1
|
Guo CCG, Xu Y, Shan L, Foka K, Memoli S, Mulveen C, Gijsbrechts B, Verheij MM, Homberg JR. Quantifying multilabeled brain cells in the whole prefrontal cortex reveals reduced inhibitory and a subtype of excitatory neuronal marker expression in serotonin transporter knockout rats. Cereb Cortex 2025; 35:bhae486. [PMID: 39932853 DOI: 10.1093/cercor/bhae486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 11/25/2024] [Accepted: 12/05/2024] [Indexed: 02/13/2025] Open
Abstract
The prefrontal cortex regulates emotions and is influenced by serotonin. Rodents lacking the serotonin transporter (5-HTT) show increased anxiety and changes in excitatory and inhibitory cell markers in the prefrontal cortex. However, these observations are constrained by limitations in brain representation and cell segmentation, as standard immunohistochemistry is inadequate to consider volume variations in regions of interest. We utilized the deep learning network of the StarDist method in combination with novel open-source methods for automated cell counts in a wide range of prefrontal cortex subregions. We found that 5-HTT knockout rats displayed increased anxiety and diminished relative numbers of subclass excitatory VGluT2+ and activated ΔFosB+ cells in the infralimbic and prelimbic cortices and of inhibitory GAD67+ cells in the prelimbic cortex. Anxiety levels and ΔFosB cell counts were positively correlated in wild-type, but not in knockout, rats. In conclusion, we present a novel method to quantify whole brain subregions of multilabeled cells in animal models and demonstrate reduced excitatory and inhibitory neuronal marker expression in prefrontal cortex subregions of 5-HTT knockout rats.
Collapse
Affiliation(s)
- Chao Ciu-Gwok Guo
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Kapittelweg 29, 6525 EN, Nijmegen, the Netherlands
| | - Yifan Xu
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Kapittelweg 29, 6525 EN, Nijmegen, the Netherlands
| | - Ling Shan
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA, Amsterdam, the Netherlands
| | - Kyriaki Foka
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | - Simone Memoli
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Kapittelweg 29, 6525 EN, Nijmegen, the Netherlands
| | - Calum Mulveen
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Kapittelweg 29, 6525 EN, Nijmegen, the Netherlands
| | - Barend Gijsbrechts
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Kapittelweg 29, 6525 EN, Nijmegen, the Netherlands
| | - Michel M Verheij
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Kapittelweg 29, 6525 EN, Nijmegen, the Netherlands
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Kapittelweg 29, 6525 EN, Nijmegen, the Netherlands
| |
Collapse
|
2
|
Leser FS, Júnyor FDS, Pagnoncelli IB, Delgado AB, Medeiros I, Nóbrega ACC, Andrade BDS, de Lima MN, da Silva NE, Jacob L, Boyé K, Geraldo LHM, de Souza AMT, Maron-Gutierrez T, Castro-Faria-Neto H, Follmer C, Braga C, Neves GA, Eichmann A, Romão LF, Lima FRS. CCL21-CCR7 blockade prevents neuroinflammation and degeneration in Parkinson's disease models. J Neuroinflammation 2025; 22:31. [PMID: 39894839 PMCID: PMC11789347 DOI: 10.1186/s12974-024-03318-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/02/2024] [Indexed: 02/04/2025] Open
Abstract
Parkinson's disease (PD) is a progressive degenerative disease of the central nervous system associated with neuroinflammation and microglial cell activation. Chemokine signaling regulates neuron-glia communication and triggers a microglial inflammatory profile. Herein, we identified the neuronal chemokine CCL21 as a major cause of microglial cell imbalance through the CCR7 receptor pathway with therapeutic implications for PD. In humans, we found that CCL21 transcript expression was increased in dopaminergic neurons (DANs) of the substantia nigra in PD patients. CCL21 and CCR7 expressions were spatially associated with brain regional vulnerability to synucleinopathies, as well as with the expression of microglial activation, neuroinflammation, and degeneration-related genes. Also, in mouse models of PD, we showed that CCL21 was overexpressed in DANs in vivo and in vitro. Mechanistically, neuronal CCL21 was shown to regulate microglial cell migration, proliferation, and activation in a CCR7-dependent manner through both canonical (PI3K/AKT) and non-canonical (ERK1/2/JNK) signaling pathways. Finally, we demonstrated that navarixin, a clinically relevant chemokine inhibitor with high affinity for the CCR7 receptor, could block CCL21 effects on microglia and prevent neurodegeneration and behavioral deficits in two mouse models of PD induced with either α-synuclein oligomers (αSynO) or 3,4-dihydroxyphenylacetaldehyde (DOPAL). Altogether, our data indicate that navarixin blocks CCL21/CCR7-mediated neuron-microglia communication and could be used as a therapeutic strategy against PD.
Collapse
Affiliation(s)
- Felipe Saceanu Leser
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France
| | - Flavio de Souza Júnyor
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Iohanna Bianca Pagnoncelli
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Anna Beatriz Delgado
- Laboratory of Neurobiology Applied to Biomedicine, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Isabelle Medeiros
- Laboratory of Neurobiology Applied to Biomedicine, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Ana Clara Campanelli Nóbrega
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Brenda da Silva Andrade
- Laboratory of Molecular Pharmacology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Maiara Nascimento de Lima
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Nícolas Emanoel da Silva
- Laboratory Molecular Modeling & QSAR, Pharmaceutical Sciences Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Laurent Jacob
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France
| | - Kevin Boyé
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France
| | - Luiz Henrique Medeiros Geraldo
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France
- Department of Internal Medicine, Department of Cellular and Molecular Physiology, Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, 06510-3221, USA
| | - Alessandra Mendonça Teles de Souza
- Laboratory Molecular Modeling & QSAR, Pharmaceutical Sciences Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Tatiana Maron-Gutierrez
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Hugo Castro-Faria-Neto
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Cristian Follmer
- Laboratory of Physical Chemistry of Proteins and Peptides (Lafipp), Chemistry Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-909, Brazil
| | - Carolina Braga
- Núcleo Multidisciplinar de Pesquisas em Biologia, NUMPEX-Bio, Universidade Federal do Rio de Janeiro, Duque de Caxias, RJ, 25240-005, Brasil
| | - Gilda Angela Neves
- Laboratory of Molecular Pharmacology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Anne Eichmann
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France.
- Department of Internal Medicine, Department of Cellular and Molecular Physiology, Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, 06510-3221, USA.
| | - Luciana Ferreira Romão
- Laboratory of Neurobiology Applied to Biomedicine, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Flavia Regina Souza Lima
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil.
| |
Collapse
|
3
|
Lima J, Panayi MC, Sharp T, McHugh SB, Bannerman DM. More and Less Fear in Serotonin Transporter Knockout Mice. GENES, BRAIN, AND BEHAVIOR 2025; 24:e70016. [PMID: 39917838 PMCID: PMC11803413 DOI: 10.1111/gbb.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 12/22/2024] [Accepted: 01/14/2025] [Indexed: 02/11/2025]
Abstract
Recent theories suggest that reduced serotonin transporter (5-HTT) function, which increases serotonin (5-HT) levels at the synapse, enhances neural plasticity and affects sensitivity to environmental cues. This may promote learning about emotionally relevant events. However, the boundaries that define such emotional learning remain to be established. This was investigated using 5-HTT knockout (5-HTTKO) mice which provide a model of long-term elevated 5-HT transmission and are associated with increased anxiety. Compared to wild-type controls, 5-HTTKO mice were faster to discriminate between an auditory cue that predicted footshock (CS+) and a cue predicting no footshock (CS-). Notably, this enhanced discrimination performance was driven not by faster learning that the CS+ predicted footshock, but rather by faster learning that the CS- cue signals the absence of footshock and thus provides temporary relief from fear/anxiety. Similarly, 5-HTTKO mice were also faster to reduce their fear of the CS+ cue during subsequent extinction. These findings are consistent with facilitated inhibitory learning that predicts the absence of potential threats in 5-HTTKO mice. However, 5-HTTKO mice also exhibited increased generalisation of fear learning about ambiguous aversive cues in a novel context, different from the training context. Thus, 5-HTTKO mice can exhibit both more and less fear compared to wild-type controls. Taken together, our results support the idea that loss of 5-HTT function, and corresponding increases in synaptic 5-HT availability, may facilitate learning by priming of aversive memories. This both facilitates inhibitory learning for fear memories but also enhances generalisation of fear.
Collapse
Affiliation(s)
- João Lima
- Department of Experimental PsychologyUniversity of OxfordOxfordUK
- Danish Research Centre for Magnetic Resonance (DRCMR), Department of Radiology and Nuclear MedicineCopenhagen University Hospital—Amager and HvidovreCopenhagenDenmark
| | - Marios C. Panayi
- Department of Experimental PsychologyUniversity of OxfordOxfordUK
- School of PsychologyUniversity of New South WalesSydneyNew South WalesAustralia
| | - Trevor Sharp
- Department of PharmacologyUniversity of OxfordOxfordUK
| | - Stephen B. McHugh
- Department of Experimental PsychologyUniversity of OxfordOxfordUK
- Medical Research Council Brain Network Dynamics UnitOxfordUK
| | | |
Collapse
|
4
|
Bale R, Doshi G. Deciphering the role of siRNA in anxiety and depression. Eur J Pharmacol 2024; 981:176868. [PMID: 39128805 DOI: 10.1016/j.ejphar.2024.176868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 07/02/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Anxiety and depression are central nervous system illnesses that are among the most prevalent medical concerns of the twenty-first century. Patients with this condition and their families bear psychological, financial, and societal hardship. There are currently restrictions when utilizing the conventional course of treatment. RNA interference is expected to become an essential approach in anxiety and depression due to its potent and targeted gene silencing. Silencing of genes by post-transcriptional modification is the mechanism of action of small interfering RNA (siRNA). The suppression of genes linked to disease is typically accomplished by siRNA molecules in an efficient and targeted manner. Unfavourable immune responses, off-target effects, naked siRNA instability, nuclease vulnerability, and the requirement to create an appropriate delivery method are some of the challenges facing the clinical application of siRNA. This review focuses on the use of siRNA in the treatment of anxiety and depression.
Collapse
Affiliation(s)
- Rajeshwari Bale
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India.
| |
Collapse
|
5
|
Illera JC, Jimenez-Blanco F, Centenera L, Gil-Cabrera F, Crespo B, Lopez PR, Silvan G, Caceres S. Addressing Combative Behaviour in Spanish Bulls by Measuring Hormonal Indicators. Vet Sci 2024; 11:182. [PMID: 38668449 PMCID: PMC11053816 DOI: 10.3390/vetsci11040182] [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: 12/28/2023] [Revised: 02/02/2024] [Accepted: 04/19/2024] [Indexed: 04/29/2024] Open
Abstract
The fighting bull is characterised by its natural aggressiveness, but the physiological mechanisms that underlie its aggressive behaviour are poorly studied. This study determines the hormonal component of aggressiveness in fighting bulls by analysing their behaviour during a fight and correlating it to their serotonin, dopamine and testosterone levels. We also determine whether aggressive behaviour can be estimated in calves. Using 195 animals, samples were obtained when the animals were calves and after 5 years. Aggressiveness scores were obtained by an observational method during bullfights, and serotonin, dopamine and testosterone levels were determined in all animals using validated enzyme immunoassay kits. The results revealed a strong correlation of serotonin and dopamine levels with aggressiveness scores in bulls during fights, but no correlation was found with respect to testosterone. These correlations led to established cut-off point and linear regression curves to obtain expected aggressiveness scores for calves at shoeing. There were no significant differences between the expected scores obtained in calves and the observed scores in bulls. Therefore, this study demonstrates that hormone determination in calves may be a great indicator of combativeness in bulls and can reliably be used in the selection of fighting bulls.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gema Silvan
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (J.C.I.); (F.J.-B.); (L.C.); (F.G.-C.); (B.C.); (P.R.L.); (S.C.)
| | | |
Collapse
|
6
|
Homberg JR, Brivio P, Greven CU, Calabrese F. Individuals being high in their sensitivity to the environment: Are sensitive period changes in play? Neurosci Biobehav Rev 2024; 159:105605. [PMID: 38417743 DOI: 10.1016/j.neubiorev.2024.105605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/13/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024]
Abstract
All individuals on planet earth are sensitive to the environment, but some more than others. These individual differences in sensitivity to environments are seen across many animal species including humans, and can influence personalities as well as vulnerability and resilience to mental disorders. Yet, little is known about the underlying brain mechanisms. Key genes that contribute to individual differences in environmental sensitivity are the serotonin transporter, dopamine D4 receptor and brain-derived neurotrophic factor genes. By synthesizing neurodevelopmental findings of these genetic factors, and discussing them through the lens of mechanisms related to sensitive periods, which are phases of heightened neuronal plasticity during which a certain network is being finetuned by experiences, we propose that these genetic factors delay but extend postnatal sensitive periods. This may explain why sensitive individuals show behavioral features that are characteristic of a young brain state at the level of sensory information processing, such as reduced filtering or blockade of irrelevant information, resulting in a sensory processing system that 'keeps all options open'.
