1
|
Özçete ÖD, Banerjee A, Kaeser PS. Mechanisms of neuromodulatory volume transmission. Mol Psychiatry 2024; 29:3680-3693. [PMID: 38789677 PMCID: PMC11540752 DOI: 10.1038/s41380-024-02608-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024]
Abstract
A wealth of neuromodulatory transmitters regulate synaptic circuits in the brain. Their mode of signaling, often called volume transmission, differs from classical synaptic transmission in important ways. In synaptic transmission, vesicles rapidly fuse in response to action potentials and release their transmitter content. The transmitters are then sensed by nearby receptors on select target cells with minimal delay. Signal transmission is restricted to synaptic contacts and typically occurs within ~1 ms. Volume transmission doesn't rely on synaptic contact sites and is the main mode of monoamines and neuropeptides, important neuromodulators in the brain. It is less precise than synaptic transmission, and the underlying molecular mechanisms and spatiotemporal scales are often not well understood. Here, we review literature on mechanisms of volume transmission and raise scientific questions that should be addressed in the years ahead. We define five domains by which volume transmission systems can differ from synaptic transmission and from one another. These domains are (1) innervation patterns and firing properties, (2) transmitter synthesis and loading into different types of vesicles, (3) architecture and distribution of release sites, (4) transmitter diffusion, degradation, and reuptake, and (5) receptor types and their positioning on target cells. We discuss these five domains for dopamine, a well-studied monoamine, and then compare the literature on dopamine with that on norepinephrine and serotonin. We include assessments of neuropeptide signaling and of central acetylcholine transmission. Through this review, we provide a molecular and cellular framework for volume transmission. This mechanistic knowledge is essential to define how neuromodulatory systems control behavior in health and disease and to understand how they are modulated by medical treatments and by drugs of abuse.
Collapse
Affiliation(s)
- Özge D Özçete
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
2
|
Fantini J, Azzaz F, Bennaï R, Yahi N, Chahinian H. Cholesterol-Dependent Serotonin Insertion Controlled by Gangliosides in Model Lipid Membranes. Int J Mol Sci 2024; 25:10194. [PMID: 39337677 PMCID: PMC11432689 DOI: 10.3390/ijms251810194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/11/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Serotonin is distinct among synaptic neurotransmitters because it is amphipathic and released from synaptic vesicles at concentrations superior to its water solubility limit (270 mM in synaptic vesicles for a solubility limit of 110 mM). Hence, serotonin is mostly aggregated in the synaptic cleft, due to extensive aromatic stacking. This important characteristic has received scant attention, as most representations of the serotonergic synapse take as warranted that serotonin molecules are present as monomers after synaptic vesicle exocytosis. Using a combination of in silico and physicochemical approaches and a new experimental device mimicking synaptic conditions, we show that serotonin aggregates are efficiently dissolved by gangliosides (especially GM1) present in postsynaptic membranes. This initial interaction, driven by electrostatic forces, attracts serotonin from insoluble aggregates and resolves micelles into monomers. Serotonin also interacts with cholesterol via a set of CH-π and van der Waals interactions. Thus, gangliosides and cholesterol act together as a functional serotonin-collecting funnel on brain cell membranes. Based on this unique mode of interaction with postsynaptic membranes, we propose a new model of serotonergic transmission that takes into account the post-exocytosis solubilizing effect of gangliosides and cholesterol on serotonin aggregates.
Collapse
Affiliation(s)
| | | | | | - Nouara Yahi
- Department of Biology, Faculty of Medicine, University of Aix-Marseille, INSERM UA16, 13015 Marseille, France; (J.F.); (F.A.); (R.B.); (H.C.)
| | | |
Collapse
|
3
|
Yeoman MS, Fidalgo S, Marcelli G, Patel BA. Amperometry approach curve profiling to understand the regulatory mechanisms governing the concentration of intestinal extracellular serotonin. Sci Rep 2024; 14:10479. [PMID: 38714793 PMCID: PMC11076564 DOI: 10.1038/s41598-024-61296-9] [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: 12/28/2023] [Accepted: 05/03/2024] [Indexed: 05/10/2024] Open
Abstract
Enterochromaffin (EC) cells located within the intestinal mucosal epithelium release serotonin (5-HT) to regulate motility tones, barrier function and the immune system. Electroanalytical methodologies have been able to monitor steady state basal extracellular 5-HT levels but are unable to provide insight into how these levels are influenced by key regulatory processes such as release and uptake. We established a new measurement approach, amperometry approach curve profiling, which monitors the extracellular 5-HT level at different electrode-tissue (E-T) distances. Analysis of the current profile can provide information on contributions of regulatory components on the observed extracellular 5-HT level. Measurements were conducted from ex vivo murine ileum and colon using a boron-doped diamond (BDD) microelectrode. Amperometry approach curve profiling coupled with classical pharmacology demonstrated that extracellular 5-HT levels were significantly lower in the colon when compared to the ileum. This difference was due to a greater degree of activity of the 5-HT transporter (SERT) and a reduced amount of 5-HT released from colonic EC cells. The presence of an inhibitory 5-HT4 autoreceptor was observed in the colon, where a 40% increase in extracellular 5-HT was the half maximal inhibitory concentration for activation of the autoreceptor. This novel electroanalytical approach allows estimates of release and re-uptake and their contribution to 5-HT extracellular concentration from intestinal tissue be obtained from a single series of measurements.
Collapse
Affiliation(s)
- Mark S Yeoman
- School of Applied Sciences, University of Brighton, Huxley Building, Brighton, BN2 4GJ, UK
- Centre for Lifelong Health, University of Brighton, Huxley Building, Brighton, BN2 4GJ, UK
| | - Sara Fidalgo
- School of Applied Sciences, University of Brighton, Huxley Building, Brighton, BN2 4GJ, UK
- Centre for Lifelong Health, University of Brighton, Huxley Building, Brighton, BN2 4GJ, UK
| | - Gianluca Marcelli
- School of Engineering, University of Kent, Jennison Building, Canterbury, CT2 7NZ, UK
| | - Bhavik Anil Patel
- School of Applied Sciences, University of Brighton, Huxley Building, Brighton, BN2 4GJ, UK.
- Centre for Lifelong Health, University of Brighton, Huxley Building, Brighton, BN2 4GJ, UK.
| |
Collapse
|
4
|
Lee WL, Westergaard X, Hwu C, Hwu J, Fiala T, Lacefield C, Boltaev U, Mendieta AM, Lin L, Sonders MS, Brown KR, He K, Asher WB, Javitch JA, Sulzer D, Sames D. Molecular Design of SERTlight: A Fluorescent Serotonin Probe for Neuronal Labeling in the Brain. J Am Chem Soc 2024; 146:9564-9574. [PMID: 38557024 DOI: 10.1021/jacs.3c11617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The serotonergic transmitter system plays fundamental roles in the nervous system in neurotransmission, synaptic plasticity, pathological processes, and therapeutic effects of antidepressants and psychedelics, as well as in the gastrointestinal and circulatory systems. We introduce a novel small molecule fluorescent agent, termed SERTlight, that specifically labels serotonergic neuronal cell bodies, dendrites, and axonal projections as a serotonin transporter (SERT) fluorescent substrate. SERTlight was developed by an iterative molecular design process, based on an aminoethyl-quinolone system, to integrate structural elements that impart SERT substrate activity, sufficient fluorescent brightness, and a broad absence of pharmacological activity, including at serotonin (5-hydroxytryptamine, 5HT) receptors, other G protein-coupled receptors (GPCRs), ion channels, and monoamine transporters. The high labeling selectivity is not achieved by high affinity binding to SERT itself but rather by a sufficient rate of SERT-mediated transport of SERTlight, resulting in accumulation of these molecules in 5HT neurons and yielding a robust and selective optical signal in the mammalian brain. SERTlight provides a stable signal, as it is not released via exocytosis nor by reverse SERT transport induced by 5HT releasers such as MDMA. SERTlight is optically, pharmacologically, and operationally orthogonal to a wide range of genetically encoded sensors, enabling multiplexed imaging. SERTlight enables labeling of distal 5HT axonal projections and simultaneous imaging of the release of endogenous 5HT using the GRAB5HT sensor, providing a new versatile molecular tool for the study of the serotonergic system.
