parallels their developmental origin and likely impacts their role in both behavior and psychopathology. Implications and further subdivisions within these areas are discussed.
Collapse
Affiliation(s)
- Kathryn G Commons
- Department of Anesthesiology, Perioperative, and Pain Medicine, Boston Children's Hospital, 300 Longwood Ave., Boston, MA, 02115, USA. .,Department of Anaesthesia, Harvard Medical School, Boston, USA.
| |
Collapse
|
36
|
Qesseveur G, Petit AC, Nguyen HT, Dahan L, Colle R, Rotenberg S, Seif I, Robert P, David D, Guilloux JP, Gardier AM, Verstuyft C, Becquemont L, Corruble E, Guiard BP. Genetic dysfunction of serotonin 2A receptor hampers response to antidepressant drugs: A translational approach. Neuropharmacology 2016; 105:142-153. [PMID: 26764241 DOI: 10.1016/j.neuropharm.2015.12.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/23/2015] [Accepted: 12/24/2015] [Indexed: 01/31/2023]
Abstract
Pharmacological studies have yielded valuable insights into the role of the serotonin 2A (5-HT2A) receptor in major depressive disorder (MDD) and antidepressant drugs (ADs) response. However, it is still unknown whether genetic variants in the HTR2A gene affect the therapeutic outcome of ADs and the mechanism underlying the regulation of such response remains poorly described. In this context, a translational human-mouse study offers a unique opportunity to address the possibility that variations in the HTR2A gene may represent a relevant marker to predict the efficacy of ADs. In a first part of this study, we investigated in depressed patients the effect of three HTR2A single nucleotide polymorphisms (SNPs), selected for their potential functional consequences on 5-HT2A receptor (rs6313, rs6314 and rs7333412), on response and remission rates after 3 months of antidepressant treatments. We also explored the consequences of the constitutive genetic inactivation of the 5-HT2A receptor (i.e. in 5-HT2A(-/-) mice) on the activity of acute and prolonged administration of SSRIs. Our clinical data indicate that GG patients for the rs7333412 SNP were less prone to respond to ADs than AA/AG patients. In the preclinical study, we demonstrated that the 5-HT2A receptor exerts an inhibitory influence on the neuronal activity of the serotonergic system after acute administration of SSRIs. However, while the chronic administration of the SSRIs escitalopram or fluoxetine elicited a progressive increased in the firing rate of 5-HT neurons in 5-HT2A(+/+) mice, it failed to do so in 5-HT2A(-/-) mutants. These electrophysiological impairments were associated with a decreased ability of the chronic administration of fluoxetine to stimulate hippocampal plasticity and to produce antidepressant-like activities. Genetic loss of the 5-HT2A receptor compromised the activity of chronic treatment with SSRIs, making this receptor a putative marker to predict ADs response.
Collapse
Affiliation(s)
- Gaël Qesseveur
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, Fac Pharmacie, Châtenay Malabry, 92290, France
| | - Anne Cécile Petit
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, CESP, Fac Médecine Paris Sud, 94275, Le Kremlin Bicêtre, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital de Bicêtre, Service de Psychiatrie, Le Kremlin Bicêtre, F-94275, France
| | - Hai Thanh Nguyen
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, Fac Pharmacie, Châtenay Malabry, 92290, France
| | - Lionel Dahan
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| | - Romain Colle
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, CESP, Fac Médecine Paris Sud, 94275, Le Kremlin Bicêtre, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital de Bicêtre, Service de Psychiatrie, Le Kremlin Bicêtre, F-94275, France
| | - Samuel Rotenberg
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, CESP, Fac Médecine Paris Sud, 94275, Le Kremlin Bicêtre, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital de Bicêtre, Service de Psychiatrie, Le Kremlin Bicêtre, F-94275, France
| | - Isabelle Seif
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, Fac Pharmacie, Châtenay Malabry, 92290, France
| | - Pauline Robert
- UMS IPSIT (INST. Paris-Saclay d'innovation Thérapeutique), Paris Sud, France
| | - Denis David
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, Fac Pharmacie, Châtenay Malabry, 92290, France
| | - Jean-Philippe Guilloux
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, Fac Pharmacie, Châtenay Malabry, 92290, France
| | - Alain M Gardier
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, Fac Pharmacie, Châtenay Malabry, 92290, France
| | - Céline Verstuyft
- INSERM U1184, Le Kremlin Bicêtre, F-94276, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital de Bicêtre, Service de Génétique moléculaire, Pharmacogénétique et Hormonologie, Le Kremlin Bicêtre, F-94275, France
| | - Laurent Becquemont
- INSERM U1184, Le Kremlin Bicêtre, F-94276, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital de Bicêtre, Service de Génétique moléculaire, Pharmacogénétique et Hormonologie, Le Kremlin Bicêtre, F-94275, France
| | - Emmanuelle Corruble
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, CESP, Fac Médecine Paris Sud, 94275, Le Kremlin Bicêtre, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital de Bicêtre, Service de Psychiatrie, Le Kremlin Bicêtre, F-94275, France.