Collapse
Affiliation(s)
- Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Paola Brivio
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Corina U Greven
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Karakter Child and Adolescent Psychiatry University Center, Nijmegen, the Netherlands; King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry Center, London, United Kingdom
| | - Francesca Calabrese
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
7
|
Huang C, van Wijnen AJ, Im HJ. Serotonin Transporter (5-Hydroxytryptamine Transporter, SERT, SLC6A4) and Sodium-dependent Reuptake Inhibitors as Modulators of Pain Behaviors and Analgesic Responses. THE JOURNAL OF PAIN 2024; 25:618-631. [PMID: 37852405 PMCID: PMC11781314 DOI: 10.1016/j.jpain.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/20/2023]
Abstract
The serotonin transporter (5-hydroxytryptamine transporter [5-HTT], Serotonin Transporter (SERT), SLC6A4) modulates the activity of serotonin via sodium-dependent reuptake. Given the established importance of serotonin in the control of pain, 5-HTT has received much interest in studies of pain states and as a pharmacological target for serotonin reuptake inhibitors (SRIs). Animal models expressing varying levels of 5-HTT activity show marked differences in pain behaviors and analgesic responses, as well as many serotonin-related physiological effects. In humans, functional nucleotide variations in the SLC6A4 gene, which encodes the serotonin transporter 5-HTT, are associated with certain pathologic pain conditions and differences in responses to pharmacological therapy. These findings collectively reflect the importance of 5-HTT in the intricate physiology and management of pain, as well as the scientific and clinical challenges that need to be considered for the optimization of 5-HTT-related analgesic therapies. PERSPECTIVE: The serotonin transporter 5-HTT/SCL6A4 is sensitive to pharmacological SRIs. Experimental studies on the physiological functions of serotonin, as well as genetic mouse models and clinical phenotype/genotype correlations of nucleotide variation in the human 5-HTT/SCL6A4 gene, provide new insights for the use of SRIs in chronic pain management.
Collapse
Affiliation(s)
- Cary Huang
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois; Department of Anesthesiology, NewYork-Presbyterian/Weill Cornell Medical Center, New York, New York.
| | - Andre J van Wijnen
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois; Department of Biochemistry, University of Vermont, Burlington, Vermont.
| | - Hee-Jeong Im
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois; Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, Illinois.
| |
Collapse
|
8
|
Dahchour A, Ward RJ. Changes in serotonin neurotransmission as assayed by microdialysis after acute, intermittent or chronic ethanol administration and withdrawal. Fundam Clin Pharmacol 2024; 38:42-59. [PMID: 37712258 DOI: 10.1111/fcp.12949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 07/08/2023] [Accepted: 08/07/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND The serotonergic neurotransmitter system is involved in many ethanol-induced changes, including many behavioural alterations, as well as contributing to alcohol dependence and its withdrawal. AIMS This review has evaluated microdialysis studies where alterations in the serotonin system, that is, serotonin, 5-HT, or its metabolite 5-hydroxyindoleacetic acid, 5-HIAA, have been reported during different ethanol intoxication states, as well as in animals showing alcohol preference or not. Changes in 5-HT receptors and the 5-HT transporter are briefly reviewed to comprehend the significance of changes in microdialysate 5-HT concentrations. MATERIALS AND METHODS Changes in 5-HT content following acute, chronic and during ethanol withdrawal states are evaluated. In addition, the serotoninergic system was assessed in animals that have been genetically selected for alcohol preference to ascertain whether changes in this monoamine microdialysate content may contribute to alcohol preference. RESULTS AND DISCUSSION Changes occurred in 5-HT signalling in the limbic brain regions, increasing after acute ethanol administration in specific brain regions, particularly at higher doses, while chronic alcohol exposure essentially decreased serotonergic transmission. Such changes may play a pivotal role in emotion-driven craving and relapse. Depending on the dosage, mode of administration and consumption rate, ethanol affects specific brain regions in different ways, enhancing or reducing 5-HT microdialysate content, thereby inducing behavioural and cognitive functions and enhancing ethanol consumption. CONCLUSION Microdialysis studies demonstrated that ethanol induces several alterations in 5-HT content as well as its metabolites, 5-HIAA and 5-HTOL, not only in its release from a specific brain region but also in the modifications of its different receptor subtypes and its transporter.
Collapse
Affiliation(s)
- Abdelkader Dahchour
- Clinical Neurosciences Laboratory, Faculty of Medicine and Pharmacy. Department of Biology, Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Roberta J Ward
- Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
9
|
Nguyen TTH, Nguyen CM, Huynh MA, Vu HH, Nguyen TK, Nguyen NT. Field effect transistor based wearable biosensors for healthcare monitoring. J Nanobiotechnology 2023; 21:411. [PMID: 37936115 PMCID: PMC10629051 DOI: 10.1186/s12951-023-02153-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
The rapid advancement of wearable biosensors has revolutionized healthcare monitoring by screening in a non-invasive and continuous manner. Among various sensing techniques, field-effect transistor (FET)-based wearable biosensors attract increasing attention due to their advantages such as label-free detection, fast response, easy operation, and capability of integration. This review explores the innovative developments and applications of FET-based wearable biosensors for healthcare monitoring. Beginning with an introduction to the significance of wearable biosensors, the paper gives an overview of structural and operational principles of FETs, providing insights into their diverse classifications. Next, the paper discusses the fabrication methods, semiconductor surface modification techniques and gate surface functionalization strategies. This background lays the foundation for exploring specific FET-based biosensor designs, including enzyme, antibody and nanobody, aptamer, as well as ion-sensitive membrane sensors. Subsequently, the paper investigates the incorporation of FET-based biosensors in monitoring biomarkers present in physiological fluids such as sweat, tears, saliva, and skin interstitial fluid (ISF). Finally, we address challenges, technical issues, and opportunities related to FET-based biosensor applications. This comprehensive review underscores the transformative potential of FET-based wearable biosensors in healthcare monitoring. By offering a multidimensional perspective on device design, fabrication, functionalization and applications, this paper aims to serve as a valuable resource for researchers in the field of biosensing technology and personalized healthcare.
Collapse
Affiliation(s)
- Thi Thanh-Ha Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD, 4111, Australia
- School of Engineering and Built Environment, Griffith University, Nathan, QLD, 4111, Australia
| | - Cong Minh Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD, 4111, Australia
- School of Environment and Science (ESC), Griffith University, Nathan, QLD, 4111, Australia
| | - Minh Anh Huynh
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD, 4111, Australia
- School of Engineering and Built Environment, Griffith University, Nathan, QLD, 4111, Australia
| | - Hoang Huy Vu
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD, 4111, Australia
- School of Engineering and Built Environment, Griffith University, Nathan, QLD, 4111, Australia
| | - Tuan-Khoa Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD, 4111, Australia
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD, 4111, Australia.
| |
Collapse
|
10
|
MORIYA Y, KASAHARA Y, ISHIHARA K, HALL FS, HAGINO Y, HEN R, IKEDA K, UHL GR, SORA I. Heterozygous and homozygous gene knockout of the 5-HT1B receptor have different effects on methamphetamine-induced behavioral sensitization. Behav Pharmacol 2023; 34:393-403. [PMID: 37668157 PMCID: PMC10527357 DOI: 10.1097/fbp.0000000000000745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
The psychostimulant drug methamphetamine (METH) causes euphoria in humans and locomotor hyperactivity in rodents by acting on the mesolimbic dopamine (DA) pathway and has severe abuse and addiction liability. Behavioral sensitization, an increased behavioral response to a drug with repeated administration, can persist for many months after the last administration. Research has shown that the serotonin 1B (5-HT1B) receptor plays a critical role in the development and maintenance of drug addiction, as well as other addictive behaviors. This study examined the role of 5-HT1B receptors in METH-induced locomotor sensitization using 5-HT1B knockout (KO) mice. To clarify the action of METH in 5-HT1B KO mice the effects of METH on extracellular levels of DA (DAec) and 5-HT (5-HTec) in the caudate putamen (CPu) and the nucleus accumbens (NAc) were examined. Locomotor sensitization and extracellular monoamine levels were determined in wild-type mice (5-HT1B +/+), heterozygous 5-HT1B receptor KO (5-HT1B +/-) mice and homozygous 5-HT1B receptor KO mice (5-HT1B -/-). Behavioral sensitization to METH was enhanced in 5-HT1B -/- mice compared to 5-HT1B +/+ mice but was attenuated in 5-HT1B +/- mice compared to 5-HT1B +/+ and 5-HT1B -/- mice. In vivo, microdialysis demonstrated that acute administration of METH increases DAec levels in the CPu and NAc of 5-HT1B KO mice compared to saline groups. In 5-HT1B +/- mice, METH increased 5-HTec levels in the CPu, and DAec levels in the NAc were higher than in others.5-HT1B receptors play an important role in regulating METH-induced behavioral sensitization.
Collapse
Affiliation(s)
- Yuki MORIYA
- Department of Biological Psychiatry, Graduate School of Medicine, Sendai, Tohoku University, Japan
- Department of Disaster Psychiatry, International Research Institute of Disaster Science (IRIDeS), Graduate School of Medicine, Tohoku University, Sendai, Japan
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yoshiyuki KASAHARA
- Department of Biological Psychiatry, Graduate School of Medicine, Sendai, Tohoku University, Japan
- Department of Disaster Psychiatry, International Research Institute of Disaster Science (IRIDeS), Graduate School of Medicine, Tohoku University, Sendai, Japan
- Department of Maternal and Fetal Therapeutics, Tohoku University Graduate, School of Medicine, Sendai, Japan
- Department of Maternal and Child Healthcare Medical Science, Tohoku, University Graduate School of Medicine, Sendai, Japan
| | - Kana ISHIHARA
- Department of Biological Psychiatry, Graduate School of Medicine, Sendai, Tohoku University, Japan
| | - F. Scott HALL
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH, USA
| | - Yoko HAGINO
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - René HEN
- Department of Neuroscience and Pharmacology, Columbia University Medical Center, NY, USA
| | - Kazutaka IKEDA
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - George R. UHL
- Departments of Neurology and Pharmacology, University of Maryland School of Medicine, and VA Maryland Healthcare System, MD, USA
| | - Ichiro SORA
- Department of Biological Psychiatry, Graduate School of Medicine, Sendai, Tohoku University, Japan
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
11
|
Moncrieff J, Cooper RE, Stockmann T, Amendola S, Hengartner MP, Horowitz MA. The serotonin theory of depression: a systematic umbrella review of the evidence. Mol Psychiatry 2023; 28:3243-3256. [PMID: 35854107 PMCID: PMC10618090 DOI: 10.1038/s41380-022-01661-0] [Citation(s) in RCA: 363] [Impact Index Per Article: 181.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 05/31/2022] [Accepted: 06/07/2022] [Indexed: 12/31/2022]
Abstract
The serotonin hypothesis of depression is still influential. We aimed to synthesise and evaluate evidence on whether depression is associated with lowered serotonin concentration or activity in a systematic umbrella review of the principal relevant areas of research. PubMed, EMBASE and PsycINFO were searched using terms appropriate to each area of research, from their inception until December 2020. Systematic reviews, meta-analyses and large data-set analyses in the following areas were identified: serotonin and serotonin metabolite, 5-HIAA, concentrations in body fluids; serotonin 5-HT1A receptor binding; serotonin transporter (SERT) levels measured by imaging or at post-mortem; tryptophan depletion studies; SERT gene associations and SERT gene-environment interactions. Studies of depression associated with physical conditions and specific subtypes of depression (e.g. bipolar depression) were excluded. Two independent reviewers extracted the data and assessed the quality of included studies using the AMSTAR-2, an adapted AMSTAR-2, or the STREGA for a large genetic study. The certainty of study results was assessed using a modified version of the GRADE. We did not synthesise results of individual meta-analyses because they included overlapping studies. The review was registered with PROSPERO (CRD42020207203). 17 studies were included: 12 systematic reviews and meta-analyses, 1 collaborative meta-analysis, 1 meta-analysis of large cohort studies, 1 systematic review and narrative synthesis, 1 genetic association study and 1 umbrella review. Quality of reviews was variable with some genetic studies of high quality. Two meta-analyses of overlapping studies examining the serotonin metabolite, 5-HIAA, showed no association with depression (largest n = 1002). One meta-analysis of cohort studies of plasma serotonin showed no relationship with depression, and evidence that lowered serotonin concentration was associated with antidepressant use (n = 1869). Two meta-analyses of overlapping studies examining the 5-HT1A receptor (largest n = 561), and three meta-analyses of overlapping studies examining SERT binding (largest n = 1845) showed weak and inconsistent evidence of reduced binding in some areas, which would be consistent with increased synaptic availability of serotonin in people with depression, if this was the original, causal abnormaly. However, effects of prior antidepressant use were not reliably excluded. One meta-analysis of tryptophan depletion studies found no effect in most healthy volunteers (n = 566), but weak evidence of an effect in those with a family history of depression (n = 75). Another systematic review (n = 342) and a sample of ten subsequent studies (n = 407) found no effect in volunteers. No systematic review of tryptophan depletion studies has been performed since 2007. The two largest and highest quality studies of the SERT gene, one genetic association study (n = 115,257) and one collaborative meta-analysis (n = 43,165), revealed no evidence of an association with depression, or of an interaction between genotype, stress and depression. The main areas of serotonin research provide no consistent evidence of there being an association between serotonin and depression, and no support for the hypothesis that depression is caused by lowered serotonin activity or concentrations. Some evidence was consistent with the possibility that long-term antidepressant use reduces serotonin concentration.
Collapse
Affiliation(s)
- Joanna Moncrieff
- Division of Psychiatry, University College London, London, UK.