Collapse
Affiliation(s)
- Wei-Li Lee
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Xavier Westergaard
- Department of Biological Sciences, Columbia University, New York, New York 10027, United States
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Christopher Hwu
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Jennifer Hwu
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Tomas Fiala
- Department of Chemistry, Columbia University, New York, New York 10027, United States
- Laboratory of Organic Chemistry, ETH Zürich, D-CHAB, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Clay Lacefield
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10032, United States
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, New York 10032, United States
| | - Umed Boltaev
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Adriana M Mendieta
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Lisa Lin
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10032, United States
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Mark S Sonders
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10032, United States
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York 10032, United States
| | - Keaon R Brown
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Keer He
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Wesley B Asher
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10032, United States
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Jonathan A Javitch
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10032, United States
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York 10032, United States
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York 10032, United States
| | - David Sulzer
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10032, United States
- Department of Neurology, Columbia University Irving Medical Center, New York, New York 10032, United States
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York 10032, United States
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York 10032, United States
| | - Dalibor Sames
- Department of Chemistry, Columbia University, New York, New York 10027, United States
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York 10027, United States
| |
Collapse
|
5
|
Deng F, Wan J, Li G, Dong H, Xia X, Wang Y, Li X, Zhuang C, Zheng Y, Liu L, Yan Y, Feng J, Zhao Y, Xie H, Li Y. Improved green and red GRAB sensors for monitoring spatiotemporal serotonin release in vivo. Nat Methods 2024; 21:692-702. [PMID: 38443508 PMCID: PMC11377854 DOI: 10.1038/s41592-024-02188-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 01/19/2024] [Indexed: 03/07/2024]
Abstract
The serotonergic system plays important roles in both physiological and pathological processes, and is a therapeutic target for many psychiatric disorders. Although several genetically encoded GFP-based serotonin (5-HT) sensors were recently developed, their sensitivities and spectral profiles are relatively limited. To overcome these limitations, we optimized green fluorescent G-protein-coupled receptor (GPCR)-activation-based 5-HT (GRAB5-HT) sensors and developed a red fluorescent GRAB5-HT sensor. These sensors exhibit excellent cell surface trafficking and high specificity, sensitivity and spatiotemporal resolution, making them suitable for monitoring 5-HT dynamics in vivo. Besides recording subcortical 5-HT release in freely moving mice, we observed both uniform and gradient 5-HT release in the mouse dorsal cortex with mesoscopic imaging. Finally, we performed dual-color imaging and observed seizure-induced waves of 5-HT release throughout the cortex following calcium and endocannabinoid waves. In summary, these 5-HT sensors can offer valuable insights regarding the serotonergic system in both health and disease.
Collapse
Affiliation(s)
- Fei Deng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jinxia Wan
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Guochuan Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Hui Dong
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Xiju Xia
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yipan Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Xuelin Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Chaowei Zhuang
- Department of Automation, Tsinghua University, Beijing, China
| | - Yu Zheng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Laixin Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yuqi Yan
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Jiesi Feng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Yulin Zhao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Hao Xie
- Department of Automation, Tsinghua University, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China.
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
- Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
6
|
Honan LE, Fraser-Spears R, Daws LC. Organic cation transporters in psychiatric and substance use disorders. Pharmacol Ther 2024; 253:108574. [PMID: 38072333 PMCID: PMC11052553 DOI: 10.1016/j.pharmthera.2023.108574] [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: 10/06/2023] [Revised: 11/01/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Psychiatric and substance use disorders inflict major public health burdens worldwide. Their widespread burden is compounded by a dearth of effective treatments, underscoring a dire need to uncover novel therapeutic targets. In this review, we summarize the literature implicating organic cation transporters (OCTs), including three subtypes of OCTs (OCT1, OCT2, and OCT3) and the plasma membrane monoamine transporter (PMAT), in the neurobiology of psychiatric and substance use disorders with an emphasis on mood and anxiety disorders, alcohol use disorder, and psychostimulant use disorder. OCTs transport monoamines with a low affinity but high capacity, situating them to play a central role in regulating monoamine homeostasis. Preclinical evidence discussed here suggests that OCTs may serve as promising targets for treatment of psychiatric and substance use disorders and encourage future research into their therapeutic potential.
Collapse
Affiliation(s)
- Lauren E Honan
- The University of Texas Health Science Center at San Antonio, Department of Cellular & Integrative Physiology, USA
| | - Rheaclare Fraser-Spears
- University of the Incarnate Word, Feik School of Pharmacy, Department of Pharmaceutical Sciences, USA
| | - Lynette C Daws
- The University of Texas Health Science Center at San Antonio, Department of Cellular & Integrative Physiology, USA; The University of Texas Health Science Center at San Antonio, Department of Pharmacology, USA.
| |
Collapse
|
7
|
Gianni G, Pasqualetti M. Wiring and Volume Transmission: An Overview of the Dual Modality for Serotonin Neurotransmission. ACS Chem Neurosci 2023; 14:4093-4104. [PMID: 37966717 DOI: 10.1021/acschemneuro.3c00648] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023] Open
Abstract
Serotonin is a neurotransmitter involved in the modulation of a multitude of physiological and behavioral processes. In spite of the relatively reduced number of serotonin-producing neurons present in the mammalian CNS, a complex long-range projection system provides profuse innervation to the whole brain. Heterogeneity of serotonin receptors, grouped in seven families, and their spatiotemporal expression pattern account for its widespread impact. Although neuronal communication occurs primarily at tiny gaps called synapses, wiring transmission, another mechanism based on extrasynaptic diffusion of neuroactive molecules and referred to as volume transmission, has been described. While wiring transmission is a rapid and specific one-to-one modality of communication, volume transmission is a broader and slower mode in which a single element can simultaneously act on several different targets in a one-to-many mode. Some experimental evidence regarding ultrastructural features, extrasynaptic localization of receptors and transporters, and serotonin-glia interactions collected over the past four decades supports the existence of a serotonergic system of a dual modality of neurotransmission, in which wiring and volume transmission coexist. To date, in spite of the radical difference in the two modalities, limited information is available on the way they are coordinated to mediate the specific activities in which serotonin participates. Understanding how wiring and volume transmission modalities contribute to serotonergic neurotransmission is of utmost relevance for the comprehension of serotonin functions in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Giulia Gianni
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56127 Pisa, Italy
| | - Massimo Pasqualetti
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56127 Pisa, Italy
- Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy
- Centro per l'Integrazione della Strumentazione Scientifica dell'Università di Pisa (CISUP), 56126 Pisa, Italy
| |
Collapse
|
8
|
Nithianandam P, Tzu-li L, Chen S, Yizhen J, Dong Y, Saul M, Tedeschi A, Wenjing S, Jinghua L. Flexible, Miniaturized Sensing Probes Inspired by Biofuel Cells for Monitoring Synaptically Released Glutamate in the Mouse Brain. Angew Chem Int Ed Engl 2023; 62:e202310245. [PMID: 37632702 PMCID: PMC10592105 DOI: 10.1002/anie.202310245] [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: 07/18/2023] [Revised: 08/20/2023] [Accepted: 08/24/2023] [Indexed: 08/28/2023]
Abstract
Chemical biomarkers in the central nervous system can provide valuable quantitative measures to gain insight into the etiology and pathogenesis of neurological diseases. Glutamate, one of the most important excitatory neurotransmitters in the brain, has been found to be upregulated in various neurological disorders, such as traumatic brain injury, Alzheimer's disease, stroke, epilepsy, chronic pain, and migraines. However, quantitatively monitoring glutamate release in situ has been challenging. This work presents a novel class of flexible, miniaturized probes inspired by biofuel cells for monitoring synaptically released glutamate in the nervous system. The resulting sensors, with dimensions as low as 50 by 50 μm, can detect real-time changes in glutamate within the biologically relevant concentration range. Experiments exploiting the hippocampal circuit in mice models demonstrate the capability of the sensors in monitoring glutamate release via electrical stimulation using acute brain slices. These advances could aid in basic neuroscience studies and translational engineering, as the sensors provide a diagnostic tool for neurological disorders. Additionally, adapting the biofuel cell design to other neurotransmitters can potentially enable the detailed study of the effect of neurotransmitter dysregulation on neuronal cell signaling pathways and revolutionize neuroscience.
Collapse
Affiliation(s)
- Prasad Nithianandam
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Liu Tzu-li
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Shulin Chen
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Jia Yizhen
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Yan Dong
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Morgan Saul
- Department of Neuroscience, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Andrea Tedeschi
- Department of Neuroscience, The Ohio State University College of Medicine, Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA
| | - Sun Wenjing
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Li Jinghua
- Department of Materials Science and Engineering, Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
9
|
Long DR, Kinser A, Olalde-Welling A, Brewer L, Lim J, Matheny D, Long B, Roossien DH. 5-HT1A regulates axon outgrowth in a subpopulation of Drosophila serotonergic neurons. Dev Neurobiol 2023; 83:268-281. [PMID: 37714743 DOI: 10.1002/dneu.22928] [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: 10/12/2022] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/17/2023]
Abstract
Serotonergic neurons produce extensively branched axons that fill most of the central nervous system, where they modulate a wide variety of behaviors. Many behavioral disorders have been correlated with defective serotonergic axon morphologies. Proper behavioral output therefore depends on the precise outgrowth and targeting of serotonergic axons during development. To direct outgrowth, serotonergic neurons utilize serotonin as a signaling molecule prior to it assuming its neurotransmitter role. This process, termed serotonin autoregulation, regulates axon outgrowth, branching, and varicosity development of serotonergic neurons. However, the receptor that mediates serotonin autoregulation is unknown. Here we asked if serotonin receptor 5-HT1A plays a role in serotonergic axon outgrowth and branching. Using cultured Drosophila serotonergic neurons, we found that exogenous serotonin reduced axon length and branching only in those expressing 5-HT1A. Pharmacological activation of 5-HT1A led to reduced axon length and branching, whereas the disruption of 5-HT1A rescued outgrowth in the presence of exogenous serotonin. Altogether this suggests that 5-HT1A is a serotonin autoreceptor in a subpopulation of serotonergic neurons and initiates signaling pathways that regulate axon outgrowth and branching during Drosophila development.