| | - Bruno P Guiard
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, Fac Pharmacie, Châtenay Malabry, 92290, France; Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| |
Collapse
|
37
|
Hwa LS, Shimamoto A, Kayyali T, Norman KJ, Valentino RJ, DeBold JF, Miczek KA. Dissociation of μ-opioid receptor and CRF-R1 antagonist effects on escalated ethanol consumption and mPFC serotonin in C57BL/6J mice. Addict Biol 2016; 21:111-24. [PMID: 25262980 DOI: 10.1111/adb.12189] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Both the opioid antagonist naltrexone and corticotropin-releasing factor type-1 receptor (CRF-R1) antagonists have been investigated for the treatment of alcoholism. The current study examines the combination of naltrexone and CP154526 to reduce intermittent access ethanol drinking [intermittent access to alcohol (IAA)] in C57BL/6J male mice, and if these compounds reduce drinking via serotonergic mechanisms in the dorsal raphe nucleus (DRN). Systemic injections and chronic intracerebroventricular infusions of naltrexone, CP154526 or CP376395 transiently decreased IAA drinking. Immunohistochemistry revealed CRF-R1 or μ-opioid receptor immunoreactivity was co-localized in tryptophan hydroxylase (TPH)-immunoreactive neurons as well as non-TPH neurons in the DRN. Mice with a history of IAA or continuous access to alcohol were microinjected with artificial cerebral spinal fluid, naltrexone, CP154526 or the combination into the DRN or the median raphe nucleus (MRN). Either intra-DRN naltrexone or CP154526 reduced IAA in the initial 2 hours of fluid access, but the combination did not additively suppress IAA, suggesting a common mechanism via which these two compounds affect intermittent drinking. These alcohol-reducing effects were localized to the DRN of IAA drinkers, as intra-MRN injections only significantly suppressed water drinking, and continuous access drinkers were not affected by CRF-R1 antagonism. Extracellular serotonin was measured in the medial prefrontal cortex (mPFC) using in vivo microdialysis after intra-DRN microinjections in another group of mice. Intra-DRN CP154526 increased serotonin impulse flow to the mPFC while naltrexone did not. This suggests the mPFC may not be an essential location to intermittent drinking, as evidenced by different effects on serotonin signaling to the forebrain yet similar behavioral findings.
Collapse
Affiliation(s)
- Lara S. Hwa
- Department of Psychology; Tufts University; Medford MA USA
| | | | - Tala Kayyali
- Department of Psychology; Tufts University; Medford MA USA
| | | | - Rita J. Valentino
- Division of Stress Neurobiology; Children's Hospital of Philadelphia; Philadelphia PA USA
| | | | - Klaus A. Miczek
- Department of Psychology; Tufts University; Medford MA USA
- Department of Neuroscience; Tufts University; Medford MA USA
| |
Collapse
|
38
|
Teissier A, Chemiakine A, Inbar B, Bagchi S, Ray RS, Palmiter RD, Dymecki SM, Moore H, Ansorge MS. Activity of Raphé Serotonergic Neurons Controls Emotional Behaviors. Cell Rep 2015; 13:1965-76. [PMID: 26655908 DOI: 10.1016/j.celrep.2015.10.061] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 09/08/2015] [Accepted: 10/19/2015] [Indexed: 01/19/2023] Open
Abstract
Despite the well-established role of serotonin signaling in mood regulation, causal relationships between serotonergic neuronal activity and behavior remain poorly understood. Using a pharmacogenetic approach, we find that selectively increasing serotonergic neuronal activity in wild-type mice is anxiogenic and reduces floating in the forced-swim test, whereas inhibition has no effect on the same measures. In a developmental mouse model of altered emotional behavior, increased anxiety and depression-like behaviors correlate with reduced dorsal raphé and increased median raphé serotonergic activity. These mice display blunted responses to serotonergic stimulation and behavioral rescues through serotonergic inhibition. Furthermore, we identify opposing consequences of dorsal versus median raphé serotonergic neuron inhibition on floating behavior, together suggesting that median raphé hyperactivity increases anxiety, whereas a low dorsal/median raphé serotonergic activity ratio increases depression-like behavior. Thus, we find a critical role of serotonergic neuronal activity in emotional regulation and uncover opposing roles of median and dorsal raphé function.