- Research and Development Department, Goodmayes Hospital, North East London NHS Foundation Trust, Essex, UK.
| | - Ruth E Cooper
- Faculty of Education, Health and Human Sciences, University of Greenwich, London, UK
| | | | - Simone Amendola
- Department of Dynamic and Clinical Psychology, and Health Studies, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Michael P Hengartner
- Department of Applied Psychology, Zurich University of Applied Sciences, Zurich, Switzerland
| | - Mark A Horowitz
- Division of Psychiatry, University College London, London, UK
- Research and Development Department, Goodmayes Hospital, North East London NHS Foundation Trust, Essex, UK
| |
Collapse
|
12
|
Shoji H, Ikeda K, Miyakawa T. Behavioral phenotype, intestinal microbiome, and brain neuronal activity of male serotonin transporter knockout mice. Mol Brain 2023; 16:32. [PMID: 36991468 PMCID: PMC10061809 DOI: 10.1186/s13041-023-01020-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/16/2023] [Indexed: 03/31/2023] Open
Abstract
The serotonin transporter (5-HTT) plays a critical role in the regulation of serotonin neurotransmission. Mice genetically deficient in 5-HTT expression have been used to study the physiological functions of 5-HTT in the brain and have been proposed as a potential animal model for neuropsychiatric and neurodevelopmental disorders. Recent studies have provided evidence for a link between the gut-brain axis and mood disorders. However, the effects of 5-HTT deficiency on gut microbiota, brain function, and behavior remain to be fully characterized. Here we investigated the effects of 5-HTT deficiency on different types of behavior, the gut microbiome, and brain c-Fos expression as a marker of neuronal activation in response to the forced swim test for assessing depression-related behavior in male 5-HTT knockout mice. Behavioral analysis using a battery of 16 different tests showed that 5-HTT-/- mice exhibited markedly reduced locomotor activity, decreased pain sensitivity, reduced motor function, increased anxiety-like and depression-related behavior, altered social behavior in novel and familiar environments, normal working memory, enhanced spatial reference memory, and impaired fear memory compared to 5-HTT+/+ mice. 5-HTT+/- mice showed slightly reduced locomotor activity and impaired social behavior compared to 5-HTT+/+ mice. Analysis of 16S rRNA gene amplicons showed that 5-HTT-/- mice had altered gut microbiota abundances, such as a decrease in Allobaculum, Bifidobacterium, Clostridium sensu stricto, and Turicibacter, compared to 5-HTT+/+ mice. This study also showed that after exposure to the forced swim test, the number of c-Fos-positive cells was higher in the paraventricular thalamus and lateral hypothalamus and was lower in the prefrontal cortical regions, nucleus accumbens shell, dorsolateral septal nucleus, hippocampal regions, and ventromedial hypothalamus in 5-HTT-/- mice than in 5-HTT+/+ mice. These phenotypes of 5-HTT-/- mice partially recapitulate clinical observations in humans with major depressive disorder. The present findings indicate that 5-HTT-deficient mice serve as a good and valid animal model to study anxiety and depression with altered gut microbial composition and abnormal neuronal activity in the brain, highlighting the importance of 5-HTT in brain function and the mechanisms underlying the regulation of anxiety and depression.
Collapse
Affiliation(s)
- Hirotaka Shoji
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan.
| |
Collapse
|
13
|
Zhao C, Man T, Cao Y, Weiss PS, Monbouquette HG, Andrews AM. Flexible and Implantable Polyimide Aptamer-Field-Effect Transistor Biosensors. ACS Sens 2022; 7:3644-3653. [PMID: 36399772 PMCID: PMC9982941 DOI: 10.1021/acssensors.2c01909] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Monitoring neurochemical signaling across time scales is critical to understanding how brains encode and store information. Flexible (vs stiff) devices have been shown to improve in vivo monitoring, particularly over longer times, by reducing tissue damage and immunological responses. Here, we report our initial steps toward developing flexible and implantable neuroprobes with aptamer-field-effect transistor (FET) biosensors for neurotransmitter monitoring. A high-throughput process was developed to fabricate thin, flexible polyimide probes using microelectromechanical-system (MEMS) technologies, where 150 flexible probes were fabricated on each 4 in. Si wafer. Probes were 150 μm wide and 7 μm thick with two FETs per tip. The bending stiffness was 1.2 × 10-11 N·m2. Semiconductor thin films (3 nm In2O3) were functionalized with DNA aptamers for target recognition, which produces aptamer conformational rearrangements detected via changes in FET conductance. Flexible aptamer-FET neuroprobes detected serotonin at femtomolar concentrations in high-ionic strength artificial cerebrospinal fluid. A straightforward implantation process was developed, where microfabricated Si carrier devices assisted with implantation such that flexible neuroprobes detected physiological relevant serotonin in a tissue-hydrogel brain mimic.
Collapse
Affiliation(s)
- Chuanzhen Zhao
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States,California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Tianxing Man
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Yan Cao
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States,Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Paul S. Weiss
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States,California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States,Departments of Bioengineering and Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Harold G. Monbouquette
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States,Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Anne M. Andrews
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States,California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States,Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience & Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States,To whom correspondence should be addressed to:
| |
Collapse
|
14
|
Otsuka T, Le HT, Thein ZL, Ihara H, Sato F, Nakao T, Kohsaka A. Deficiency of the circadian clock gene Rev-erbα induces mood disorder-like behaviours and dysregulation of the serotonergic system in mice. Physiol Behav 2022; 256:113960. [PMID: 36115382 DOI: 10.1016/j.physbeh.2022.113960] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 10/31/2022]
Abstract
Mood disorders such as depression, anxiety, and bipolar disorder are highly associated with disrupted daily rhythms of activity, which are often observed in shift work and sleep disturbance in humans. Recent studies have proposed the REV-ERBα protein as a key circadian nuclear receptor that links behavioural rhythms to mood regulation. However, how the Rev-erbα gene participates in the regulation of mood remains poorly understood. Here, we show that the regulation of the serotonergic (5-HTergic) system, which plays a central role in stress-induced mood behaviours, is markedly disrupted in Rev-erbα-/- mice. Rev-erbα-/- mice exhibit both negative and positive behavioural phenotypes, including anxiety-like and mania-like behaviours, when subjected to a stressful environment. Importantly, Rev-erbα-/- mice show a significant decrease in the expression of a gene that encodes the rate-limiting enzyme of serotonin (5-HT) synthesis in the raphe nuclei (RN). In addition, 5-HT levels in Rev-erbα-/- mice are significantly reduced in the prefrontal cortex, which receives strong inputs from the RN and controls stress-related behaviours. Our findings indicate that Rev-erbα plays an important role in controlling the 5-HTergic system and thus regulates mood and behaviour.
Collapse
Affiliation(s)
- Tsuyoshi Otsuka
- Faculty of Applied Biological Sciences, Gifu University, Gifu, 501-1193, Japan; The Second Department of Physiology, Wakayama Medical University, Wakayama 641-8509, Japan.
| | - Hue Thi Le
- The Second Department of Physiology, Wakayama Medical University, Wakayama 641-8509, Japan; Department of Biomedical Engineering, National Cerebral and Cardiovascular Center, Osaka, 564-8565, Japan
| | - Zaw Lin Thein
- The Second Department of Physiology, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Hayato Ihara
- The Department of Radioisotope Laboratory Center, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Fuyuki Sato
- Department of Diagnostic Pathology, Shizuoka Cancer Center, Suntogun, Shizuoka 411-8777, Japan; The Departments of Pathology, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Tomomi Nakao
- The Second Department of Physiology, Wakayama Medical University, Wakayama 641-8509, Japan; The First Department of Internal Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Akira Kohsaka
- The Second Department of Physiology, Wakayama Medical University, Wakayama 641-8509, Japan
| |
Collapse
|
15
|
Armand S, Ozenne B, Svart N, Frøkjaer VG, Knudsen GM, Fisher PM, Stenbaek DS. Brain serotonin transporter is associated with cognitive-affective biases in healthy individuals. Hum Brain Mapp 2022; 43:4174-4184. [PMID: 35607850 PMCID: PMC9374883 DOI: 10.1002/hbm.25946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/21/2022] [Accepted: 05/02/2022] [Indexed: 12/26/2022] Open
Abstract
Cognitive affective biases describe the tendency to process negative information or positive information over the other. These biases can be modulated by changing extracellular serotonin (5-HT) levels in the brain, for example, by pharmacologically blocking and downregulating the 5-HT transporter (5-HTT), which remediates negative affective bias. This suggests that higher levels of 5-HTT are linked to a priority of negative information over positive, but this link remains to be tested in vivo in healthy individuals. We, therefore, evaluated the association between 5-HTT levels, as measured with [11 C]DASB positron emission tomography (PET), and affective biases, hypothesising that higher 5-HTT levels are associated with a more negative bias. We included 98 healthy individuals with measures of [11 C]DASB binding potential (BPND ) and affective biases using The Emotional Faces Identification Task by subtracting the per cent hit rate for happy from that of sad faces (EFITAB ). We evaluated the association between [11 C]DASB BPND and EFITAB in a linear latent variable model, with the latent variable (5-HTTLV ) modelled from [11 C]DASB BPND in the fronto-striatal and fronto-limbic networks implicated in affective cognition. We observed an inverse association between 5-HTTLV and EFITAB (β = -8% EFITAB per unit 5-HTTLV , CI = -14% to -3%, p = .002). These findings show that higher 5-HTT levels are linked to a more negative bias in healthy individuals. High 5-HTT supposedly leads to high clearance of 5-HT, and thus, a negative bias could result from low extracellular 5-HT. Future studies must reveal if a similar inverse association exists in individuals with affective disorders.
Collapse
Affiliation(s)
- Sophia Armand
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Brice Ozenne
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Public Health, Section of Biostatistics, University of Copenhagen, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Nanna Svart
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Vibe G Frøkjaer
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Psychiatric Center Copenhagen, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Gitte M Knudsen
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Patrick M Fisher
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Dea S Stenbaek
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Psychology, University of Copenhagen, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
16
|
Dagher M, Perrotta KA, Erwin SA, Hachisuka A, Iyer R, Masmanidis SC, Yang H, Andrews AM. Optogenetic Stimulation of Midbrain Dopamine Neurons Produces Striatal Serotonin Release. ACS Chem Neurosci 2022; 13:946-958. [PMID: 35312275 PMCID: PMC9040469 DOI: 10.1021/acschemneuro.1c00715] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Targeting neurons with light-driven opsins is widely used to investigate cell-specific responses. We transfected midbrain dopamine neurons with the excitatory opsin Chrimson. Extracellular basal and stimulated neurotransmitter levels in the dorsal striatum were measured by microdialysis in awake mice. Optical activation of dopamine cell bodies evoked terminal dopamine release in the striatum. Multiplexed analysis of dialysate samples revealed that the evoked dopamine was accompanied by temporally coupled increases in striatal 3-methoxytyramine, an extracellular dopamine metabolite, and in serotonin. We investigated a mechanism for dopamine-serotonin interactions involving striatal dopamine receptors. However, the evoked serotonin associated with optical stimulation of dopamine neurons was not abolished by striatal D1- or D2-like receptor inhibition. Although the mechanisms underlying the coupling of striatal dopamine and serotonin remain unclear, these findings illustrate advantages of multiplexed measurements for uncovering functional interactions between neurotransmitter systems. Furthermore, they suggest that the output of optogenetic manipulations may extend beyond opsin-expressing neuronal populations.
Collapse
Affiliation(s)
- Merel Dagher
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Katie A. Perrotta
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Sara A. Erwin
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Ayaka Hachisuka
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Rahul Iyer
- Department of Electrical Engineering, University of California, Los Angeles, Los Angeles, CA, 94720
| | - Sotiris C. Masmanidis
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA 90095, United States
- Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, United States
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Hongyan Yang
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience & Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Anne M. Andrews
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, United States
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, United States
- Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, United States
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience & Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA 90095, United States
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, United States
| |
Collapse
|
17
|
Thorne BN, Ellenbroek BA, Day DJ. The serotonin reuptake transporter modulates mitochondrial copy number and mitochondrial respiratory complex gene expression in the frontal cortex and cerebellum in a sexually dimorphic manner. J Neurosci Res 2022; 100:869-879. [PMID: 35043462 DOI: 10.1002/jnr.25010] [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: 09/14/2021] [Revised: 12/05/2021] [Accepted: 12/28/2021] [Indexed: 12/27/2022]
Abstract
Neuropsychiatric and neurodevelopmental disorders such as major depressive disorder (MDD) and autism spectrum disorder (ASD) are complex conditions attributed to both genetic and environmental factors. There is a growing body of evidence showing that serotonergic signaling and mitochondrial dysfunction contribute to the pathophysiology of these disorders and are linked as signaling through specific serotonin (5-HT) receptors drives mitochondrial biogenesis. The serotonin transporter (SERT) is important in these disorders as it regulates synaptic serotonin and therapeutically is the target of selective serotonin reuptake inhibitors which are a major class of anti-depressant drug. Human allelic variants of the serotonin transporter-linked polymorphic region (5-HTTLPR) such as the S/S variant, are associated with reduced SERT expression and increased susceptibility for developing neuropsychiatric disorders. Using a rat model that is haploinsufficient for SERT and displays reduced SERT expression similar to the human S/S variant, we demonstrate that reduced SERT expression modulates mitochondrial copy number and expression of respiratory chain electron transfer components in the brain. In the frontal cortex, genotype-related trends were opposing for males and females, such that reduced SERT expression led to increased expression of the Complex I subunit mt-Nd1 in males but reduced expression in females. Our findings suggest that SERT expression and serotonergic signaling have a role in regulating mitochondrial biogenesis and adenosine triphosphate (ATP) production in the brain. We speculate that the sexual dimorphism in mitochondrial abundance and gene expression contributes to the sex bias found in the incidence of neuropsychiatric disorders such as MDD and ASD.