Collapse
Affiliation(s)
- Delaney R Long
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | - Ava Kinser
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | | | - Luke Brewer
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | - Juri Lim
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | - Dayle Matheny
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | - Breanna Long
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | | |
Collapse
|
10
|
Bartlett EA, Zanderigo F, Stanley B, Choo TH, Galfalvy HC, Pantazatos SP, Sublette ME, Miller JM, Oquendo MA, Mann JJ. In vivo serotonin transporter and 1A receptor binding potential and ecological momentary assessment (EMA) of stress in major depression and suicidal behavior. Eur Neuropsychopharmacol 2023; 70:1-13. [PMID: 36780841 PMCID: PMC10121874 DOI: 10.1016/j.euroneuro.2023.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/17/2023] [Accepted: 01/24/2023] [Indexed: 02/13/2023]
Abstract
We examined relationships between the serotonin system and stress in major depression and suicidal behavior. Twenty-five medication-free depressed participants (13 suicide attempters) underwent same-day [11C]DASB and [11C]CUMI-101 positron emission tomography (PET) imaging. Binding potential (BPND) to the serotonin transporter (5-HTT) and serotonin 1A (5-HT1A) receptor, respectively, was quantified using the NRU 5-HT atlas, reflecting distinct spatial distributions of multiple serotonin targets. Ecological momentary assessment (EMA) measured current stress over one week proximal to imaging. EMA stress did not differ between attempters and non-attempters. In all depressed participants, 5-HTT and 5-HT1A BPND were unrelated to EMA stress. There were region-specific effects of 5-HTT (p=0.002) and 5-HT1A BPND (p=0.03) in attempters vs. nonattempters. In attempters, region-specific associations between 5-HTT (p=0.03) and 5-HT1A (p=0.005) BPND and EMA stress emerged. While no post-hoc 5-HTT BPND correlations were significant, 5-HT1A BPND correlated positively with EMA stress in attempters in 9/10 regions (p-values<0.007), including the entire cortex except the largely occipital region 5. Brodmann-based regional analyses found diminished effects for 5-HTT and subcortically localized positive corrrelations between 5-HT1A and EMA stress, in attempters only. Given comparable depression severity and childhood and current stress between attempters and nonattempters, lower 5-HTT binding in attempters vs. nonattempters may suggest a biological risk marker. Localized lower 5-HTT and widespread higher 5-HT1A binding with stress among attempters specifically may suggest that a serotonergic phenotype might be a key determinant of risk or resiliency for suicidal behavior.
Collapse
Affiliation(s)
- Elizabeth A Bartlett
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, USA.
| | - Francesca Zanderigo
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, USA
| | - Barbara Stanley
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, USA
| | - Tse-Hwei Choo
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, USA
| | - Hanga C Galfalvy
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, USA
| | - Spiro P Pantazatos
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, USA
| | - M Elizabeth Sublette
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, USA
| | - Jeffrey M Miller
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, USA
| | - Maria A Oquendo
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - J John Mann
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, USA; Department of Radiology, Columbia University Irving Medical Center, New York, USA
| |
Collapse
|
11
|
Plausible Role of Stem Cell Types for Treating and Understanding the Pathophysiology of Depression. Pharmaceutics 2023; 15:pharmaceutics15030814. [PMID: 36986674 PMCID: PMC10058940 DOI: 10.3390/pharmaceutics15030814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/09/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Major Depressive Disorder (MDD), colloquially known as depression, is a debilitating condition affecting an estimated 3.8% of the population globally, of which 5.0% are adults and 5.7% are above the age of 60. MDD is differentiated from common mood changes and short-lived emotional responses due to subtle alterations in gray and white matter, including the frontal lobe, hippocampus, temporal lobe, thalamus, striatum, and amygdala. It can be detrimental to a person’s overall health if it occurs with moderate or severe intensity. It can render a person suffering terribly to perform inadequately in their personal, professional, and social lives. Depression, at its peak, can lead to suicidal thoughts and ideation. Antidepressants manage clinical depression and function by modulating the serotonin, norepinephrine, and dopamine neurotransmitter levels in the brain. Patients with MDD positively respond to antidepressants, but 10–30% do not recuperate or have a partial response accompanied by poor life quality, suicidal ideation, self-injurious behavior, and an increased relapse rate. Recent research shows that mesenchymal stem cells and iPSCs may be responsible for lowering depression by producing more neurons with increased cortical connections. This narrative review discusses the plausible functions of various stem cell types in treating and understanding depression pathophysiology.
Collapse
|
12
|
Zheng D, Pisano F, Collard L, Balena A, Pisanello M, Spagnolo B, Mach-Batlle R, Tantussi F, Carbone L, De Angelis F, Valiente M, de la Prida LM, Ciracì C, De Vittorio M, Pisanello F. Toward Plasmonic Neural Probes: SERS Detection of Neurotransmitters through Gold-Nanoislands-Decorated Tapered Optical Fibers with Sub-10 nm Gaps. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2200902. [PMID: 36479741 DOI: 10.1002/adma.202200902] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 10/21/2022] [Indexed: 06/17/2023]
Abstract
Integration of plasmonic nanostructures with fiber-optics-based neural probes enables label-free detection of molecular fingerprints via surface-enhanced Raman spectroscopy (SERS), and it represents a fascinating technological horizon to investigate brain function. However, developing neuroplasmonic probes that can interface with deep brain regions with minimal invasiveness while providing the sensitivity to detect biomolecular signatures in a physiological environment is challenging, in particular because the same waveguide must be employed for both delivering excitation light and collecting the resulting scattered photons. Here, a SERS-active neural probe based on a tapered optical fiber (TF) decorated with gold nanoislands (NIs) that can detect neurotransmitters down to the micromolar range is presented. To do this, a novel, nonplanar repeated dewetting technique to fabricate gold NIs with sub-10 nm gaps, uniformly distributed on the wide (square millimeter scale in surface area), highly curved surface of TF is developed. It is experimentally and numerically shown that the amplified broadband near-field enhancement of the high-density NIs layer allows for achieving a limit of detection in aqueous solution of 10-7 m for rhodamine 6G and 10-5 m for serotonin and dopamine through SERS at near-infrared wavelengths. The NIs-TF technology is envisioned as a first step toward the unexplored frontier of in vivo label-free plasmonic neural interfaces.
Collapse
Affiliation(s)
- Di Zheng
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Filippo Pisano
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Liam Collard
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Antonio Balena
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Marco Pisanello
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Barbara Spagnolo
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Rosa Mach-Batlle
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Francesco Tantussi
- Istituto Italiano di Tecnologia, Center for Convergent Technologies, Genova, 16163, Italy
| | - Luigi Carbone
- CNR NANOTEC - Institute of Nanotechnology, University of Salento, Lecce, 73100, Italy
| | - Francesco De Angelis
- Istituto Italiano di Tecnologia, Center for Convergent Technologies, Genova, 16163, Italy
| | - Manuel Valiente
- Brain Metastasis Group, Spanish National Cancer Research Center (CNIO), Madrid, 28029, Spain
| | | | - Cristian Ciracì
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Massimo De Vittorio
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
- Dipartimento di Ingegneria Dell'Innovazione, Università del Salento, Lecce, 73100, Italy
| | - Ferruccio Pisanello
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| |
Collapse
|
13
|
Next generation genetically encoded fluorescent sensors for serotonin. Nat Commun 2022; 13:7525. [PMID: 36473867 PMCID: PMC9726753 DOI: 10.1038/s41467-022-35200-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
We developed a family of genetically encoded serotonin (5-HT) sensors (sDarken) on the basis of the native 5-HT1A receptor and circularly permuted GFP. sDarken 5-HT sensors are bright in the unbound state and diminish their fluorescence upon binding of 5-HT. Sensor variants with different affinities for serotonin were engineered to increase the versatility in imaging of serotonin dynamics. Experiments in vitro and in vivo showed the feasibility of imaging serotonin dynamics with high temporal and spatial resolution. As demonstrated here, the designed sensors show excellent membrane expression, have high specificity and a superior signal-to-noise ratio, detect the endogenous release of serotonin and are suitable for two-photon in vivo imaging.