Collapse
Affiliation(s)
- Anne Teissier
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Alexei Chemiakine
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Benjamin Inbar
- New York State Psychiatric Institute, New York, NY 10032, USA
| | - Sneha Bagchi
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Russell S Ray
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Richard D Palmiter
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Susan M Dymecki
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Holly Moore
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | - Mark S Ansorge
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA.
| |
Collapse
|
39
|
Iwai M, Muroi Y, Kinoshita KI, Ishii T. Serotonin modulates the dehydration-induced changes in tolerance for bitter water. Physiol Behav 2015; 151:545-50. [PMID: 26325013 DOI: 10.1016/j.physbeh.2015.08.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/25/2015] [Accepted: 08/26/2015] [Indexed: 11/26/2022]
Abstract
Drinking behavior is regulated by endogenous factors such as the hydration condition of animals and exogenous factors such as the taste of ingested fluids. These factors have been suggested to interact with each other via serotonergic (5-HT) signaling to regulate drinking behavior. In the present study, we examined how dehydration affects the intake of bitter water, which suppresses drinking behavior, via 5-HT signaling. Water deprivation increased water intake for 1h, depending on the duration of water deprivation. The intake of 1mM quinine, which is a bitter tastant, was lower than that of water in mice deprived of water for 24h but not 48 h. We next examined the involvement of the dorsal raphe nucleus (DRN) and median raphe nucleus (MRN), which contain a large population of 5-HT neurons, in changing tolerance for quinine intake after water deprivation. The intake of quinine following water deprivation for 24h, but not 48 h, increased the number of tryptophan hydroxylase-positive neurons expressing c-Fos in the DRN, but not in the MRN. Moreover, administration of paroxetine, a selective serotonin reuptake inhibitor, decreased the intake of quinine solution, but not water, in mice deprived of water for 48 h, indicating that paroxetine treatment restored the aversion to quinine. These results suggest that unresponsiveness of 5-HT neurons in the DRN may be involved in the dehydration-induced increase in tolerance for bitter water.
Collapse
Affiliation(s)
- Masaki Iwai
- Department of Basic Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan.
| | - Yoshikage Muroi
- Department of Basic Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan.
| | - Ken-ichi Kinoshita
- Department of Basic Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan.
| | - Toshiaki Ishii
- Department of Basic Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan.