Collapse
Affiliation(s)
- Bryony N Thorne
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Bart A Ellenbroek
- School of Psychology, Victoria University of Wellington Faculty of Science, Wellington, New Zealand
| | - Darren J Day
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
18
|
Sato A, Kotajima-Murakami H, Tanaka M, Katoh Y, Ikeda K. Influence of Prenatal Drug Exposure, Maternal Inflammation, and Parental Aging on the Development of Autism Spectrum Disorder. Front Psychiatry 2022; 13:821455. [PMID: 35222122 PMCID: PMC8863673 DOI: 10.3389/fpsyt.2022.821455] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/12/2022] [Indexed: 12/17/2022] Open
Abstract
Autism spectrum disorder (ASD) affects reciprocal social interaction and produces abnormal repetitive, restrictive behaviors and interests. The diverse causes of ASD are divided into genetic alterations and environmental risks. The prevalence of ASD has been rising for several decades, which might be related to environmental risks as it is difficult to consider that the prevalence of genetic disorders related to ASD would increase suddenly. The latter includes (1) exposure to medications, such as valproic acid (VPA) and selective serotonin reuptake inhibitors (SSRIs) (2), maternal complications during pregnancy, including infection and hypertensive disorders of pregnancy, and (3) high parental age. Epidemiological studies have indicated a pathogenetic role of prenatal exposure to VPA and maternal inflammation in the development of ASD. VPA is considered to exert its deleterious effects on the fetal brain through several distinct mechanisms, such as alterations of γ-aminobutyric acid signaling, the inhibition of histone deacetylase, the disruption of folic acid metabolism, and the activation of mammalian target of rapamycin. Maternal inflammation that is caused by different stimuli converges on a higher load of proinflammatory cytokines in the fetal brain. Rodent models of maternal exposure to SSRIs generate ASD-like behavior in offspring, but clinical correlations with these preclinical findings are inconclusive. Hypertensive disorders of pregnancy and advanced parental age increase the risk of ASD in humans, but the mechanisms have been poorly investigated in animal models. Evidence of the mechanisms by which environmental factors are related to ASD is discussed, which may contribute to the development of preventive and therapeutic interventions for ASD.
Collapse
Affiliation(s)
- Atsushi Sato
- Department of Pediatrics, The University of Tokyo Hospital, Tokyo, Japan.,Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | - Miho Tanaka
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Psychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshihisa Katoh
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
19
|
Zhao C, Cheung KM, Huang IW, Yang H, Nakatsuka N, Liu W, Cao Y, Man T, Weiss PS, Monbouquette HG, Andrews AM. Implantable aptamer-field-effect transistor neuroprobes for in vivo neurotransmitter monitoring. SCIENCE ADVANCES 2021; 7:eabj7422. [PMID: 34818033 PMCID: PMC8612678 DOI: 10.1126/sciadv.abj7422] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
While tools for monitoring in vivo electrophysiology have been extensively developed, neurochemical recording technologies remain limited. Nevertheless, chemical communication via neurotransmitters plays central roles in brain information processing. We developed implantable aptamer–field-effect transistor (FET) neuroprobes for monitoring neurotransmitters. Neuroprobes were fabricated using high-throughput microelectromechanical system (MEMS) technologies, where 150 probes with shanks of either 150- or 50-μm widths and thicknesses were fabricated on 4-inch Si wafers. Nanoscale FETs with ultrathin (~3 to 4 nm) In2O3 semiconductor films were prepared using sol-gel processing. The In2O3 surfaces were coupled with synthetic oligonucleotide receptors (aptamers) to recognize and to detect the neurotransmitter serotonin. Aptamer-FET neuroprobes enabled femtomolar serotonin detection limits in brain tissue with minimal biofouling. Stimulated serotonin release was detected in vivo. This study opens opportunities for integrated neural activity recordings at high spatiotemporal resolution by combining these aptamer-FET sensors with other types of Si-based implantable probes to advance our understanding of brain function.
Collapse
Affiliation(s)
- Chuanzhen Zhao
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kevin M. Cheung
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - I-Wen Huang
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hongyan Yang
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nako Nakatsuka
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Wenfei Liu
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yan Cao
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tianxing Man
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Paul S. Weiss
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Harold G. Monbouquette
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Anne M. Andrews
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Corresponding author.
| |
Collapse
|
20
|
Movassaghi CS, Perrotta KA, Yang H, Iyer R, Cheng X, Dagher M, Fillol MA, Andrews AM. Simultaneous serotonin and dopamine monitoring across timescales by rapid pulse voltammetry with partial least squares regression. Anal Bioanal Chem 2021; 413:6747-6767. [PMID: 34686897 PMCID: PMC8551120 DOI: 10.1007/s00216-021-03665-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/11/2021] [Accepted: 09/14/2021] [Indexed: 11/12/2022]
Abstract
Many voltammetry methods have been developed to monitor brain extracellular dopamine levels. Fewer approaches have been successful in detecting serotonin in vivo. No voltammetric techniques are currently available to monitor both neurotransmitters simultaneously across timescales, even though they play integrated roles in modulating behavior. We provide proof-of-concept for rapid pulse voltammetry coupled with partial least squares regression (RPV-PLSR), an approach adapted from multi-electrode systems (i.e., electronic tongues) used to identify multiple components in complex environments. We exploited small differences in analyte redox profiles to select pulse steps for RPV waveforms. Using an intentionally designed pulse strategy combined with custom instrumentation and analysis software, we monitored basal and stimulated levels of dopamine and serotonin. In addition to faradaic currents, capacitive currents were important factors in analyte identification arguing against background subtraction. Compared to fast-scan cyclic voltammetry-principal components regression (FSCV-PCR), RPV-PLSR better differentiated and quantified basal and stimulated dopamine and serotonin associated with striatal recording electrode position, optical stimulation frequency, and serotonin reuptake inhibition. The RPV-PLSR approach can be generalized to other electrochemically active neurotransmitters and provides a feedback pipeline for future optimization of multi-analyte, fit-for-purpose waveforms and machine learning approaches to data analysis.
Collapse
Affiliation(s)
- Cameron S Movassaghi
- Department of Chemistry & Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Katie A Perrotta
- Department of Chemistry & Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hongyan Yang
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Rahul Iyer
- Department of Electrical Engineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xinyi Cheng
- Department of Chemistry & Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Merel Dagher
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Miguel Alcañiz Fillol
- Interuniversity Research Institute for Molecular Recognition and Technological Development, Universitat Politècnica de València - Universitat de València, Camino de Vera s/n, 46022, Valencia, Spain.
| | - Anne M Andrews
- Department of Chemistry & Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
21
|
Chan JZ, Fernandes MF, Hashemi A, Grewal RS, Mardian EB, Bradley RM, Duncan RE. Age-associated increase in anxiety-like behavior in Lpaatδ/Agpat4 knockout mice. CURRENT RESEARCH IN BEHAVIORAL SCIENCES 2021. [DOI: 10.1016/j.crbeha.2021.100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
22
|
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the most commonly prescribed medications for psychiatric disorders, yet they leave the majority of patients without full symptom relief. Therefore, a major research challenge is to identify novel targets for the improved treatment of these disorders. SSRIs act by blocking the serotonin transporter (SERT), the high-affinity, low-capacity, uptake-1 transporter for serotonin. Other classes of antidepressant work by blocking the norepinephrine or dopamine transporters (NET and DAT), the high-affinity, low-capacity uptake-1 transporters for norepinephrine and dopamine, or by blocking combinations of SERT, NET, and DAT. It has been proposed that uptake-2 transporters, which include organic cation transporters (OCTs) and the plasma membrane monoamine transporter (PMAT), undermine the therapeutic utility of uptake-1 acting antidepressants. Uptake-2 transporters for monoamines have low affinity for these neurotransmitters, but a high capacity to transport them. Thus, activity of these transporters may limit the increase of extracellular monoamines thought to be essential for ultimate therapeutic benefit. Here preclinical evidence supporting a role for OCT2, OCT3, and PMAT in behaviors relevant to psychiatric disorders is presented. Importantly, preclinical evidence revealing these transporters as targets for the development of novel therapeutics for psychiatric disorders is discussed.
Collapse
|
23
|
Popp S, Schmitt-Böhrer A, Langer S, Hofmann U, Hommers L, Schuh K, Frantz S, Lesch KP, Frey A. 5-HTT Deficiency in Male Mice Affects Healing and Behavior after Myocardial Infarction. J Clin Med 2021; 10:jcm10143104. [PMID: 34300270 PMCID: PMC8308004 DOI: 10.3390/jcm10143104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 12/13/2022] Open
Abstract
Anxiety disorders and depression are common comorbidities in cardiac patients. Mice lacking the serotonin transporter (5-HTT) exhibit increased anxiety-like behavior. However, the role of 5-HTT deficiency on cardiac aging, and on healing and remodeling processes after myocardial infarction (MI), remains unclear. Cardiological evaluation of experimentally naïve male mice revealed a mild cardiac dysfunction in ≥4-month-old 5-HTT knockout (−/−) animals. Following induction of chronic cardiac dysfunction (CCD) by MI vs. sham operation 5-HTT−/− mice with infarct sizes >30% experienced 100% mortality, while 50% of 5-HTT+/− and 37% of 5-HTT+/+ animals with large MI survived the 8-week observation period. Surviving (sham and MI < 30%) 5-HTT−/− mutants displayed reduced exploratory activity and increased anxiety-like behavior in different approach-avoidance tasks. However, CCD failed to provoke a depressive-like behavioral response in either 5-Htt genotype. Mechanistic analyses were performed on mice 3 days post-MI. Electrocardiography, histology and FACS of inflammatory cells revealed no abnormalities. However, gene expression of inflammation-related cytokines (TGF-β, TNF-α, IL-6) and MMP-2, a protein involved in the breakdown of extracellular matrix, was significantly increased in 5-HTT−/− mice after MI. This study shows that 5-HTT deficiency leads to age-dependent cardiac dysfunction and disrupted early healing after MI probably due to alterations of inflammatory processes in mice.
Collapse
Affiliation(s)
- Sandy Popp
- Comprehensive Heart Failure Center, University Hospital of Würzburg, 97078 Würzburg, Germany; (S.P.); (S.L.); (U.H.); (S.F.); (K.-P.L.)
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, Division of Molecular Psychiatry, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Angelika Schmitt-Böhrer
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, 97080 Würzburg, Germany; (A.S.-B.); (L.H.)
| | - Simon Langer
- Comprehensive Heart Failure Center, University Hospital of Würzburg, 97078 Würzburg, Germany; (S.P.); (S.L.); (U.H.); (S.F.); (K.-P.L.)
| | - Ulrich Hofmann
- Comprehensive Heart Failure Center, University Hospital of Würzburg, 97078 Würzburg, Germany; (S.P.); (S.L.); (U.H.); (S.F.); (K.-P.L.)
- Medical Clinic and Policlinic I, University Hospital of Würzburg, 97080 Würzburg, Germany
- Interdisciplinary Center for Clinical Research, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Leif Hommers
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, 97080 Würzburg, Germany; (A.S.-B.); (L.H.)
- Interdisciplinary Center for Clinical Research, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Kai Schuh
- Institute of Physiology I, University of Würzburg, 97070 Würzburg, Germany;
| | - Stefan Frantz
- Comprehensive Heart Failure Center, University Hospital of Würzburg, 97078 Würzburg, Germany; (S.P.); (S.L.); (U.H.); (S.F.); (K.-P.L.)
- Medical Clinic and Policlinic I, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Klaus-Peter Lesch
- Comprehensive Heart Failure Center, University Hospital of Würzburg, 97078 Würzburg, Germany; (S.P.); (S.L.); (U.H.); (S.F.); (K.-P.L.)
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, Division of Molecular Psychiatry, University Hospital of Würzburg, 97080 Würzburg, Germany
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, 97080 Würzburg, Germany; (A.S.-B.); (L.H.)
- Department of Translational Neuroscience, School for Mental Health and Neuroscience, Maastricht University, 6229 Maastricht, The Netherlands
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Anna Frey
- Comprehensive Heart Failure Center, University Hospital of Würzburg, 97078 Würzburg, Germany; (S.P.); (S.L.); (U.H.); (S.F.); (K.-P.L.)
- Medical Clinic and Policlinic I, University Hospital of Würzburg, 97080 Würzburg, Germany
- Interdisciplinary Center for Clinical Research, University Hospital of Würzburg, 97080 Würzburg, Germany
- Correspondence: ; Tel.: +49-931-201-39927
| |
Collapse
|
24
|
Wilson C, Rogers J, Chen F, Li S, Adlard PA, Hannan AJ, Renoir T. Exercise ameliorates aberrant synaptic plasticity without enhancing adult-born cell survival in the hippocampus of serotonin transporter knockout mice. Brain Struct Funct 2021; 226:1991-1999. [PMID: 34052925 DOI: 10.1007/s00429-021-02283-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 04/21/2021] [Indexed: 12/28/2022]
Abstract
Deficits in hippocampal cellular and synaptic plasticity are frequently associated with cognitive and mood disorders, and indeed common mechanisms of antidepressants are thought to involve neuroplastic processes. Here, we investigate hippocampal adult-born cell survival and synaptic plasticity (long-term potentiation, LTP, and long-term depression, LTD) in serotonin transporter (5-HTT) knockout (KO) mice. From 8 weeks of age, mice either continued in standard-housing conditions or were given access to voluntary running wheels for 1 month. Electrophysiology was performed on hippocampal slices to measure LTP and LTD, and immunohistochemistry was used to assess cell proliferation and subsequent survival in the dentate gyrus. The results revealed a reduced LTP in 5-HTT KO mice that was restored to wild-type (WT) levels after chronic exercise. While LTD appeared normal in 5-HTT KO, exercise decreased the magnitude of LTD in both WT and 5-HTT KO mice. Furthermore, although 5-HTT KO mice had normal hippocampal adult-born cell survival, they did not benefit from the pro-proliferative effects of exercise observed in WT animals. Taken together, these findings suggest that reduced 5-HTT expression is associated with significant alterations to functional neuroplasticity. Interestingly, 5-HTT appeared necessary for exercise-induced augmentation of adult-born hippocampal cell survival, yet exercise corrected the LTP impairment displayed by 5-HTT KO mice. Together, our findings further highlight the salience of serotonergic signalling in mediating the neurophysiological benefits of exercise.