Collapse
|
14
|
Andrews PW, Bosyj C, Brenton L, Green L, Gasser PJ, Lowry CA, Pickel VM. All the brain's a stage for serotonin: the forgotten story of serotonin diffusion across cell membranes. Proc Biol Sci 2022; 289:20221565. [PMID: 36321487 PMCID: PMC9627707 DOI: 10.1098/rspb.2022.1565] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022] Open
Abstract
In the conventional model of serotonin neurotransmission, serotonin released by neurons in the midbrain raphe nuclei exerts its actions on forebrain neurons by interacting with a large family of post-synaptic receptors. The actions of serotonin are terminated by active transport of serotonin back into the releasing neuron, which is mediated by the serotonin reuptake transporter (SERT). Because SERT is expressed pre-synaptically and is widely thought to be the only serotonin transporter in the forebrain, the conventional model does not include serotonin transport into post-synaptic neurons. However, a large body of evidence accumulating since the 1970s has shown that serotonin, despite having a positive charge, can cross cell membranes through a diffusion-like process. Multiple low-affinity, high-capacity, sodium-independent transporters, widely expressed in the brain, allow the carrier-mediated diffusion of serotonin into forebrain neurons. The amount of serotonin crossing cell membranes through this mechanism under physiological conditions is considerable. Most prominent textbooks fail to include this alternative method of serotonin uptake in the brain, and even most neuroscientists are unaware of it. This failure has limited our understanding of a key regulator of serotonergic neurotransmission, impeded research on the potential intracellular actions of serotonin in post-synaptic neurons and glial cells, and may have impeded our understanding of the mechanism by which antidepressant medications reduce depressive symptoms.
Collapse
Affiliation(s)
- Paul W. Andrews
- Department of Psychology, Neuroscience and Behaviour, McMaster University, Hamilton, Ontario, Canada
| | - Catherine Bosyj
- Department of Psychology, Neuroscience and Behaviour, McMaster University, Hamilton, Ontario, Canada
| | - Luke Brenton
- Department of Psychology, Neuroscience and Behaviour, McMaster University, Hamilton, Ontario, Canada
| | - Laura Green
- Neuroscience Institute, New York University, New York, NY, USA
| | - Paul J. Gasser
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Christopher A. Lowry
- Department of Integrative Physiology, Center for Neuroscience, and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, USA
| | - Virginia M. Pickel
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
15
|
Pan Y, Zhang K, Wei H, Xiong T, Liu Y, Mao L, Yu P. Double-Barreled Micropipette Enables Neuron-Compatible In Vivo Analysis. Anal Chem 2022; 94:15671-15677. [DOI: 10.1021/acs.analchem.2c02739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Yifei Pan
- Key Laboratory of Analytical Chemistry for Living Biosystems Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing100190, China
- School of Chemical Science, University of Chinese Academy of Sciences (CAS), Beijing100190, China
| | - Kailin Zhang
- Key Laboratory of Analytical Chemistry for Living Biosystems Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing100190, China
- School of Chemical Science, University of Chinese Academy of Sciences (CAS), Beijing100190, China
| | - Huan Wei
- Key Laboratory of Analytical Chemistry for Living Biosystems Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing100190, China
- College of Chemistry, Beijing Normal University, Beijing100875, China
| | - Tianyi Xiong
- Key Laboratory of Analytical Chemistry for Living Biosystems Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing100190, China
- School of Chemical Science, University of Chinese Academy of Sciences (CAS), Beijing100190, China
| | - Ying Liu
- Key Laboratory of Analytical Chemistry for Living Biosystems Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing100190, China
- School of Chemical Science, University of Chinese Academy of Sciences (CAS), Beijing100190, China
| | - Lanqun Mao
- College of Chemistry, Beijing Normal University, Beijing100875, China
| | - Ping Yu
- Key Laboratory of Analytical Chemistry for Living Biosystems Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing100190, China
- School of Chemical Science, University of Chinese Academy of Sciences (CAS), Beijing100190, China
| |
Collapse
|
16
|
Flavell SW, Gogolla N, Lovett-Barron M, Zelikowsky M. The emergence and influence of internal states. Neuron 2022; 110:2545-2570. [PMID: 35643077 PMCID: PMC9391310 DOI: 10.1016/j.neuron.2022.04.030] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 02/11/2022] [Accepted: 04/27/2022] [Indexed: 01/09/2023]
Abstract
Animal behavior is shaped by a variety of "internal states"-partially hidden variables that profoundly shape perception, cognition, and action. The neural basis of internal states, such as fear, arousal, hunger, motivation, aggression, and many others, is a prominent focus of research efforts across animal phyla. Internal states can be inferred from changes in behavior, physiology, and neural dynamics and are characterized by properties such as pleiotropy, persistence, scalability, generalizability, and valence. To date, it remains unclear how internal states and their properties are generated by nervous systems. Here, we review recent progress, which has been driven by advances in behavioral quantification, cellular manipulations, and neural population recordings. We synthesize research implicating defined subsets of state-inducing cell types, widespread changes in neural activity, and neuromodulation in the formation and updating of internal states. In addition to highlighting the significance of these findings, our review advocates for new approaches to clarify the underpinnings of internal brain states across the animal kingdom.
Collapse
Affiliation(s)
- Steven W Flavell
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Nadine Gogolla
- Emotion Research Department, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Circuits for Emotion Research Group, Max Planck Institute of Neurobiology, 82152 Martinsried, Germany.
| | - Matthew Lovett-Barron
- Division of Biological Sciences-Neurobiology Section, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Moriel Zelikowsky
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
17
|
Debnath A, Williams PDE, Bamber BA. Reduced Ca2+ transient amplitudes may signify increased or decreased depolarization depending on the neuromodulatory signaling pathway. Front Neurosci 2022; 16:931328. [PMID: 35937887 PMCID: PMC9354622 DOI: 10.3389/fnins.2022.931328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Neuromodulators regulate neuronal excitability and bias neural circuit outputs. Optical recording of neuronal Ca2+ transients is a powerful approach to study the impact of neuromodulators on neural circuit dynamics. We are investigating the polymodal nociceptor ASH in Caenorhabditis elegans to better understand the relationship between neuronal excitability and optically recorded Ca2+ transients. ASHs depolarize in response to the aversive olfactory stimulus 1-octanol (1-oct) with a concomitant rise in somal Ca2+, stimulating an aversive locomotory response. Serotonin (5-HT) potentiates 1-oct avoidance through Gαq signaling, which inhibits L-type voltage-gated Ca2+ channels in ASH. Although Ca2+ signals in the ASH soma decrease, depolarization amplitudes increase because Ca2+ mediates inhibitory feedback control of membrane potential in this context. Here, we investigate octopamine (OA) signaling in ASH to assess whether this negative correlation between somal Ca2+ and depolarization amplitudes is a general phenomenon, or characteristic of certain neuromodulatory pathways. Like 5-HT, OA reduces somal Ca2+ transient amplitudes in ASH neurons. However, OA antagonizes 5-HT modulation of 1-oct avoidance behavior, suggesting that OA may signal through a different pathway. We further show that the pathway for OA diminution of ASH somal Ca2+ consists of the OCTR-1 receptor, the Go heterotrimeric G-protein, and the G-protein activated inwardly rectifying channels IRK-2 and IRK-3, and this pathway reduces depolarization amplitudes in parallel with somal Ca2+ transient amplitudes. Therefore, even within a single neuron, somal Ca2+ signal reduction may indicate either increased or decreased depolarization amplitude, depending on which neuromodulatory signaling pathways are activated, underscoring the need for careful interpretation of Ca2+ imaging data in neuromodulatory studies.
Collapse
Affiliation(s)
- Arunima Debnath
- Department of Biological Sciences, The University of Toledo, Toledo, OH, United States
| | - Paul D. E. Williams
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Bruce A. Bamber
- Department of Biological Sciences, The University of Toledo, Toledo, OH, United States
- *Correspondence: Bruce A. Bamber,
| |
Collapse
|
18
|
Holmes J, Lau T, Saylor R, Fernández-Novel N, Hersey M, Keen D, Hampel L, Horschitz S, Ladewig J, Parke B, Reed MC, Nijhout HF, Best J, Koch P, Hashemi P. Voltammetric Approach for Characterizing the Biophysical and Chemical Functionality of Human Induced Pluripotent Stem Cell-Derived Serotonin Neurons. Anal Chem 2022; 94:8847-8856. [PMID: 35713335 DOI: 10.1021/acs.analchem.1c05082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Depression is quickly becoming one of the world's most pressing public health crises, and there is an urgent need for better diagnostics and therapeutics. Behavioral models in animals and humans have not adequately addressed the diagnosis and treatment of depression, and biomarkers of mental illnesses remain ill-defined. It has been very difficult to identify biomarkers of depression because of in vivo measurement challenges. While our group has made important strides in developing in vivo tools to measure such biomarkers (e.g., serotonin) in mice using voltammetry, these tools cannot be easily applied for depression diagnosis and drug screening in humans due to the inaccessibility of the human brain. In this work, we take a chemical approach, ex vivo, to introduce a human-derived system to investigate brain serotonin. We utilize human induced pluripotent stem cells differentiated into serotonin neurons and establish a new ex vivo model of real-time serotonin neurotransmission measurements. We show that evoked serotonin release responds to stimulation intensity and tryptophan preloading, and that serotonin release and reuptake kinetics resemble those found in vivo in rodents. Finally, after selective serotonin reuptake inhibitor (SSRI) exposure, we find dose-dependent internalization of the serotonin reuptake transporters (a signature of the in vivo response to SSRI). Our new human-derived chemical model has great potential to provide an ex vivo chemical platform as a translational tool for in vivo neuropsychopharmacology.