| |
Collapse
|
40
|
Rysz M, Bromek E, Haduch A, Sadakierska-Chudy A, Daniel WA. Damage to the Brain Serotonergic System Increases the Expression of Liver Cytochrome P450. Drug Metab Dispos 2015; 43:1345-52. [PMID: 26059263 DOI: 10.1124/dmd.115.064980] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/09/2015] [Indexed: 02/13/2025] Open
Abstract
Genes coding for cytochrome P450 are regulated by endogenous hormones such as the growth hormone, corticosteroids, thyroid, and sex hormones. Secretion of these hormones is regulated by the respective hypothalamus-pituitary-secretory organ axes. Since the brain sends its serotonergic projections from the raphe nuclei to the hypothalamus, we have assumed that damage to these nuclei may affect the neuroendocrine regulation of cytochrome P450 expression in the liver. Thereby, 5,7-dihydroxytryptamine (5,7-DHT), a serotonergic neurotoxin, was injected into the dorsal and median raphe nuclei of male Wistar rats. Ten days after the neurotoxin injections, the brain concentrations of neurotransmitters, serum hormone, and cytokine levels, as well as the expression of cytochrome P450 in the liver were measured. Injection of 5,7-DHT decreased serotonin concentration in the brain followed by a significant rise in the levels of the growth hormone, corticosterone, and testosterone, and a drop in triiodothyronine concentration in the serum. No changes in interleukin (IL) levels (IL-2 and IL-6) were observed. Simultaneously, the activity and protein level of liver CYP1A, CYP3A1, and CYP2C11 rose (the activity of CYP2A/2B/2C6/2D was not significantly changed). Similarly, the mRNA levels of CYP1A1, CYP1A2, CYP2C11, and CYP3A1 were elevated. This is the first report demonstrating the effect of intracerebral administration of serotonergic neurotoxin on liver cytochrome P450. The obtained results indicate involvement of the brain serotonergic system in the neuroendocrine regulation of liver cytochrome P450 expression. The physiologic and pharmacological significance of the findings is discussed.
Collapse
Affiliation(s)
- Marta Rysz
- Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Ewa Bromek
- Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Anna Haduch
- Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | | | | |
Collapse
|
41
|
Nobuta H, Cilio MR, Danhaive O, Tsai HH, Tupal S, Chang SM, Murnen A, Kreitzer F, Bravo V, Czeisler C, Gokozan HN, Gygli P, Bush S, Weese-Mayer DE, Conklin B, Yee SP, Huang EJ, Gray PA, Rowitch D, Otero JJ. Dysregulation of locus coeruleus development in congenital central hypoventilation syndrome. Acta Neuropathol 2015; 130:171-83. [PMID: 25975378 PMCID: PMC4503865 DOI: 10.1007/s00401-015-1441-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 05/01/2015] [Accepted: 05/02/2015] [Indexed: 12/29/2022]
Abstract
Human congenital central hypoventilation syndrome (CCHS), resulting from mutations in transcription factor PHOX2B, manifests with impaired responses to hypoxemia and hypercapnia especially during sleep. To identify brainstem structures developmentally affected in CCHS, we analyzed two postmortem neonatal-lethal cases with confirmed polyalanine repeat expansion (PARM) or Non-PARM (PHOX2B∆8) mutation of PHOX2B. Both human cases showed neuronal losses within the locus coeruleus (LC), which is important for central noradrenergic signaling. Using a conditionally active transgenic mouse model of the PHOX2B∆8 mutation, we found that early embryonic expression (<E10.5) caused failure of LC neuronal specification and perinatal respiratory lethality. In contrast, later onset (E11.5) of PHOX2B∆8 expression was not deleterious to LC development and perinatal respiratory lethality was rescued, despite failure of chemosensor retrotrapezoid nucleus formation. Our findings indicate that early-onset mutant PHOX2B expression inhibits LC neuronal development in CCHS. They further suggest that such mutations result in dysregulation of central noradrenergic signaling, and therefore, potential for early pharmacologic intervention in humans with CCHS.
Collapse
|
42
|
Versteeg RI, Serlie MJ, Kalsbeek A, la Fleur SE. Serotonin, a possible intermediate between disturbed circadian rhythms and metabolic disease. Neuroscience 2015; 301:155-67. [PMID: 26047725 DOI: 10.1016/j.neuroscience.2015.05.067] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 05/21/2015] [Accepted: 05/27/2015] [Indexed: 01/27/2023]
Abstract
It is evident that eating in misalignment with the biological clock (such as in shift work, eating late at night and skipping breakfast) is associated with increased risk for obesity and diabetes. The biological clock located in the suprachiasmatic nucleus dictates energy balance including feeding behavior and glucose metabolism. Besides eating and sleeping patterns, glucose metabolism also exhibits clear diurnal variations with higher blood glucose concentrations, glucose tolerance and insulin sensitivity prior to waking up. The daily variation in plasma glucose concentrations in rats, is independent of the rhythm in feeding behavior. On the other hand, feeding itself has profound effects on glucose metabolism, but differential effects occur depending on the time of the day. We here review data showing that a disturbed diurnal eating pattern results in alterations in glucose metabolism induced by a disrupted circadian clock. We first describe the role of central serotonin on feeding behavior and glucose metabolism and subsequently describe the effects of central serotonin on the circadian system. We next explore the interaction between the serotonergic system and the circadian clock in conditions of disrupted diurnal rhythms in feeding and how this might be involved in the metabolic dysregulation that occurs with chronodisruption.