Collapse
Affiliation(s)
- Carey Wilson
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia
| | - Jake Rogers
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia
| | - Feng Chen
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia
| | - Shanshan Li
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia
| | - Paul A Adlard
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia.,Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia.,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Melbourne, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia. .,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Melbourne, Australia.
| |
Collapse
|
25
|
Dalvi-Garcia F, Fonseca LL, Vasconcelos ATR, Hedin-Pereira C, Voit EO. A model of dopamine and serotonin-kynurenine metabolism in cortisolemia: Implications for depression. PLoS Comput Biol 2021; 17:e1008956. [PMID: 33970902 PMCID: PMC8136856 DOI: 10.1371/journal.pcbi.1008956] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/20/2021] [Accepted: 04/10/2021] [Indexed: 12/31/2022] Open
Abstract
A major factor contributing to the etiology of depression is a neurochemical imbalance of the dopaminergic and serotonergic systems, which is caused by persistently high levels of circulating stress hormones. Here, a computational model is proposed to investigate the interplay between dopaminergic and serotonergic-kynurenine metabolism under cortisolemia and its consequences for the onset of depression. The model was formulated as a set of nonlinear ordinary differential equations represented with power-law functions. Parameter values were obtained from experimental data reported in the literature, biological databases, and other general information, and subsequently fine-tuned through optimization. Model simulations predict that changes in the kynurenine pathway, caused by elevated levels of cortisol, can increase the risk of neurotoxicity and lead to increased levels of 3,4-dihydroxyphenylaceltahyde (DOPAL) and 5-hydroxyindoleacetaldehyde (5-HIAL). These aldehydes contribute to alpha-synuclein aggregation and may cause mitochondrial fragmentation. Further model analysis demonstrated that the inhibition of both serotonin transport and kynurenine-3-monooxygenase decreased the levels of DOPAL and 5-HIAL and the neurotoxic risk often associated with depression. The mathematical model was also able to predict a novel role of the dopamine and serotonin metabolites DOPAL and 5-HIAL in the ethiology of depression, which is facilitated through increased cortisol levels. Finally, the model analysis suggests treatment with a combination of inhibitors of serotonin transport and kynurenine-3-monooxygenase as a potentially effective pharmacological strategy to revert the slow-down in monoamine neurotransmission that is often triggered by inflammation.
Collapse
Affiliation(s)
- Felipe Dalvi-Garcia
- Bioinformatics Lab, National Laboratory for Scientific Computing, Petrópolis, Rio de Janeiro, Brazil
- School of Medicine and Surgery, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luis L. Fonseca
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Ana Tereza R. Vasconcelos
- Bioinformatics Lab, National Laboratory for Scientific Computing, Petrópolis, Rio de Janeiro, Brazil
| | - Cecilia Hedin-Pereira
- Center of Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eberhard O. Voit
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| |
Collapse
|
26
|
Kolter JF, Hildenbrand MF, Popp S, Nauroth S, Bankmann J, Rother L, Waider J, Deckert J, Asan E, Jakob PM, Lesch KP, Schmitt-Böhrer A. Serotonin transporter genotype modulates resting state and predator stress-induced amygdala perfusion in mice in a sex-dependent manner. PLoS One 2021; 16:e0247311. [PMID: 33606835 PMCID: PMC7895400 DOI: 10.1371/journal.pone.0247311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/04/2021] [Indexed: 12/16/2022] Open
Abstract
The serotonin transporter (5-HTT) is a key molecule of serotoninergic neurotransmission and target of many anxiolytics and antidepressants. In humans, 5-HTT gene variants resulting in lower expression levels are associated with behavioral traits of anxiety. Furthermore, functional magnetic resonance imaging (fMRI) studies reported increased cerebral blood flow (CBF) during resting state (RS) and amygdala hyperreactivity. 5-HTT deficient mice as an established animal model for anxiety disorders seem to be well suited for investigating amygdala (re-)activity in an fMRI study. We investigated wildtype (5-HTT+/+), heterozygous (5-HTT+/-), and homozygous 5-HTT-knockout mice (5-HTT-/-) of both sexes in an ultra-high-field 17.6 Tesla magnetic resonance scanner. CBF was measured with continuous arterial spin labeling during RS, stimulation state (SS; with odor of rats as aversive stimulus), and post-stimulation state (PS). Subsequently, post mortem c-Fos immunohistochemistry elucidated neural activation on cellular level. The results showed that in reaction to the aversive odor CBF in total brain and amygdala of all mice significantly increased. In male 5-HTT+/+ mice amygdala RS CBF levels were found to be significantly lower than in 5-HTT+/- mice. From RS to SS 5-HTT+/+ amygdala perfusion significantly increased compared to both 5-HTT+/- and 5-HTT-/- mice. Perfusion level changes of male mice correlated with the density of c-Fos-immunoreactive cells in the amygdaloid nuclei. In female mice the perfusion was not modulated by the 5-Htt-genotype, but by estrous cycle stages. We conclude that amygdala reactivity is modulated by the 5-Htt genotype in males. In females, gonadal hormones have an impact which might have obscured genotype effects. Furthermore, our results demonstrate experimental support for the tonic model of 5-HTTLPR function.
Collapse
Affiliation(s)
- Jann F. Kolter
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
- Division of Molecular Psychiatry, Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Markus F. Hildenbrand
- Department of Magnetic Resonance and X-Ray Imaging, Fraunhofer Development Center X-Ray Technology, Wuerzburg, Germany
| | - Sandy Popp
- Division of Molecular Psychiatry, Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Stephan Nauroth
- Division of Molecular Psychiatry, Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Julian Bankmann
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Lisa Rother
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Jonas Waider
- Division of Molecular Psychiatry, Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Jürgen Deckert
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Esther Asan
- Institute of Anatomy and Cell Biology, University of Wuerzburg, Wuerzburg, Germany
| | - Peter M. Jakob
- Department of Experimental Physics 5, University of Wuerzburg, Wuerzburg, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Angelika Schmitt-Böhrer
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
- * E-mail:
| |
Collapse
|
27
|
Reduced Motivation in Perinatal Fluoxetine-Treated Mice: A Hypodopaminergic Phenotype. J Neurosci 2021; 41:2723-2732. [PMID: 33536200 DOI: 10.1523/jneurosci.2608-20.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 12/17/2022] Open
Abstract
Early life is a sensitive period, in which enhanced neural plasticity allows the developing brain to adapt to its environment. This plasticity can also be a risk factor in which maladaptive development can lead to long-lasting behavioral deficits. Here, we test how early-life exposure to the selective-serotonin-reuptake-inhibitor (SSRI), fluoxetine, affects motivation, and dopaminergic signaling in adulthood. We show for the first time that mice exposed to fluoxetine in the early postnatal period exhibit a reduction in effort-related motivation. These mice also show blunted responses to amphetamine and reduced dopaminergic activation in a sucrose reward task. Interestingly, we find that the reduction in motivation can be rescued in the adult by administering bupropion, a dopamine-norepinephrine reuptake inhibitor used as an antidepressant and a smoke cessation aid but not by fluoxetine. Taken together, our studies highlight the effects of early postnatal exposure of fluoxetine on motivation and demonstrate the involvement of the dopaminergic system in this process.SIGNIFICANCE STATEMENT The developmental period is characterized by enhanced plasticity. During this period, environmental factors have the potential to lead to enduring behavioral changes. Here, we show that exposure to the SSRI fluoxetine during a restricted period in early life leads to a reduction in adult motivation. We further show that this reduction is associated with decreased dopaminergic responsivity. Finally, we show that motivational deficits induced by early-life fluoxetine exposure can be rescued by adult administration of bupropion but not by fluoxetine.
Collapse
|
28
|
Abstract
The adult brain is the result of a multistages complex neurodevelopmental process involving genetic, molecular and microenvironmental factors as well as diverse patterns of electrical activity. In the postnatal life, immature neuronal circuits undergo an experience-dependent maturation during critical periods of plasticity, but the brain still retains plasticity during adult life. In all these stages, the neurotransmitter GABA plays a pivotal role. In this chapter, we will describe the interaction of 5-HT with GABA in regulating neurodevelopment and plasticity.
Collapse
|
29
|
Abstract
Inhibitors of Na+/Cl- dependent high affinity transporters for norepinephrine (NE), serotonin (5-HT), and/or dopamine (DA) represent frequently used drugs for treatment of psychological disorders such as depression, anxiety, obsessive-compulsive disorder, attention deficit hyperactivity disorder, and addiction. These transporters remove NE, 5-HT, and/or DA after neuronal excitation from the interstitial space close to the synapses. Thereby they terminate transmission and modulate neuronal behavioral circuits. Therapeutic failure and undesired central nervous system side effects of these drugs have been partially assigned to neurotransmitter removal by low affinity transport. Cloning and functional characterization of the polyspecific organic cation transporters OCT1 (SLC22A1), OCT2 (SLC22A2), OCT3 (SLC22A3) and the plasma membrane monoamine transporter PMAT (SLC29A4) revealed that every single transporter mediates low affinity uptake of NE, 5-HT, and DA. Whereas the organic transporters are all located in the blood brain barrier, OCT2, OCT3, and PMAT are expressed in neurons or in neurons and astrocytes within brain areas that are involved in behavioral regulation. Areas of expression include the dorsal raphe, medullary motoric nuclei, hypothalamic nuclei, and/or the nucleus accumbens. Current knowledge of the transport of monoamine neurotransmitters by the organic cation transporters, their interactions with psychotropic drugs, and their locations in the brain is reported in detail. In addition, animal experiments including behavior tests in wildtype and knockout animals are reported in which the impact of OCT2, OCT3, and/or PMAT on regulation of salt intake, depression, mood control, locomotion, and/or stress effect on addiction is suggested.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute of Anatomy and Cell Biology, University Würzburg, Würzburg, Germany.
| |
Collapse
|
30
|
Hypocretinergic interactions with the serotonergic system regulate REM sleep and cataplexy. Nat Commun 2020; 11:6034. [PMID: 33247179 PMCID: PMC7699625 DOI: 10.1038/s41467-020-19862-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 10/30/2020] [Indexed: 12/31/2022] Open
Abstract
Loss of muscle tone triggered by emotions is called cataplexy and is the pathognomonic symptom of narcolepsy, which is caused by hypocretin deficiency. Cataplexy is classically considered to be an abnormal manifestation of REM sleep and is treated by selective serotonin (5HT) reuptake inhibitors. Here we show that deleting the 5HT transporter in hypocretin knockout mice suppressed cataplexy while dramatically increasing REM sleep. Additionally, double knockout mice showed a significant deficit in the buildup of sleep need. Deleting one allele of the 5HT transporter in hypocretin knockout mice strongly increased EEG theta power during REM sleep and theta and gamma powers during wakefulness. Deleting hypocretin receptors in the dorsal raphe neurons of adult mice did not induce cataplexy but consolidated REM sleep. Our results indicate that cataplexy and REM sleep are regulated by different mechanisms and both states and sleep need are regulated by the hypocretinergic input into 5HT neurons. Narcolepsy is characterized by a sudden loss of muscle tone (cataplexy) similar to REM sleep and is caused by hypocretin deficiency. Here, the authors show that deleting the serotonin transporter gene in hypocretin knockout mice suppresses cataplexy while dramatically increasing REM sleep, indicating that these are two different states but are both regulated by hypocretinergic input to serotonergic neurons.
Collapse
|
31
|
Gallardo CM, Martin CS, Steele AD. Food Anticipatory Activity on Circadian Time Scales Is Not Dependent on Central Serotonin: Evidence From Tryptophan Hydroxylase-2 and Serotonin Transporter Knockout Mice. Front Mol Neurosci 2020; 13:534238. [PMID: 33041772 PMCID: PMC7517832 DOI: 10.3389/fnmol.2020.534238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 08/13/2020] [Indexed: 01/14/2023] Open
Abstract
A number of studies implicate biogenic amines in regulating circadian rhythms. In particular, dopamine and serotonin influence the entrainment of circadian rhythms to daily food availability. To study circadian entrainment to feeding, food availability is typically restricted to a short period within the light cycle daily. This results in a notable increase in pre-meal activity, termed "food anticipatory activity" (FAA), which typically develops within about 1 week of scheduled feeding. Several studies have implicated serotonin as a negative regulator of FAA: (1) aged rats treated with serotonin 5-HT2 and 3 receptor antagonists showed enhanced FAA, (2) mice lacking for the 2C serotonin receptor demonstrate enhanced FAA, and (3) pharmacologically increased serotonin levels suppressed FAA while decreased serotonin levels enhanced FAA in mice. We sought to confirm and extend these findings using genetic models with impairments in central serotonin production or re-uptake, but were surprised to find that both serotonin transporter (Slc6a4) and tryptophan hydroxylase-2 knockout mice demonstrated a normal behavioral response to timed, calorie restricted feeding. Our data suggest that FAA is largely independent of central serotonin and/or serotonin reuptake and that serotonin may not be a robust negative regulator of FAA.