Collapse
Affiliation(s)
- Jordan Holmes
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Thorsten Lau
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Rachel Saylor
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Nadine Fernández-Novel
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Melinda Hersey
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States.,Department of Pharmacology, Physiology, & Neuroscience, University of South Carolina, Columbia, South Carolina 29209, United States
| | - Deanna Keen
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Lena Hampel
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Sandra Horschitz
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Julia Ladewig
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Brenna Parke
- Department of Bioengineering, Imperial College London, London SW7 2AZ, U.K
| | - Michael C Reed
- Department of Mathematics, Duke University, Durham, North Carolina 27708, United States
| | - H Frederik Nijhout
- Department of Biology, Duke University, Durham, North Carolina 27708, United States
| | - Janet Best
- Department of Mathematics, The Ohio State University, Columbus, Ohio 43210, United States
| | - Philipp Koch
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Parastoo Hashemi
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States.,Department of Bioengineering, Imperial College London, London SW7 2AZ, U.K
| |
Collapse
|
19
|
Cooke P, Janowitz H, Dougherty SE. Neuronal Redevelopment and the Regeneration of Neuromodulatory Axons in the Adult Mammalian Central Nervous System. Front Cell Neurosci 2022; 16:872501. [PMID: 35530177 PMCID: PMC9074815 DOI: 10.3389/fncel.2022.872501] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/24/2022] [Indexed: 01/09/2023] Open
Abstract
One reason that many central nervous system injuries, including those arising from traumatic brain injury, spinal cord injury, and stroke, have limited recovery of function is that neurons within the adult mammalian CNS lack the ability to regenerate their axons following trauma. This stands in contrast to neurons of the adult mammalian peripheral nervous system (PNS). New evidence, provided by single-cell expression profiling, suggests that, following injury, both mammalian central and peripheral neurons can revert to an embryonic-like growth state which is permissive for axon regeneration. This “redevelopment” strategy could both facilitate a damage response necessary to isolate and repair the acute damage from injury and provide the intracellular machinery necessary for axon regrowth. Interestingly, serotonin neurons of the rostral group of raphe nuclei, which project their axons into the forebrain, display a robust ability to regenerate their axons unaided, counter to the widely held view that CNS axons cannot regenerate without experimental intervention after injury. Furthermore, initial evidence suggests that norepinephrine neurons within the locus coeruleus possess similar regenerative abilities. Several morphological characteristics of serotonin axon regeneration in adult mammals, observable using longitudinal in vivo imaging, are distinct from the known characteristics of unaided peripheral nerve regeneration, or of the regeneration seen in the spinal cord and optic nerve that occurs with experimental intervention. These results suggest that there is an alternative CNS program for axon regeneration that likely differs from that displayed by the PNS.
Collapse
Affiliation(s)
- Patrick Cooke
- Linden Lab, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Haley Janowitz
- Linden Lab, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sarah E Dougherty
- Linden Lab, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
20
|
Characterization of a new type of neuronal 5-HT G- protein coupled receptor in the cestode nervous system. PLoS One 2021; 16:e0259104. [PMID: 34762657 PMCID: PMC8584985 DOI: 10.1371/journal.pone.0259104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 10/12/2021] [Indexed: 11/29/2022] Open
Abstract
Cestodes are platyhelminth parasites with a wide range of hosts that cause neglected diseases. Neurotransmitter signaling is of critical importance for these parasites which lack circulatory, respiratory and digestive systems. For example, serotonin (5-HT) and serotonergic G-protein coupled receptors (5-HT GPCRs) play major roles in cestode motility, development and reproduction. In previous work, we deorphanized a group of 5-HT7 type GPCRs from cestodes. However, little is known about another type of 5-HT GPCR, the 5-HT1 clade, which has been studied in several invertebrate phyla but not in platyhelminthes. Three putative 5-HT GPCRs from Echinococcus canadensis, Mesocestoides vogae (syn. M. corti) and Hymenolepis microstoma were cloned, sequenced and bioinformatically analyzed. Evidence grouped these new sequences within the 5-HT1 clade of GPCRs but differences in highly conserved GPCR motifs were observed. Transcriptomic analysis, heterologous expression and immunolocalization studies were performed to characterize the E. canadensis receptor, called Eca-5-HT1a. Functional heterologous expression studies showed that Eca-5-HT1a is highly specific for serotonin. 5-Methoxytryptamine and α-methylserotonin, both known 5-HT GPCR agonists, give stimulatory responses whereas methysergide, a known 5-HT GPCR ligand, give an antagonist response in Eca-5-HT1a. Mutants obtained by the substitution of key predicted residues resulted in severe impairment of receptor activity, confirming that indeed, these residues have important roles in receptor function. Immunolocalization studies on the protoscolex stage from E. canadensis, showed that Eca-5-HT1a is localized in branched fibers which correspond to the nervous system of the parasite. The patterns of immunoreactive fibers for Eca-5-HT1a and for serotonin were intimately intertwined but not identical, suggesting that they are two separate groups of fibers. These data provide the first functional, pharmacological and localization report of a serotonergic receptor that putatively belongs to the 5-HT1 type of GPCRs in cestodes. The serotonergic GPCR characterized here may represent a new target for antiparasitic intervention.
Collapse
|
21
|
Wu X, Morishita W, Beier KT, Heifets BD, Malenka RC. 5-HT modulation of a medial septal circuit tunes social memory stability. Nature 2021; 599:96-101. [PMID: 34616037 DOI: 10.1038/s41586-021-03956-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 08/25/2021] [Indexed: 12/16/2022]
Abstract
Social memory-the ability to recognize and remember familiar conspecifics-is critical for the survival of an animal in its social group1,2. The dorsal CA2 (dCA2)3-5 and ventral CA1 (vCA1)6 subregions of the hippocampus, and their projection targets6,7, have important roles in social memory. However, the relevant extrahippocampal input regions remain poorly defined. Here we identify the medial septum (MS) as a dCA2 input region that is critical for social memory and reveal that modulation of the MS by serotonin (5-HT) bidirectionally controls social memory formation, thereby affecting memory stability. Novel social interactions increase activity in dCA2-projecting MS neurons and induce plasticity at glutamatergic synapses from MS neurons onto dCA2 pyramidal neurons. The activity of dCA2-projecting MS cells is enhanced by the neuromodulator 5-HT acting on 5-HT1B receptors. Moreover, optogenetic manipulation of median raphe 5-HT terminals in the MS bidirectionally regulates social memory stability. This work expands our understanding of the neural mechanisms by which social interactions lead to social memory and provides evidence that 5-HT has a critical role in promoting not only prosocial behaviours8,9, but also social memory, by influencing distinct target structures.
Collapse
Affiliation(s)
- Xiaoting Wu
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Wade Morishita
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Kevin T Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Boris D Heifets
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Robert C Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
| |
Collapse
|
22
|
Hersey M, Hashemi P, Reagan LP. Integrating the monoamine and cytokine hypotheses of depression: Is histamine the missing link? Eur J Neurosci 2021; 55:2895-2911. [PMID: 34265868 DOI: 10.1111/ejn.15392] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 06/26/2021] [Accepted: 07/06/2021] [Indexed: 12/28/2022]
Abstract
Psychiatric diseases, like depression, largely affect the central nervous system (CNS). While the underlying neuropathology of depressive illness remains to be elucidated, several hypotheses have been proposed as molecular underpinnings for major depressive disorder, including the monoamine hypothesis and the cytokine hypothesis. The monoamine hypothesis has been largely supported by the pharmaceuticals that target monoamine neurotransmitters as a treatment for depression. However, these antidepressants have come under scrutiny due to their limited clinical efficacy, side effects, and delayed onset of action. The more recent, cytokine hypothesis of depression is supported by the ability of immune-active agents to induce "sickness behaviour" akin to that seen with depression. However, treatments that more selectively target inflammation have yielded inconsistent antidepressive results. As such, neither of these hypotheses can fully explain depressive illness pathology, implying that the underlying neuropathological mechanisms may encompass aspects of both theories. The goal of the current review is to integrate these two well-studied hypotheses and to propose a role for histamine as a potential unifying factor that links monoamines to cytokines. Additionally, we will focus on stress-induced depression, to provide an updated perspective of depressive illness research and thereby identify new potential targets for the treatment of major depressive disorder.