Collapse
Affiliation(s)
- R I Versteeg
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - M J Serlie
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - A Kalsbeek
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - S E la Fleur
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
43
|
da Silva ES, Flores RA, Cella EC, Levone BR, Taschetto AP, Kochenborger L, Terenzi MG, Faria MS, Paschoalini MA. Blockade of median raphe nucleus α1-adrenoceptor subtypes increases food intake in rats. Pharmacol Biochem Behav 2014; 124:350-5. [DOI: 10.1016/j.pbb.2014.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 06/11/2014] [Accepted: 06/15/2014] [Indexed: 10/25/2022]
|
44
|
Abstract
The dorsal raphe nucleus (DRN) in the midbrain is a key center for serotonin (5-hydroxytryptamine; 5-HT)-expressing neurons. Serotonergic neurons in the DRN have been theorized to encode punishment by opposing the reward signaling of dopamine neurons. Here, we show that DRN neurons encode reward, but not punishment, through 5-HT and glutamate. Optogenetic stimulation of DRN Pet-1 neurons reinforces mice to explore the stimulation-coupled spatial region, shifts sucrose preference, drives optical self-stimulation, and directs sensory discrimination learning. DRN Pet-1 neurons increase their firing activity during reward tasks, and this activation can be used to rapidly change neuronal activity patterns in the cortex. Although DRN Pet-1 neurons are often associated with 5-HT, they also release glutamate, and both neurotransmitters contribute to reward signaling. These experiments demonstrate the ability of DRN neurons to organize reward behaviors and might provide insights into the underlying mechanisms of learning facilitation and anhedonia treatment.
Collapse
|
45
|
Gq/5-HT2c receptor signals activate a local GABAergic inhibitory feedback circuit to modulate serotonergic firing and anxiety in mice. Proc Natl Acad Sci U S A 2014; 111:6479-84. [PMID: 24733892 DOI: 10.1073/pnas.1321576111] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Serotonin 2c receptors (5-HT2c-Rs) are drug targets for certain mental disorders, including schizophrenia, depression, and anxiety. 5-HT2c-Rs are expressed throughout the brain, making it difficult to link behavioral changes to circuit specific receptor expression. Various 5-HT-Rs, including 5-HT2c-Rs, are found in the dorsal raphe nucleus (DRN); however, the function of 5-HT2c-Rs and their influence on the serotonergic signals mediating mood disorders remain unclear. To investigate the role of 5-HT2c-Rs in the DRN in mice, we developed a melanopsin-based optogenetic probe for activation of Gq signals in cellular domains, where 5-HT2c-Rs are localized. Our results demonstrate that precise temporal control of Gq signals in 5-HT2c-R domains in GABAergic neurons upstream of 5-HT neurons provides negative feedback regulation of serotonergic firing to modulate anxiety-like behavior in mice.
Collapse
|
46
|
Abstract
More than half of all patients with diabetes develop neuropathic disorders affecting the distal sensory and/or motor nerves, or autonomic or cranial nerve functions. Glycemic control can decrease the incidence of neuropathy but is not adequate alone to prevent or treat the disease. This chapter introduces diabetic neuropathy with a morphological description of the disease then describes our current understanding of metabolic and molecular mechanisms that contribute to neurovascular dysfunctions. Key mechanisms include glucose and lipid imbalances and insulin resistance that are interconnected via oxidative stress, inflammation, and altered gene expression. These complex interactions should be considered for the development of new treatment strategies against the onset or progression of neuropathy. Advances in understanding the combined metabolic stressors and the novel study of epigenetics suggest new therapeutic targets to combat this morbid and intractable disease affecting millions of patients with type 1 or type 2 diabetes.