Collapse
Affiliation(s)
- Christian M Gallardo
- Division of Biology, California Institute of Technology, Pasadena, CA, United States
| | - Camille S Martin
- Department of Biological Sciences, California State Polytechnic University Pomona, Pomona, CA, United States
| | - Andrew D Steele
- Department of Biological Sciences, California State Polytechnic University Pomona, Pomona, CA, United States
| |
Collapse
|
32
|
El-Sherbeni AA, Stocco MR, Wadji FB, Tyndale RF. Addressing the instability issue of dopamine during microdialysis: the determination of dopamine, serotonin, methamphetamine and its metabolites in rat brain. J Chromatogr A 2020; 1627:461403. [PMID: 32823108 PMCID: PMC7484461 DOI: 10.1016/j.chroma.2020.461403] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/10/2020] [Accepted: 07/12/2020] [Indexed: 12/27/2022]
Abstract
Dopamine is a catecholamine neurotransmitter that degrades rapidly in aqueous solutions; hence, its analysis following brain microdialysis is challenging. The aim of the current study was to develop and validate a new microdialysis coupled LC-MS/MS system with improved accuracy, precision, simplicity and turnaround time for dopamine, serotonin, methamphetamine, amphetamine, 4-hydroxymethamphetamine and 4-hydroxyamphetamine analysis in the brain. Dopamine degradation was studied with different stabilizing agents under different storage conditions. The modified microdialysis system was tested in vitro, and was optimized for best probe recovery, assessed by %gain. LC-MS/MS assay was developed and validated for the targeted compounds. Stabilizing agents (ascorbic acid, EDTA and acetic acid) as well as internal and cold standards were added on-line to the dialysate flow. Assay linearity range was 0.01-100 ng/mL, precision and accuracy passed criteria, and LOQ and LLOQ were 0.2 and 1.0 pg, respectively. The new microdialysis coupled LC-MS/MS system was used in Wistar rats striatum after 4 mg/kg subcutaneous methamphetamine. Methamphetamine rapidly distributed to rat striatum reaching an average ~200 ng/mL maximum, ~82.5 min post-dose. Amphetamine, followed by 4-hydroxymethamphetamine, was the most abundant metabolite. Dopamine was released following methamphetamine injection, while serotonin was not altered. In conclusion, we proposed and tested an innovative and simplified solution to improve stability, accuracy and turnover time to monitor unstable molecules, such as dopamine, by microdialysis.
Collapse
Affiliation(s)
- Ahmed A El-Sherbeni
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Marlaina R Stocco
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Fariba Baghai Wadji
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Rachel F Tyndale
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
33
|
Liu Q, Zhao C, Chen M, Liu Y, Zhao Z, Wu F, Li Z, Weiss PS, Andrews AM, Zhou C. Flexible Multiplexed In 2O 3 Nanoribbon Aptamer-Field-Effect Transistors for Biosensing. iScience 2020; 23:101469. [PMID: 33083757 PMCID: PMC7509003 DOI: 10.1016/j.isci.2020.101469] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/03/2020] [Accepted: 08/13/2020] [Indexed: 11/05/2022] Open
Abstract
Flexible sensors are essential for advancing implantable and wearable bioelectronics toward monitoring chemical signals within and on the body. Developing biosensors for monitoring multiple neurotransmitters in real time represents a key in vivo application that will increase understanding of information encoded in brain neurochemical fluxes. Here, arrays of devices having multiple In2O3 nanoribbon field-effect transistors (FETs) were fabricated on 1.4-μm-thick polyethylene terephthalate (PET) substrates using shadow mask patterning techniques. Thin PET-FET devices withstood crumpling and bending such that stable transistor performance with high mobility was maintained over >100 bending cycles. Real-time detection of the small-molecule neurotransmitters serotonin and dopamine was achieved by immobilizing recently identified high-affinity nucleic-acid aptamers on individual In2O3 nanoribbon devices. Limits of detection were 10 fM for serotonin and dopamine with detection ranges spanning eight orders of magnitude. Simultaneous sensing of temperature, pH, serotonin, and dopamine enabled integration of physiological and neurochemical data from individual bioelectronic devices. We fabricated flexible In2O3 nanoribbon transistors using cleanroom-free processes Flexible In2O3 transistors withstood crumpling and bending with stable performance Flexible aptamer biosensors detect neurotransmitters in real time Multiplexed sensors monitor temperature, pH, serotonin, and dopamine simultaneously
Collapse
Affiliation(s)
- Qingzhou Liu
- Ming Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089, USA.,Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Chuanzhen Zhao
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mingrui Chen
- Ming Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Yihang Liu
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Zhiyuan Zhao
- Ming Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Fanqi Wu
- Ming Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Zhen Li
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Paul S Weiss
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Departments of Bioengineering and Materials Science and Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Anne M Andrews
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chongwu Zhou
- Ming Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089, USA.,Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
34
|
Forero A, Ku HP, Malpartida AB, Wäldchen S, Alhama-Riba J, Kulka C, Aboagye B, Norton WHJ, Young AMJ, Ding YQ, Blum R, Sauer M, Rivero O, Lesch KP. Serotonin (5-HT) neuron-specific inactivation of Cadherin-13 impacts 5-HT system formation and cognitive function. Neuropharmacology 2020; 168:108018. [PMID: 32113967 DOI: 10.1016/j.neuropharm.2020.108018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 02/15/2020] [Accepted: 02/23/2020] [Indexed: 02/06/2023]
Abstract
Genome-wide screening approaches identified the cell adhesion molecule Cadherin-13 (CDH13) as a risk factor for neurodevelopmental disorders, nevertheless the contribution of CDH13 to the disease mechanism remains obscure. CDH13 is involved in neurite outgrowth and axon guidance during early brain development and we previously provided evidence that constitutive CDH13 deficiency influences the formation of the raphe serotonin (5-HT) system by modifying neuron-radial glia interaction. Here, we dissect the specific impact of CDH13 on 5-HT system development and function using a 5-HT neuron-specific Cdh13 knockout mouse model (conditional Cdh13 knockout, Cdh13 cKO). Our results show that exclusive inactivation of CDH13 in 5-HT neurons selectively increases 5-HT neuron density in the embryonic dorsal raphe, with persistence into adulthood, and serotonergic innervation of the developing prefrontal cortex. At the behavioral level, adult Cdh13 cKO mice display delayed acquisition of several learning tasks and a subtle impulsive-like phenotype, with decreased latency in a sociability paradigm alongside with deficits in visuospatial memory. Anxiety-related traits were not observed in Cdh13 cKO mice. Our findings further support the critical role of CDH13 in the development of dorsal raphe 5-HT circuitries, a mechanism that may underlie specific clinical features observed in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Andrea Forero
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany.
| | - Hsing-Ping Ku
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Ana Belén Malpartida
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Sina Wäldchen
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Judit Alhama-Riba
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Christina Kulka
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Benjamin Aboagye
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - William H J Norton
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | - Andrew M J Young
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | - Yu-Qiang Ding
- Institute of Brain Sciences, Fudan University, Shanghai, 200031, China
| | - Robert Blum
- Institute of Clinical Neurobiology, University of Würzburg, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Olga Rivero
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
35
|
Wilson C, Li S, Hannan AJ, Renoir T. Antidepressant-like effects of ketamine in a mouse model of serotonergic dysfunction. Neuropharmacology 2020; 168:107998. [PMID: 32061666 DOI: 10.1016/j.neuropharm.2020.107998] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 02/05/2020] [Accepted: 02/09/2020] [Indexed: 12/28/2022]
Abstract
Traditional monoaminergic treatments of depression frequently exhibit suboptimal tolerability and effectiveness. The 'short' (s) allele variant of 5-HTTLPR is known to compromise transcriptional efficacy of the serotonin transporter (5-HTT) and can reduce treatment response to traditional antidepressants (e.g. selective serotonin reuptake inhibitors or SSRIs). This study sought to establish the 5-HTT knock-out (KO) line as a mouse model of SSRI-resistant depression and assess its response to a novel glutamatergic antidepressant, ketamine, a non-competitive N-methyl-d-aspartate receptor (NMDAR) antagonist. Following acute antidepressant treatment, 5-HTT KO mice and wild-type (WT) controls were subjected to the forced-swim test (FST), one of the most widely used techniques to detect acute antidepressant response. As hypothesised, when assessed 30 min after administration in the FST, the SSRI sertraline (20 mg/kg, i.p.) produced antidepressant-like effects in WT control but not in 5-HTT KO mice. In contrast, ketamine (20 mg/kg, i.p.) induced antidepressant-like effects in both genotypes. 5-HTT KO mice also exhibited a reduced locomotor response to both MK-801 (another NMDAR antagonist) and ketamine, and reduced GluN2A protein levels in the hippocampus, suggesting glutamatergic dysfunction in this model. These results highlight the utility of 5-HTT KO mice as a relevant model of SSRI-resistant depression and demonstrate that ketamine can produce acute antidepressant-like effects in conditions of 5-HTT deficiency. These findings extend existing literature that indicates ketamine is effective in ameliorating symptoms of treatment-resistant depression and may have implications for understanding the cellular and molecular mechanisms underlying the antidepressant effects of ketamine. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.
Collapse
Affiliation(s)
- Carey Wilson
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia; Melbourne School of Psychological Science, University of Melbourne, Parkville, Australia
| | - Shanshan Li
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia; Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia; Facssulty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia.
| |
Collapse
|
36
|
Annamalai B, Ragu Varman D, Horton RE, Daws LC, Jayanthi LD, Ramamoorthy S. Histamine Receptors Regulate the Activity, Surface Expression, and Phosphorylation of Serotonin Transporters. ACS Chem Neurosci 2020; 11:466-476. [PMID: 31916747 DOI: 10.1021/acschemneuro.9b00664] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Reuptake and clearance of released serotonin (5-HT) are critical in serotonergic neurotransmission. Serotonin transporter (SERT) is mainly responsible for clearing the extracellular 5-HT. Controlled trafficking, phosphorylation, and protein stability have been attributed to robust SERT activity. H3 histamine receptors (H3Rs) act in conjunction and regulate 5-HT release. H3Rs are expressed in the nervous system and located at the serotonergic terminals, where they act as heteroreceptors. Although histaminergic and serotonergic neurotransmissions are thought to be two separate events, whether H3Rs influence SERT in the CNS to control 5-HT reuptake has never been addressed. With a priori knowledge gained from our studies, we explored the possibility of using rat hippocampal synaptosomal preparations. We found that treatment with H3R/H4R-agonists immepip and (R)-(-)-α-methyl-histamine indeed resulted in a time- and concentration-dependent decrease in 5-HT transport. On the other hand, treatment with H3R/H4R-inverse agonist thioperamide caused a moderate increase in 5-HT uptake while blocking the inhibitory effect of H3R/H4R agonists. When investigated further, immepip treatment reduced the level of SERT on the plasma membrane and its phosphorylation. Likewise, CaMKII inhibitor KN93 or calcineurin inhibitor cyclosporine A also inhibited SERT function; however, an additive effect with immepip was not seen. High-speed in vivo chronoamperometry demonstrated that immepip delayed 5-HT clearance while thioperamide accelerated 5-HT clearance from the extracellular space. Immepip selectively inhibited SERT activity in the hippocampus and cortex but not in the striatum, midbrain, and brain stem. Thus, we report here a novel mechanism of regulating SERT activity by H3R-mediated CaMKII/calcineurin pathway in a brain-region-specific manner and perhaps synaptic 5-HT in the CNS that controls 5-HT clearance.
Collapse
Affiliation(s)
- Balasubramaniam Annamalai
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Durairaj Ragu Varman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Rebecca E. Horton
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, United States
| | - Lynette C. Daws
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, United States
| | - Lankupalle D. Jayanthi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Sammanda Ramamoorthy
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
37
|
Neukam PT, Deza-Araujo YI, Marxen M, Pooseh S, Rietschel M, Schwarzenbolz U, Smolka MN. No evidence for the involvement of serotonin or the 5-HTTLPR genotype in intertemporal choice in a larger community sample. J Psychopharmacol 2019; 33:1377-1387. [PMID: 31547761 DOI: 10.1177/0269881119874417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Serotonin has been implicated in impulsive behaviours such as temporal discounting. While animal studies and theoretical approaches suggest that reduced tonic serotonin levels increase temporal discounting rates and vice versa, evidence from human studies is scarce and inconclusive. Furthermore, an important modulator of serotonin signalling, a genetic variation in the promoter region of the serotonin transporter gene (5-HTTLPR), has not been investigated for temporal discounting so far. OBJECTIVE First, the purpose of this study was to test for a significant association between 5-HTTLPR and temporal discounting. Second, we wished to investigate the effect of high/low tonic serotonin levels on intertemporal choice and blood oxygen-level-dependent response, controlling for 5-HTTLPR. METHODS We tested the association of 5-HTTLPR with temporal discounting rates using an intertemporal choice task in 611 individuals. We then manipulated tonic serotonin levels with acute tryptophan interventions (depletion, loading, balanced) in a subsample of 45 short (S)-allele and 45 long (L)/L-allele carriers in a randomised double-blind crossover design using functional magnetic resonance imaging and an intertemporal choice task. RESULTS Overall, we did not find any effect of serotonin and 5-HTTLPR on temporal discounting rates or the brain networks associated with valuation and cognitive control. CONCLUSION Our findings indicate that serotonin may not be directly involved in choices including delays on longer timescales such as days, weeks or months. We speculate that serotonin plays a stronger role in dynamic intertemporal choice tasks where the delays are on a timescale of seconds and hence are therefore directly experienced during the experiment.