Collapse
Affiliation(s)
- Melinda Hersey
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina, USA.,Department of Chemistry & Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| | - Parastoo Hashemi
- Department of Chemistry & Biochemistry, University of South Carolina, Columbia, South Carolina, USA.,Department of Bioengineering, Imperial College, London, UK
| | - Lawrence P Reagan
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina, USA.,WJB Dorn Veterans Affairs Medical Center, Columbia, South Carolina, USA
| |
Collapse
|
23
|
Litus EA, Kazakov AS, Deryusheva EI, Nemashkalova EL, Shevelyova MP, Nazipova AA, Permyakova ME, Raznikova EV, Uversky VN, Permyakov SE. Serotonin Promotes Serum Albumin Interaction with the Monomeric Amyloid β Peptide. Int J Mol Sci 2021; 22:ijms22115896. [PMID: 34072751 PMCID: PMC8199245 DOI: 10.3390/ijms22115896] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/28/2021] [Accepted: 05/28/2021] [Indexed: 11/16/2022] Open
Abstract
Prevention of amyloid β peptide (Aβ) deposition via facilitation of Aβ binding to its natural depot, human serum albumin (HSA), is a promising approach to preclude Alzheimer's disease (AD) onset and progression. Previously, we demonstrated the ability of natural HSA ligands, fatty acids, to improve the affinity of this protein to monomeric Aβ by a factor of 3 (BBRC, 510(2), 248-253). Using plasmon resonance spectroscopy, we show here that another HSA ligand related to AD pathogenesis, serotonin (SRO), increases the affinity of the Aβ monomer to HSA by a factor of 7/17 for Aβ40/Aβ42, respectively. Meanwhile, the structurally homologous SRO precursor, tryptophan (TRP), does not affect HSA's affinity to monomeric Aβ, despite slowdown of the association and dissociation processes. Crosslinking with glutaraldehyde and dynamic light scattering experiments reveal that, compared with the TRP-induced effects, SRO binding causes more marked changes in the quaternary structure of HSA. Furthermore, molecular docking reveals distinct structural differences between SRO/TRP complexes with HSA. The disintegration of the serotonergic system during AD pathogenesis may contribute to Aβ release from HSA in the central nervous system due to impairment of the SRO-mediated Aβ trapping by HSA.
Collapse
Affiliation(s)
- Ekaterina A. Litus
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (E.A.L.); (A.S.K.); (E.I.D.); (E.L.N.); (M.P.S.); (A.A.N.); (M.E.P.); (E.V.R.)
| | - Alexey S. Kazakov
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (E.A.L.); (A.S.K.); (E.I.D.); (E.L.N.); (M.P.S.); (A.A.N.); (M.E.P.); (E.V.R.)
| | - Evgenia I. Deryusheva
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (E.A.L.); (A.S.K.); (E.I.D.); (E.L.N.); (M.P.S.); (A.A.N.); (M.E.P.); (E.V.R.)
| | - Ekaterina L. Nemashkalova
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (E.A.L.); (A.S.K.); (E.I.D.); (E.L.N.); (M.P.S.); (A.A.N.); (M.E.P.); (E.V.R.)
| | - Marina P. Shevelyova
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (E.A.L.); (A.S.K.); (E.I.D.); (E.L.N.); (M.P.S.); (A.A.N.); (M.E.P.); (E.V.R.)
| | - Aliya A. Nazipova
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (E.A.L.); (A.S.K.); (E.I.D.); (E.L.N.); (M.P.S.); (A.A.N.); (M.E.P.); (E.V.R.)
| | - Maria E. Permyakova
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (E.A.L.); (A.S.K.); (E.I.D.); (E.L.N.); (M.P.S.); (A.A.N.); (M.E.P.); (E.V.R.)
| | - Elena V. Raznikova
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (E.A.L.); (A.S.K.); (E.I.D.); (E.L.N.); (M.P.S.); (A.A.N.); (M.E.P.); (E.V.R.)
| | - Vladimir N. Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Correspondence: (V.N.U.); (S.E.P.); Tel.: +7-(495)-143-7741 (S.E.P.); Fax: +7-(4967)-33-0522 (S.E.P.)
| | - Sergei E. Permyakov
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (E.A.L.); (A.S.K.); (E.I.D.); (E.L.N.); (M.P.S.); (A.A.N.); (M.E.P.); (E.V.R.)
- Correspondence: (V.N.U.); (S.E.P.); Tel.: +7-(495)-143-7741 (S.E.P.); Fax: +7-(4967)-33-0522 (S.E.P.)
| |
Collapse
|
24
|
Dey S, Surendran D, Engberg O, Gupta A, Fanibunda SE, Das A, Maity BK, Dey A, Visvakarma V, Kallianpur M, Scheidt HA, Walker G, Vaidya VA, Huster D, Maiti S. Altered Membrane Mechanics Provides a Receptor-Independent Pathway for Serotonin Action. Chemistry 2021; 27:7533-7541. [PMID: 33502812 PMCID: PMC8252079 DOI: 10.1002/chem.202100328] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Indexed: 12/20/2022]
Abstract
Serotonin, an important signaling molecule in humans, has an unexpectedly high lipid membrane affinity. The significance of this finding has evoked considerable speculation. Here we show that membrane binding by serotonin can directly modulate membrane properties and cellular function, providing an activity pathway completely independent of serotonin receptors. Atomic force microscopy shows that serotonin makes artificial lipid bilayers softer, and induces nucleation of liquid disordered domains inside the raft-like liquid-ordered domains. Solid-state NMR spectroscopy corroborates this data at the atomic level, revealing a homogeneous decrease in the order parameter of the lipid chains in the presence of serotonin. In the RN46A immortalized serotonergic neuronal cell line, extracellular serotonin enhances transferrin receptor endocytosis, even in the presence of broad-spectrum serotonin receptor and transporter inhibitors. Similarly, it increases the membrane binding and internalization of oligomeric peptides. Our results uncover a mode of serotonin-membrane interaction that can potentiate key cellular processes in a receptor-independent fashion.
Collapse
Affiliation(s)
- Simli Dey
- Department of Chemical SciencesTata Institute of Fundamental ResearchHomi Bhabha Road, ColabaMumbai400005India
| | - Dayana Surendran
- Department of Chemical SciencesTata Institute of Fundamental ResearchHomi Bhabha Road, ColabaMumbai400005India
| | - Oskar Engberg
- Institute of Medical Physics and BiophysicsUniversity of LeipzigHärtelstr. 16–1804107LeipzigGermany
| | - Ankur Gupta
- Department of Chemical SciencesTata Institute of Fundamental ResearchHomi Bhabha Road, ColabaMumbai400005India
| | - Sashaina E. Fanibunda
- Department of Biological SciencesTata Institute of Fundamental ResearchHomi Bhabha Road, ColabaMumbai400005India
- Kasturba Health SocietyMedical Research CenterMumbaiIndia
| | - Anirban Das
- Department of Chemical SciencesTata Institute of Fundamental ResearchHomi Bhabha Road, ColabaMumbai400005India
| | - Barun Kumar Maity
- Department of Chemical SciencesTata Institute of Fundamental ResearchHomi Bhabha Road, ColabaMumbai400005India
| | - Arpan Dey
- Department of Chemical SciencesTata Institute of Fundamental ResearchHomi Bhabha Road, ColabaMumbai400005India
| | - Vicky Visvakarma
- Department of Chemical SciencesTata Institute of Fundamental ResearchHomi Bhabha Road, ColabaMumbai400005India
| | - Mamata Kallianpur
- Department of Chemical SciencesTata Institute of Fundamental ResearchHomi Bhabha Road, ColabaMumbai400005India
| | - Holger A. Scheidt
- Institute of Medical Physics and BiophysicsUniversity of LeipzigHärtelstr. 16–1804107LeipzigGermany
| | - Gilbert Walker
- Department of ChemistryUniversity of TorontoTorontoOntarioM5S3H6Canada
| | - Vidita A. Vaidya
- Department of Biological SciencesTata Institute of Fundamental ResearchHomi Bhabha Road, ColabaMumbai400005India
| | - Daniel Huster
- Department of Chemical SciencesTata Institute of Fundamental ResearchHomi Bhabha Road, ColabaMumbai400005India
- Institute of Medical Physics and BiophysicsUniversity of LeipzigHärtelstr. 16–1804107LeipzigGermany
| | - Sudipta Maiti
- Department of Chemical SciencesTata Institute of Fundamental ResearchHomi Bhabha Road, ColabaMumbai400005India
| |
Collapse
|
25
|
De-Miguel FF, Leon-Pinzon C, Torres-Platas SG, Del-Pozo V, Hernández-Mendoza GA, Aguirre-Olivas D, Méndez B, Moore S, Sánchez-Sugía C, García-Aguilera MA, Martínez-Valencia A, Ramírez-Santiago G, Rubí JM. Extrasynaptic Communication. Front Mol Neurosci 2021; 14:638858. [PMID: 33994942 PMCID: PMC8119753 DOI: 10.3389/fnmol.2021.638858] [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: 12/07/2020] [Accepted: 04/01/2021] [Indexed: 12/18/2022] Open
Abstract
Streams of action potentials or long depolarizations evoke a massive exocytosis of transmitters and peptides from the surface of dendrites, axons and cell bodies of different neuron types. Such mode of exocytosis is known as extrasynaptic for occurring without utilization of synaptic structures. Most transmitters and all peptides can be released extrasynaptically. Neurons may discharge their contents with relative independence from the axon, soma and dendrites. Extrasynaptic exocytosis takes fractions of a second in varicosities or minutes in the soma or dendrites, but its effects last from seconds to hours. Unlike synaptic exocytosis, which is well localized, extrasynaptic exocytosis is diffuse and affects neuronal circuits, glia and blood vessels. Molecules that are liberated may reach extrasynaptic receptors microns away. The coupling between excitation and exocytosis follows a multistep mechanism, different from that at synapses, but similar to that for the release of hormones. The steps from excitation to exocytosis have been studied step by step for the vital transmitter serotonin in leech Retzius neurons. The events leading to serotonin exocytosis occur similarly for the release of other transmitters and peptides in central and peripheral neurons. Extrasynaptic exocytosis occurs commonly onto glial cells, which react by releasing the same or other transmitters. In the last section, we discuss how illumination of the retina evokes extrasynaptic release of dopamine and ATP. Dopamine contributes to light-adaptation; ATP activates glia, which mediates an increase in blood flow and oxygenation. A proper understanding of the workings of the nervous system requires the understanding of extrasynaptic communication.