Collapse
|
47
|
Ito H, Yanase M, Yamashita A, Kitabatake C, Hamada A, Suhara Y, Narita M, Ikegami D, Sakai H, Yamazaki M, Narita M. Analysis of sleep disorders under pain using an optogenetic tool: possible involvement of the activation of dorsal raphe nucleus-serotonergic neurons. Mol Brain 2013; 6:59. [PMID: 24370235 PMCID: PMC3879646 DOI: 10.1186/1756-6606-6-59] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 12/20/2013] [Indexed: 11/10/2022] Open
Abstract
Background Several etiological reports have shown that chronic pain significantly interferes with sleep. Inadequate sleep due to chronic pain may contribute to the stressful negative consequences of living with pain. However, the neurophysiological mechanism by which chronic pain affects sleep-arousal patterns is as yet unknown. Although serotonin (5-HT) was proposed to be responsible for sleep regulation, whether the activity of 5-HTergic neurons in the dorsal raphe nucleus (DRN) is affected by chronic pain has been studied only infrequently. On the other hand, the recent development of optogenetic tools has provided a valuable opportunity to regulate the activity in genetically targeted neural populations with high spatial and temporal precision. In the present study, we investigated whether chronic pain could induce sleep dysregulation while changing the activity of DRN-5-HTergic neurons. Furthermore, we sought to physiologically activate the DRN with channelrhodopsin-2 (ChR2) to identify a causal role for the DRN-5-HT system in promoting and maintaining wakefulness using optogenetics. Results We produced a sciatic nerve ligation model by tying a tight ligature around approximately one-third to one-half the diameter of the sciatic nerve. In mice with nerve ligation, we confirmed an increase in wakefulness and a decrease in non-rapid eye movement (NREM) sleep as monitored by electroencephalogram (EEG). Microinjection of the retrograde tracer fluoro-gold (FG) into the prefrontal cortex (PFC) revealed several retrogradely labeled-cells in the DRN. The key finding of the present study was that the levels of 5-HT released in the PFC by the electrical stimulation of DRN neurons were significantly increased in mice with sciatic nerve ligation. Using optogenetic tools in mice, we found a causal relationship among DRN neuron firing, cortical activity and sleep-to-wake transitions. In particular, the activation of DRN-5-HTergic neurons produced a significant increase in wakefulness and a significant decrease in NREM sleep. The duration of NREM sleep episodes was significantly decreased during photostimulation in these mice. Conclusions These results suggest that neuropathic pain accelerates the activity of DRN-5-HTergic neurons. Although further loss-of-function experiments are required, we hypothesize that this activation in DRN neurons may, at least in part, correlate with sleep dysregulation under a neuropathic pain-like state.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Mitsuaki Yamazaki
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| | | |
Collapse
|
48
|
Abstract
Although the role of the median raphe nucleus (MRN) in the regulation of anxiety has received less attention than that of the dorsal raphe nucleus (DRN) there is substantial evidence supporting this function. Reported results with different animal models of anxiety in rats show that whereas inactivation of serotonergic neurons in the MRN causes anxiolysis, the stimulation of the same neurons is anxiogenic. In particular, studies using the elevated T-maze comparing serotonergic interventions in the MRN and in the DRN indicate that the former affect only the inhibitory avoidance task, which has been related to generalized anxiety. In contrast, similar operations in the DRN change both the inhibitory avoidance and the one-way escape task, the latter being representative of panic disorder. Simultaneous injections of 5-HT-acting drugs in the MRN and in the dorsal hippocampus (DH) suggest that the MRN-DH pathway mediates the regulatory function of the MRN in anxiety. Overall, the results discussed in this review point to a relevant role of the MRN in the regulation of anxiety, but not panic, through the 5-HT pathway that innervates the DH.
Collapse
Affiliation(s)
- Telma Gcs Andrade
- 1Department of Biological Science, São Paulo State University-UNESP, Assis, Brazil
| | | | | |
Collapse
|
49
|
|
50
|
Own LS, Iqbal R, Patel PD. Maternal separation alters serotonergic and HPA axis gene expression independent of separation duration in c57bl/6 mice. Brain Res 2013; 1515:29-38. [DOI: 10.1016/j.brainres.2013.03.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 01/21/2013] [Accepted: 03/13/2013] [Indexed: 12/29/2022]
|