Collapse
Affiliation(s)
- Philipp T Neukam
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Yacila I Deza-Araujo
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Michael Marxen
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Shakoor Pooseh
- Freiburg Center for Data Analysis and Modeling, Freiburg, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Uwe Schwarzenbolz
- Institute of Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Michael N Smolka
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
38
|
de Assis Brasil ES, Guerino Furini CR, da Silva Rodrigues F, Nachtigall EG, Kielbovicz Behling JA, Saenger BF, Farias CP, de Carvalho Myskiw J, Izquierdo I. The blockade of the serotoninergic receptors 5-HT5A, 5-HT6 and 5-HT7 in the basolateral amygdala, but not in the hippocampus facilitate the extinction of fear memory. Behav Brain Res 2019; 372:112055. [DOI: 10.1016/j.bbr.2019.112055] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/27/2019] [Accepted: 06/20/2019] [Indexed: 01/20/2023]
|
39
|
Gopaldas M, Zanderigo F, Zhan S, Ogden RT, Miller JM, Rubin-Falcone H, Cooper TB, Oquendo MA, Sullivan G, Mann JJ, Sublette ME. Brain serotonin transporter binding, plasma arachidonic acid and depression severity: A positron emission tomography study of major depression. J Affect Disord 2019; 257:495-503. [PMID: 31319341 PMCID: PMC6886679 DOI: 10.1016/j.jad.2019.07.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 06/11/2019] [Accepted: 07/04/2019] [Indexed: 11/17/2022]
Abstract
BACKGROUND Serotonin transporter (5-HTT) binding and polyunsaturated fatty acids (PUFAs) are implicated in major depressive disorder (MDD). Links between the two systems in animal models have not been investigated in humans. METHODS Using positron emission tomography (PET) and [11C]DASB, we studied relationships between 5-HTT binding potential and plasma levels of PUFAs docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and arachidonic acid (AA) in medication-free MDD patients (n = 21). PUFAs were quantified using transesterification and gas chromatography. Binding potential BPP, and alternative outcome measures BPF and BPND, were determined for [11C]DASB in six a priori brain regions of interest (ROIs) using likelihood estimation in graphical analysis (LEGA) to calculate radioligand total distribution volume (VT), and a validated hybrid deconvolution approach (HYDECA) that estimates radioligand non-displaceable distribution volume (VND) without a reference region. Linear mixed models used PUFA levels as predictors and binding potential measures as outcomes across the specified ROIs; age and sex as fixed effects; and subject as random effect to account for across-region binding correlations. As nonlinear relationships were observed, a quadratic term was added to final models. RESULTS AA predicted both 5-HTT BPP and depression severity nonlinearly, described by an inverted U-shaped curve. 5-HTT binding potential mediated the relationship between AA and depression severity. LIMITATIONS Given the small sample and multiple comparisons, results require replication. CONCLUSIONS Our findings suggest that AA status may impact depression pathophysiology through effects on serotonin transport. Future studies should examine whether these relationships explain therapeutic effects of PUFAs in the treatment of MDD.
Collapse
Affiliation(s)
- Manesh Gopaldas
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA,Department of Psychiatry & Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Francesca Zanderigo
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - Serena Zhan
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY
| | - R. Todd Ogden
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA,Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY
| | - Jeffrey M. Miller
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - Harry Rubin-Falcone
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - Thomas B. Cooper
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA,Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Maria A. Oquendo
- Psychiatry Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - J. John Mann
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA,Department of Radiology, Columbia University, New York, NY, USA
| | - M. Elizabeth Sublette
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA,To whom correspondence should be addressed: New York State Psychiatric Institute, 1051 Riverside Drive, Unit 42, New York, NY 10032, Tel: 646 774-7514, Fax: 646 774-7589,
| |
Collapse
|
40
|
Rogers J, Chen F, Stanic D, Farzana F, Li S, Zeleznikow-Johnston AM, Nithianantharajah J, Churilov L, Adlard PA, Lanfumey L, Hannan AJ, Renoir T. Paradoxical effects of exercise on hippocampal plasticity and cognition in mice with a heterozygous null mutation in the serotonin transporter gene. Br J Pharmacol 2019; 176:3279-3296. [PMID: 31167040 DOI: 10.1111/bph.14760] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 05/08/2019] [Accepted: 05/14/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Exercise is known to improve cognitive function, but the exact synaptic and cellular mechanisms remain unclear. We investigated the potential role of the serotonin (5-HT) transporter (SERT) in mediating these effects. EXPERIMENTAL APPROACH Hippocampal long-term potentiation (LTP) and neurogenesis were measured in standard-housed and exercising (wheel running) wild-type (WT) and SERT heterozygous (HET) mice. We also assessed hippocampal-dependent cognition using the Morris water maze (MWM) and a spatial pattern separation touchscreen task. KEY RESULTS SERT HET mice had impaired hippocampal LTP regardless of the housing conditions. Exercise increased hippocampal neurogenesis in WT mice. However, this was not observed in SERT HET animals, even though both genotypes used the running wheels to a similar extent. We also found that standard-housed SERT HET mice displayed altered cognitive flexibility than WT littermate controls in the MWM reversal learning task. However, SERT HET mice no longer exhibited this phenotype after exercise. Cognitive changes, specific to SERT HET mice in the exercise condition, were also revealed on the touchscreen spatial pattern separation task, especially when the cognitive pattern separation load was at its highest. CONCLUSIONS AND IMPLICATIONS Our study is the first evidence of reduced hippocampal LTP in SERT HET mice. We also show that functional SERT is required for exercise-induced increase in adult neurogenesis. Paradoxically, exercise had a negative impact on hippocampal-dependent cognitive tasks, especially in SERT HET mice. Taken together, our results suggest unique complex interactions between exercise and altered 5-HT homeostasis.
Collapse
Affiliation(s)
- Jake Rogers
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Feng Chen
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Davor Stanic
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Farheen Farzana
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Shanshan Li
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Ariel M Zeleznikow-Johnston
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Jess Nithianantharajah
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Leonid Churilov
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC, Australia.,School of Mathematical and Geospatial Sciences, RMIT University, Melbourne, VIC, Australia
| | - Paul A Adlard
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Laurence Lanfumey
- UMR S894, Université Paris Descartes, Paris, France.,Centre de Psychiatrie et Neurosciences, Inserm UMR 894, Paris, France
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
41
|
Geng H, Peng D, Huang Y, Tang D, Gao J, Zhang Y, Zhang X. Changes in sexual performance and biochemical characterisation of functional neural regions: A study in serotonin transporter knockout male rats. Andrologia 2019; 51:e13291. [PMID: 31037750 DOI: 10.1111/and.13291] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/17/2019] [Accepted: 03/22/2019] [Indexed: 11/29/2022] Open
Affiliation(s)
- Hao Geng
- Department of Urology The First Affiliated Hospital of Anhui Medical University Hefei China
| | - Dangwei Peng
- Department of Urology The First Affiliated Hospital of Anhui Medical University Hefei China
| | - Yuanyuan Huang
- Department of Urology The Fourth Affiliated Hospital of Anhui Medical University Hefei China
| | - Dongdong Tang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology The First Affiliated Hospital of Anhui Medical University Hefei China
| | - Jingjing Gao
- Department of Urology The First Affiliated Hospital of Anhui Medical University Hefei China
| | - Yao Zhang
- Department of Urology The First Affiliated Hospital of Anhui Medical University Hefei China
| | - Xiansheng Zhang
- Department of Urology The First Affiliated Hospital of Anhui Medical University Hefei China
| |
Collapse
|
42
|
Garbarino VR, Gilman TL, Daws LC, Gould GG. Extreme enhancement or depletion of serotonin transporter function and serotonin availability in autism spectrum disorder. Pharmacol Res 2019; 140:85-99. [PMID: 30009933 PMCID: PMC6345621 DOI: 10.1016/j.phrs.2018.07.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/22/2018] [Accepted: 07/12/2018] [Indexed: 12/15/2022]
Abstract
A variety of human and animal studies support the hypothesis that serotonin (5-hydroxytryptamine or 5-HT) system dysfunction is a contributing factor to the development of autism in some patients. However, many questions remain about how developmental manipulation of various components that influence 5-HT signaling (5-HT synthesis, transport, metabolism) persistently impair social behaviors. This review will summarize key aspects of central 5-HT function important for normal brain development, and review evidence implicating perinatal disruptions in 5-HT signaling in the pathophysiology of autism spectrum disorder. We discuss the importance, and relative dearth, of studies that explore the possible correlation to autism in the interactions between important intrinsic and extrinsic factors that may disrupt 5-HT homeostasis during development. In particular, we focus on exposure to 5-HT transport altering mechanisms such as selective serotonin-reuptake inhibitors or genetic polymorphisms in primary or auxiliary transporters of 5-HT, and how they relate to neurological stores of serotonin and its precursors. A deeper understanding of the many mechanisms by which 5-HT signaling can be disrupted, alone and in concert, may contribute to an improved understanding of the etiologies and heterogeneous nature of this disorder. We postulate that extreme bidirectional perturbations of these factors during development likely compound or synergize to facilitate enduring neurochemical changes resulting in insufficient or excessive 5-HT signaling, that could underlie the persistent behavioral characteristics of autism spectrum disorder.
Collapse
Affiliation(s)
- Valentina R Garbarino
- Department of Cellular and Integrative Physiology, United States; The Sam and Ann Barshop Institute for Longevity and Aging Studies, United States.
| | - T Lee Gilman
- Department of Cellular and Integrative Physiology, United States; Addiction Research, Treatment & Training Center of Excellence, United States.
| | - Lynette C Daws
- Department of Cellular and Integrative Physiology, United States; Addiction Research, Treatment & Training Center of Excellence, United States; Department of Pharmacology, United States.
| | - Georgianna G Gould
- Department of Cellular and Integrative Physiology, United States; Center for Biomedical Neuroscience, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
43
|
Pinheiro IL, da Silva AI, Reginato A, da Silva Filho RC, Galindo LCM, Matos RJB, de Souza Ferraz JC, Toscano Meneses da Silva Castro AE, Milanski Ferreira M, Manhães de Castro R, de Souza SL. Neonatal fluoxetine exposure modulates serotonergic neurotransmission and disturb inhibitory action of serotonin on food intake. Behav Brain Res 2019; 357-358:65-70. [DOI: 10.1016/j.bbr.2017.07.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/12/2017] [Accepted: 07/25/2017] [Indexed: 12/15/2022]
|
44
|
Tanaka M, Sato A, Kasai S, Hagino Y, Kotajima-Murakami H, Kashii H, Takamatsu Y, Nishito Y, Inagaki M, Mizuguchi M, Hall FS, Uhl GR, Murphy D, Sora I, Ikeda K. Brain hyperserotonemia causes autism-relevant social deficits in mice. Mol Autism 2018; 9:60. [PMID: 30498565 PMCID: PMC6258166 DOI: 10.1186/s13229-018-0243-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 11/08/2018] [Indexed: 12/19/2022] Open
Abstract
Background Hyperserotonemia in the brain is suspected to be an endophenotype of autism spectrum disorder (ASD). Reducing serotonin levels in the brain through modulation of serotonin transporter function may improve ASD symptoms. Methods We analyzed behavior and gene expression to unveil the causal mechanism of ASD-relevant social deficits using serotonin transporter (Sert) knockout mice. Results Social deficits were observed in both heterozygous knockout mice (HZ) and homozygous knockout mice (KO), but increases in general anxiety were only observed in KO mice. Two weeks of dietary restriction of the serotonin precursor tryptophan ameliorated both brain hyperserotonemia and ASD-relevant social deficits in Sert HZ and KO mice. The expression of rather distinct sets of genes was altered in Sert HZ and KO mice, and a substantial portion of these genes was also affected by tryptophan depletion. Tryptophan depletion in Sert HZ and KO mice was associated with alterations in the expression of genes involved in signal transduction pathways initiated by changes in extracellular serotonin or melatonin, a derivative of serotonin. Only expression of the AU015836 gene was altered in both Sert HZ and KO mice. AU015836 expression and ASD-relevant social deficits normalized after dietary tryptophan restriction. Conclusions These findings reveal a Sert gene dose-dependent effect on brain hyperserotonemia and related behavioral sequelae in ASD and a possible therapeutic target to normalize brain hyperserotonemia and ASD-relevant social deficits. Electronic supplementary material The online version of this article (10.1186/s13229-018-0243-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Miho Tanaka
- 1Department of Psychiatry and Behavioral Sciences, Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan.,2Molecular and Cellular Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.,3Department of Developmental Disorders, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Atsushi Sato
- 1Department of Psychiatry and Behavioral Sciences, Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan.,4Department of Pediatrics, The University of Tokyo Hospital, Tokyo, Japan
| | - Shinya Kasai
- 1Department of Psychiatry and Behavioral Sciences, Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan
| | - Yoko Hagino
- 1Department of Psychiatry and Behavioral Sciences, Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan
| | - Hiroko Kotajima-Murakami
- 1Department of Psychiatry and Behavioral Sciences, Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan
| | - Hirofumi Kashii
- 1Department of Psychiatry and Behavioral Sciences, Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan
| | - Yukio Takamatsu
- 1Department of Psychiatry and Behavioral Sciences, Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan
| | - Yasumasa Nishito
- 5Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masumi Inagaki
- 3Department of Developmental Disorders, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Masashi Mizuguchi
- 6Department of Developmental Medical Sciences, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - F Scott Hall
- 7Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH USA
| | - George R Uhl
- 8Branch of Molecular Neurobiology, National Institute on Drug Abuse, Baltimore, MD USA.,9Research Service, New Mexico VA Health Care System, Albuquerque, NM USA
| | - Dennis Murphy
- 10Laboratory of Clinical Science, National Institutes of Health, Bethesda, MD USA
| | - Ichiro Sora
- 11Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazutaka Ikeda
- 1Department of Psychiatry and Behavioral Sciences, Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan.,2Molecular and Cellular Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
45
|
Korte-Bouws GAH, van Heesch F, Westphal KGC, Ankersmit LMJ, van Oosten EM, Güntürkün O, Korte SM. Bacterial Lipopolysaccharide Increases Serotonin Metabolism in Both Medial Prefrontal Cortex and Nucleus Accumbens in Male Wild Type Rats, but Not in Serotonin Transporter Knockout Rats. Pharmaceuticals (Basel) 2018; 11:ph11030066. [PMID: 29976854 PMCID: PMC6160917 DOI: 10.3390/ph11030066] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 06/29/2018] [Accepted: 07/02/2018] [Indexed: 11/19/2022] Open
Abstract
It is well known that bacterial lipopolysaccharides (LPS) both increases proinflammatory cytokines and produces sickness behavior, including fatigue and anhedonia (i.e., the inability to experience pleasure). Previously, we have shown that intraperitoneally (i.p.) administered LPS increased extracellular monoamine metabolite levels in the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC), which was completely, or at least partly, prevented by pretreatment with a triple reuptake inhibitor that also blocks the serotonin (5-HT) transporter (SERT). This suggests indirectly, that LPS may enhance SERT transporter activity, and consequently, increase removal of 5-HT from the synaptic cleft, and increase metabolism of 5-HT. In the present study, we focus more specifically on the role of SERT in this increased metabolism by using rats, that differ in SERT expression. Therefore, the effects of an intraperitoneal LPS injection on extracellular concentrations of 5-HT and its metabolite 5-hydroxyindoleacetic acid (5-HIAA) were investigated by in vivo microdialysis in the NAc and mPFC of wild type (SERT+/+), heterozygous (SERT+/−) and knockout (SERT−/−) rats. Here, we show that LPS-induced 5-HIAA formation in male rats, is significantly increased in SERT+/+ rats in both the NAc and mPFC, whereas this increase is partly or totally abolished in SERT+/− and SERT−/− rats, respectively. Thus, the present study supports the hypothesis that systemic LPS in male rats increases SERT function and consequently enhances 5-HT uptake and metabolism in both the NAc and mPFC.