Collapse
Affiliation(s)
- Francisco F De-Miguel
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, México City, Mexico
| | - Carolina Leon-Pinzon
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Susana G Torres-Platas
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Vanessa Del-Pozo
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | | | - Dilia Aguirre-Olivas
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Bruno Méndez
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Sharlen Moore
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Celeste Sánchez-Sugía
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | | | | | | | - J Miguel Rubí
- Facultat de Fisica, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
26
|
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
|
27
|
Vahid-Ansari F, Albert PR. Rewiring of the Serotonin System in Major Depression. Front Psychiatry 2021; 12:802581. [PMID: 34975594 PMCID: PMC8716791 DOI: 10.3389/fpsyt.2021.802581] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/17/2021] [Indexed: 12/14/2022] Open
Abstract
Serotonin is a key neurotransmitter that is implicated in a wide variety of behavioral and cognitive phenotypes. Originating in the raphe nuclei, 5-HT neurons project widely to innervate many brain regions implicated in the functions. During the development of the brain, as serotonin axons project and innervate brain regions, there is evidence that 5-HT plays key roles in wiring the developing brain, both by modulating 5-HT innervation and by influencing synaptic organization within corticolimbic structures. These actions are mediated by 14 different 5-HT receptors, with region- and cell-specific patterns of expression. More recently, the role of the 5-HT system in synaptic re-organization during adulthood has been suggested. The 5-HT neurons have the unusual capacity to regrow and reinnervate brain regions following insults such as brain injury, chronic stress, or altered development that result in disconnection of the 5-HT system and often cause depression, anxiety, and cognitive impairment. Chronic treatment with antidepressants that amplify 5-HT action, such as selective serotonin reuptake inhibitors (SSRIs), appears to accelerate the rewiring of the 5-HT system by mechanisms that may be critical to the behavioral and cognitive improvements induced in these models. In this review, we survey the possible 5-HT receptor mechanisms that could mediate 5-HT rewiring and assess the evidence that 5-HT-mediated brain rewiring is impacting recovery from mental illness. By amplifying 5-HT-induced rewiring processes using SSRIs and selective 5-HT agonists, more rapid and effective treatments for injury-induced mental illness or cognitive impairment may be achieved.
Collapse
Affiliation(s)
- Faranak Vahid-Ansari
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Paul R Albert
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| |
Collapse
|
28
|
Abstract
Precise control of monoamine neurotransmitter levels in the central nervous system (CNS) is crucial for proper brain function. Dysfunctional monoamine signaling is associated with several neuropsychiatric and neurodegenerative disorders. The plasma membrane monoamine transporter (PMAT) is a new polyspecific organic cation transporter encoded by the SLC29A4 gene. Capable of transporting monoamine neurotransmitters with low affinity and high capacity, PMAT represents a major uptake2 transporter in the brain. Broadly expressed in multiple brain regions, PMAT can complement the high-affinity, low-capacity monoamine uptake mediated by uptake1 transporters, the serotonin, dopamine, and norepinephrine transporters (SERT, DAT, and NET, respectively). This chapter provides an overview of the molecular and functional characteristics of PMAT together with its regional and cell-type specific expression in the mammalian brain. The physiological functions of PMAT in brain monoamine homeostasis are evaluated in light of its unique transport kinetics and brain location, and in comparison with uptake1 and other uptake2 transporters (e.g., OCT3) along with corroborating experimental evidences. Lastly, the possibility of PMAT's involvement in brain pathophysiological processes, such as autism, depression, and Parkinson's disease, is discussed in the context of disease pathology and potential link to aberrant monoamine pathways.
Collapse
|
29
|
Unger EK, Keller JP, Altermatt M, Liang R, Matsui A, Dong C, Hon OJ, Yao Z, Sun J, Banala S, Flanigan ME, Jaffe DA, Hartanto S, Carlen J, Mizuno GO, Borden PM, Shivange AV, Cameron LP, Sinning S, Underhill SM, Olson DE, Amara SG, Temple Lang D, Rudnick G, Marvin JS, Lavis LD, Lester HA, Alvarez VA, Fisher AJ, Prescher JA, Kash TL, Yarov-Yarovoy V, Gradinaru V, Looger LL, Tian L. Directed Evolution of a Selective and Sensitive Serotonin Sensor via Machine Learning. Cell 2020; 183:1986-2002.e26. [PMID: 33333022 PMCID: PMC8025677 DOI: 10.1016/j.cell.2020.11.040] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 06/22/2020] [Accepted: 11/20/2020] [Indexed: 12/28/2022]
Abstract
Serotonin plays a central role in cognition and is the target of most pharmaceuticals for psychiatric disorders. Existing drugs have limited efficacy; creation of improved versions will require better understanding of serotonergic circuitry, which has been hampered by our inability to monitor serotonin release and transport with high spatial and temporal resolution. We developed and applied a binding-pocket redesign strategy, guided by machine learning, to create a high-performance, soluble, fluorescent serotonin sensor (iSeroSnFR), enabling optical detection of millisecond-scale serotonin transients. We demonstrate that iSeroSnFR can be used to detect serotonin release in freely behaving mice during fear conditioning, social interaction, and sleep/wake transitions. We also developed a robust assay of serotonin transporter function and modulation by drugs. We expect that both machine-learning-guided binding-pocket redesign and iSeroSnFR will have broad utility for the development of other sensors and in vitro and in vivo serotonin detection, respectively.
Collapse
Affiliation(s)
- Elizabeth K Unger
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Jacob P Keller
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20174, USA
| | - Michael Altermatt
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ruqiang Liang
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Aya Matsui
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Chunyang Dong
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Olivia J Hon
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Zi Yao
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Junqing Sun
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Samba Banala
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20174, USA
| | - Meghan E Flanigan
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - David A Jaffe
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Samantha Hartanto
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Jane Carlen
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Grace O Mizuno
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Phillip M Borden
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20174, USA
| | - Amol V Shivange
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Lindsay P Cameron
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Steffen Sinning
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Suzanne M Underhill
- Laboratory of Molecular and Cellular Neurobiology, National Institute on Mental Health, NIH, Bethesda, MD 20892, USA
| | - David E Olson
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Susan G Amara
- Laboratory of Molecular and Cellular Neurobiology, National Institute on Mental Health, NIH, Bethesda, MD 20892, USA
| | - Duncan Temple Lang
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Gary Rudnick
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jonathan S Marvin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20174, USA
| | - Luke D Lavis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20174, USA
| | - Henry A Lester
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Andrew J Fisher
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Jennifer A Prescher
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Vladimir Yarov-Yarovoy
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Loren L Looger
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20174, USA.
| | - Lin Tian
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
30
|
Béchade C, D'Andrea I, Etienne F, Verdonk F, Moutkine I, Banas SM, Kolodziejczak M, Diaz SL, Parkhurst CN, Gan WB, Maroteaux L, Roumier A. The serotonin 2B receptor is required in neonatal microglia to limit neuroinflammation and sickness behavior in adulthood. Glia 2020; 69:638-654. [PMID: 33095507 DOI: 10.1002/glia.23918] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/11/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022]
Abstract
Severe peripheral infections induce an adaptive sickness behavior and an innate immune reaction in various organs including the brain. On the long term, persistent alteration of microglia, the brain innate immune cells, is associated with an increased risk of psychiatric disorders. It is thus critical to identify genes and mechanisms controlling the intensity and duration of the neuroinflammation induced by peripheral immune challenges. We tested the hypothesis that the 5-HT2B receptor, the main serotonin receptor expressed by microglia, might represent a valuable candidate. First, we observed that Htr2b-/- mice, knock-out for the 5-HT2B receptor gene, developed, when exposed to a peripheral lipopolysaccharide (LPS) challenge, a stronger weight loss compared to wild-type mice; in addition, comparison of inflammatory markers in brain, 4 and 24 hr after LPS injection, showed that Htr2b deficiency leads to a prolonged neuroinflammation. Second, to assess the specific contribution of the microglial 5-HT2B receptor, we investigated the response to LPS of conditional knock-out mice invalidated for Htr2b in microglia only. We found that deletion of Htr2b in microglia since birth is sufficient to cause enhanced weight loss and increased neuroinflammatory response upon LPS injection at adult stage. In contrast, mice deleted for microglial Htr2b in adulthood responded normally to LPS, revealing a neonatal developmental effect. These results highlight the role of microglia in the response to a peripheral immune challenge and suggest the existence of a developmental, neonatal period, during which instruction of microglia through 5-HT2B receptors is necessary to prevent microglia overreactivity in adulthood.