Collapse
Affiliation(s)
- Gerdien A H Korte-Bouws
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Faculty of Science, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.
| | - Floor van Heesch
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Faculty of Science, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.
| | - Koen G C Westphal
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Faculty of Science, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.
| | - Lisa M J Ankersmit
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Faculty of Science, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.
| | - Edwin M van Oosten
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Faculty of Science, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.
| | - Onur Güntürkün
- Department of Biopsychology, Faculty of Psychology, Ruhr-Universität Bochum, Universitätsstraße 150, D-44780 Bochum, Germany.
| | - S Mechiel Korte
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Faculty of Science, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.
- Department of Biopsychology, Faculty of Psychology, Ruhr-Universität Bochum, Universitätsstraße 150, D-44780 Bochum, Germany.
| |
Collapse
|
46
|
Neukam PT, Kroemer NB, Deza Araujo YI, Hellrung L, Pooseh S, Rietschel M, Witt SH, Schwarzenbolz U, Henle T, Smolka MN. Risk-seeking for losses is associated with 5-HTTLPR, but not with transient changes in 5-HT levels. Psychopharmacology (Berl) 2018; 235:2151-2165. [PMID: 29730700 DOI: 10.1007/s00213-018-4913-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/17/2018] [Indexed: 10/17/2022]
Abstract
RATIONALE Serotonin (5-HT) plays a key role in different aspects of value-based decision-making. A recent framework proposed that tonic 5-HT (together with dopamine, DA) codes future average reward expectations, providing a baseline against which possible choice outcomes are compared to guide decision-making. OBJECTIVES To test whether high 5-HT levels decrease loss aversion, risk-seeking for gains, and risk-seeking for losses. METHODS In a first session, 611 participants were genotyped for 5-HTTLPR and performed a mixed gambles (MGA) task and two probability discounting tasks for gains and losses, respectively (PDG/PDL). Afterwards, a subsample of 105 participants (44 with S/S, 6 with S/L, 55 with L/L genotype) completed the pharmacological study using a crossover design with tryptophan depletion (ATD), loading (ATL), and balanced (BAL) conditions. The same decision constructs were assessed. RESULTS We found increased risk-seeking for losses in S/S compared to L/L individuals at the first visit (p = 0.002). Neither tryptophan depletion nor loading affected decision-making, nor did we observe an interaction between intervention and 5-HTTLPR genotype. CONCLUSION Our data do not support the idea that transient changes of tonic 5-HT affect value-based decision-making. We provide evidence for an association of 5-HTTLPR with risk-seeking for losses, independent of acute 5-HT levels. This indicates that the association of 5-HTTLPR and risk-seeking for losses is mediated via other mechanisms, possibly by differences in the structural development of neural circuits of the 5-HT system during early life phases.
Collapse
Affiliation(s)
- Philipp T Neukam
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Nils B Kroemer
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany.,Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Yacila I Deza Araujo
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Lydia Hellrung
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany.,Department of Economics, University of Zürich, Zürich, Switzerland
| | - Shakoor Pooseh
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Uwe Schwarzenbolz
- Institute of Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Thomas Henle
- Institute of Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Michael N Smolka
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
47
|
Brain serotonin synthesis capacity in obsessive-compulsive disorder: effects of cognitive behavioral therapy and sertraline. Transl Psychiatry 2018; 8:82. [PMID: 29666372 PMCID: PMC5904107 DOI: 10.1038/s41398-018-0128-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 01/06/2018] [Accepted: 01/31/2018] [Indexed: 02/01/2023] Open
Abstract
Cognitive behavioral therapy (CBT) and selective serotonin reuptake inhibitors (SSRIs) are both effective treatments for some patients with obsessive-compulsive disorder (OCD), yet little is known about the neurochemical changes related to these treatment modalities. Here, we used positron emission tomography and the α-[11C]methyl-L-tryptophan tracer to examine the changes in brain regional serotonin synthesis capacity in OCD patients following treatment with CBT or SSRI treatment. Sixteen medication-free OCD patients were randomly assigned to 12 weeks of either CBT or sertraline treatment. Pre-to-post treatment changes in the α-[11C]methyl-L-tryptophan brain trapping constant, K* (ml/g/min), were assessed as a function of symptom response, and correlations with symptom improvement were examined. Responders/partial responders to treatment did not show significant changes in relative regional tracer uptake; rather, in responders/partial responders, 12 weeks of treatment led to serotonin synthesis capacity increases that were brain-wide. Irrespective of treatment modality, baseline serotonin synthesis capacity in the raphe nuclei correlated positively with clinical improvement. These observations suggest that, for some patients, successful remediation of OCD symptoms might be associated with greater serotonergic tone.
Collapse
|
48
|
Kulikov AV, Gainetdinov RR, Ponimaskin E, Kalueff AV, Naumenko VS, Popova NK. Interplay between the key proteins of serotonin system in SSRI antidepressants efficacy. Expert Opin Ther Targets 2018. [DOI: 10.1080/14728222.2018.1452912] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Alexander V. Kulikov
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Allan V. Kalueff
- School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
- Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia
- Ural Federal University, Ekaterinburg 620002, Russia
- Research Institute of Physiology and Basic Medicine, Novosibirsk 630117, Russia
- Russian Research Center for Radiology and Surgical Technologies, Pesochny 197758, Russia
| | - Vladimir S. Naumenko
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Nina K. Popova
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
49
|
Ishizu N, Yui D, Hebisawa A, Aizawa H, Cui W, Fujita Y, Hashimoto K, Ajioka I, Mizusawa H, Yokota T, Watase K. Impaired striatal dopamine release in homozygous Vps35 D620N knock-in mice. Hum Mol Genet 2018; 25:4507-4517. [PMID: 28173004 DOI: 10.1093/hmg/ddw279] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/11/2016] [Accepted: 08/15/2016] [Indexed: 12/22/2022] Open
Abstract
Point mutations in the vacuolar protein sorting 35 gene (VPS35) have been associated with an autosomal dominant form of late-onset Parkinson disease (PARK17), but there has been considerable debate over whether it is caused by a loss- or gain-of-function mechanism and over the intracellular target site of neurotoxicity. To investigate the pathogenesis of PARK17 in vivo, we generated Vps35 D620N knock-in (KI) mice, expressing the homologous mutant protein with endogenous patterns of expression, simultaneously with Vps35 deletion 1 (Del1) mice, which carry 1bp deletion in the exon15 of Vps35, by CRISPR/Cas9-mediated genome engineering. Neither homozygous nor heterozygous Vps35 D620N KI mice suffered from premature death or developed clear neurodegeneration up to 70 weeks of age. Vps35 Del1 allele appeared to be a null or at least severely hypomorphic allele and homozygous Vps35 Del1 showed early embryonic lethality. Heterozygous crossings between Del1 and D620N knock-in mice revealed that the D620N/Del1 compound heterozygous mice, but not heterozygous Del1 mice, suffered from survival disadvantage. In vivo microdialysis showed that DA release evoked by 120 mM potassium chloride was significantly reduced in the caudate putamen of adult homozygous Vps35 D620N KI mice. Taken together, these results suggest that Vps35 D620N allele is a partial-loss-of-function allele and that such a genetic predisposition and age-related alterations in the nigrostriatal dopamine system cooperatively influence the pathogenesis of PARK17.
Collapse
Affiliation(s)
- Nobutaka Ishizu
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University Graduate School of Medical and Dental Sciences, Tokyo, Japan.,Department of Neurology, Tokyo National Hospital, Tokyo Japan
| | - Daishi Yui
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Akira Hebisawa
- Department of Clinical research, Tokyo National Hospital, Tokyo Japan
| | - Hidenori Aizawa
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan,Department of Neurobiology, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Wanpeng Cui
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan,Department of Neurobiology, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuko Fujita
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Itsuki Ajioka
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Takanori Yokota
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Kei Watase
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
50
|
Cao H, Harneit A, Walter H, Erk S, Braun U, Moessnang C, Geiger LS, Zang Z, Mohnke S, Heinz A, Romanczuk-Seiferth N, Mühleisen T, Mattheisen M, Witt SH, Cichon S, Nöthen MM, Rietschel M, Meyer-Lindenberg A, Tost H. The 5-HTTLPR Polymorphism Affects Network-Based Functional Connectivity in the Visual-Limbic System in Healthy Adults. Neuropsychopharmacology 2018; 43:406-414. [PMID: 28589968 PMCID: PMC5729553 DOI: 10.1038/npp.2017.121] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 04/29/2017] [Accepted: 05/16/2017] [Indexed: 01/15/2023]
Abstract
The serotonin transporter-linked polymorphic region 5-HTTLPR is a key genetic regulator of 5-HTT expression in the human brain where the short allele S has been implicated in emotion dysregulation. However, the neural mechanism underlying the association between this variant and emotion processing is still unclear. Earlier studies suggested an effect of 5-HTTLPR on amygdala activation during emotional face processing. However, this association has been questioned in recent studies employing larger sample sizes and meta-analyses. Here, we examined a sample of 223 healthy subjects with a well-established fMRI emotional face processing task to (1) re-evaluate the association between 5-HTTLPR and amygdala activation, (2) explore potential network-based functional connectivity phenotypes for associations with 5-HTTLPR, and (3) probe the reliability, behavioral significance and potential structural confounds of the identified network phenotype. Our results revealed no significant effect of 5-HTTLPR on amygdala activation (P>0.79). However, the number of S alleles was significantly correlated with functional connectivity of a visual-limbic subnetwork (PFWE=0.03). The subnetwork cluster included brain regions that are pivotal to emotion regulation such as the hippocampus, orbitofrontal cortex, anterior cingulate gyrus, fusiform gyrus, and subcortex. Notably, individuals with lower subnetwork connectivity had significantly higher emotion suppression scores (P=0.01). Further, the connectivity metrics were test-retest reliable and independent from subnetwork gray matter volume and white matter anisotropy. Our data provide evidence for a functional network-based phenotype linking genetic variation in 5-HTTLPR to emotion regulation, and suggest that further critical evaluations of the association between 5-HTTLPR and amygdala activation are warranted.
Collapse
Affiliation(s)
- Hengyi Cao
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Anais Harneit
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy, Universitätsmedizin Charité, Berlin, Germany
| | - Susanne Erk
- Department of Psychiatry and Psychotherapy, Universitätsmedizin Charité, Berlin, Germany
| | - Urs Braun
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Carolin Moessnang
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Lena S Geiger
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Zhenxiang Zang
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Sebastian Mohnke
- Department of Psychiatry and Psychotherapy, Universitätsmedizin Charité, Berlin, Germany
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy, Universitätsmedizin Charité, Berlin, Germany
| | | | - Thomas Mühleisen
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Manuel Mattheisen
- Department of Genomic Mathematics, University of Bonn, Bonn, Germany,Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Sven Cichon
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany,Department of Biomedicine, University of Basel, Basel, Switzerland,Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany,Division of Medical Genetics, University of Basel, Basel, Switzerland
| | - Markus M Nöthen
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany,Institute of Human Genetics, University of Bonn School of Medicine & University Hospital of Bonn, Bonn, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Andreas Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Heike Tost
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany,Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Square J5, Mannheim 68159, Germany, Tel: +49 621 1703 6510, E-mail:
| |
Collapse
|