Collapse
Affiliation(s)
- Catherine Béchade
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Ivana D'Andrea
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Fanny Etienne
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Franck Verdonk
- Experimental Neuropathology, Infection and Epidemiology Department, Institut Pasteur, Paris, France
| | - Imane Moutkine
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Sophie M Banas
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Marta Kolodziejczak
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Silvina L Diaz
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Christopher N Parkhurst
- Molecular Neurobiology Program, The Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, New York, New York, USA
| | - Wenbiao B Gan
- Molecular Neurobiology Program, The Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, New York, New York, USA
| | - Luc Maroteaux
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Anne Roumier
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| |
Collapse
|
31
|
Best J, Duncan W, Sadre-Marandi F, Hashemi P, Nijhout HF, Reed M. Autoreceptor control of serotonin dynamics. BMC Neurosci 2020; 21:40. [PMID: 32967609 PMCID: PMC7509944 DOI: 10.1186/s12868-020-00587-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/29/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Serotonin is a neurotransmitter that has been linked to a wide variety of behaviors including feeding and body-weight regulation, social hierarchies, aggression and suicidality, obsessive compulsive disorder, alcoholism, anxiety, and affective disorders. Full understanding involves genomics, neurochemistry, electrophysiology, and behavior. The scientific issues are daunting but important for human health because of the use of selective serotonin reuptake inhibitors and other pharmacological agents to treat disorders. This paper presents a new deterministic model of serotonin metabolism and a new systems population model that takes into account the large variation in enzyme and transporter expression levels, tryptophan input, and autoreceptor function. RESULTS We discuss the steady state of the model and the steady state distribution of extracellular serotonin under different hypotheses on the autoreceptors and we show the effect of tryptophan input on the steady state and the effect of meals. We use the deterministic model to interpret experimental data on the responses in the hippocampus of male and female mice, and to illustrate the short-time dynamics of the autoreceptors. We show there are likely two reuptake mechanisms for serotonin and that the autoreceptors have long-lasting influence and compare our results to measurements of serotonin dynamics in the substantia nigra pars reticulata. We also show how histamine affects serotonin dynamics. We examine experimental data that show very variable response curves in populations of mice and ask how much variation in parameters in the model is necessary to produce the observed variation in the data. Finally, we show how the systems population model can potentially be used to investigate specific biological and clinical questions. CONCLUSIONS We have shown that our new models can be used to investigate the effects of tryptophan input and meals and the behavior of experimental response curves in different brain nuclei. The systems population model incorporates individual variation and can be used to investigate clinical questions and the variation in drug efficacy. The codes for both the deterministic model and the systems population model are available from the authors and can be used by other researchers to investigate the serotonergic system.
Collapse
Affiliation(s)
- Janet Best
- Department of Mathematics, The Ohio State University, 231 W 18th Ave., Columbus, OH 43210 USA
| | - William Duncan
- Department of Mathematics, Duke University, Durham, NC 27708 USA
| | | | - Parastoo Hashemi
- Department of Bioengineering, Imperial College, London, SW7 2AZ UK
| | | | - Michael Reed
- Department of Mathematics, Duke University, Durham, NC 27708 USA
| |
Collapse
|
32
|
Albertini G, Etienne F, Roumier A. Regulation of microglia by neuromodulators: Modulations in major and minor modes. Neurosci Lett 2020; 733:135000. [DOI: 10.1016/j.neulet.2020.135000] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
|
33
|
Sizemore TR, Hurley LM, Dacks AM. Serotonergic modulation across sensory modalities. J Neurophysiol 2020; 123:2406-2425. [PMID: 32401124 PMCID: PMC7311732 DOI: 10.1152/jn.00034.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/04/2020] [Accepted: 05/12/2020] [Indexed: 12/24/2022] Open
Abstract
The serotonergic system has been widely studied across animal taxa and different functional networks. This modulatory system is therefore well positioned to compare the consequences of neuromodulation for sensory processing across species and modalities at multiple levels of sensory organization. Serotonergic neurons that innervate sensory networks often bidirectionally exchange information with these networks but also receive input representative of motor events or motivational state. This convergence of information supports serotonin's capacity for contextualizing sensory information according to the animal's physiological state and external events. At the level of sensory circuitry, serotonin can have variable effects due to differential projections across specific sensory subregions, as well as differential serotonin receptor type expression within those subregions. Functionally, this infrastructure may gate or filter sensory inputs to emphasize specific stimulus features or select among different streams of information. The near-ubiquitous presence of serotonin and other neuromodulators within sensory regions, coupled with their strong effects on stimulus representation, suggests that these signaling pathways should be considered integral components of sensory systems.
Collapse
Affiliation(s)
- Tyler R Sizemore
- Department of Biology, West Virginia University, Morgantown, West Virginia
| | - Laura M Hurley
- Department of Biology, Indiana University, Bloomington, Indiana
| | - Andrew M Dacks
- Department of Biology, West Virginia University, Morgantown, West Virginia
- Department of Neuroscience, West Virginia University, Morgantown, West Virginia
| |
Collapse
|
34
|
Abstract
Neurons that synthesize and release 5-hydroxytryptamine (5-HT; serotonin) express a core set of genes that establish and maintain this neurotransmitter phenotype and distinguish these neurons from other brain cells. Beyond a shared 5-HTergic phenotype, these neurons display divergent cellular properties in relation to anatomy, morphology, hodology, electrophysiology and gene expression, including differential expression of molecules supporting co-transmission of additional neurotransmitters. This diversity suggests that functionally heterogeneous subtypes of 5-HT neurons exist, but linking subsets of these neurons to particular functions has been technically challenging. We discuss recent data from molecular genetic, genomic and functional methods that, when coupled with classical findings, yield a reframing of the 5-HT neuronal system as a conglomeration of diverse subsystems with potential to inspire novel, more targeted therapies for clinically distinct 5-HT-related disorders.
Collapse
|
35
|
Hernández-Mendoza GA, Aguirre-Olivas D, González-Gutiérrez M, Leal HJ, Qureshi N, Treviño-Palacios CG, Peón J, De-Miguel FF. Fluorescence of serotonin in the visible spectrum upon multiphotonic photoconversion. BIOMEDICAL OPTICS EXPRESS 2020; 11:1432-1448. [PMID: 32206420 PMCID: PMC7075609 DOI: 10.1364/boe.380412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/05/2019] [Accepted: 01/02/2020] [Indexed: 06/01/2023]
Abstract
The vital molecule serotonin modulates the functioning of the nervous system. The chemical characteristics of serotonin provide multiple advantages for its study in living or fixed tissue. Serotonin has the capacity to emit fluorescence directly and indirectly through chemical intermediates in response to mono- and multiphoton excitation. However, the fluorescent emissions are multifactorial and their dependence on the concentration, excitation wavelength and laser intensity still need a comprehensive study. Here we studied the fluorescence of serotonin excited multiphotonically with near-infrared light. Experiments were conducted in a custom-made multiphoton microscope coupled to a monochromator and a photomultiplier that collected the emissions. We show that the responses of serotonin to multiphoton stimulation are highly non-linear. The well-known violet emission having a 340 nm peak was accompanied by two other emissions in the visible spectrum. The best excitor wavelength to produce both emissions was 700 nm. A green emission with a ∼ 500 nm peak was similar to a previously described fluorescence in response to longer excitation wavelengths. A new blue emission with a ∼ 405 nm peak was originated from the photoconversion of serotonin to a relatively stable product. Such a reaction could be reproduced by irradiation of serotonin with high laser power for 30 minutes. The absorbance of the new compound expanded from ∼ 315 to ∼ 360 nm. Excitation of the irradiated solution monophotonically with 350 nm or biphotonically with 700 nm similarly generated the 405 nm blue emission. Our data are presented quantitatively through the design of a single geometric chart that combines the intensity of each emission in response to the serotonin concentration, excitation wavelengths and laser intensity. The autofluorescence of serotonin in addition to the formation of the two compounds emitting in the visible spectrum provides diverse possibilities for the quantitative study of the dynamics of serotonin in living tissue.
Collapse
Affiliation(s)
- Guillermo A. Hernández-Mendoza
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México. Circuito Exterior, Ciudad Universitaria, 04510, D. F., Mexico
- Instituto de Ciencias Aplicadas y Tecnología, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Ciudad de Mexico, 04510, Mexico
| | - Dilia Aguirre-Olivas
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México. Circuito Exterior, Ciudad Universitaria, 04510, D. F., Mexico
| | - Mario González-Gutiérrez
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Ciudad de Mexico, 04510, Mexico
| | - Héctor J. Leal
- Facultad de Ingeniería, Universidad Nacional Autónoma de México, Av. Universidad 3000, Ciudad Universitaria, Ciudad de Mexico, 04510, Mexico
| | - Naser Qureshi
- Instituto de Ciencias Aplicadas y Tecnología, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Ciudad de Mexico, 04510, Mexico
| | - Carlos G. Treviño-Palacios
- Instituto Nacional de Astrofísica, Óptica y Electrónica, Luis Enrique Erro #1, 72840 Tonantzintla, Puebla, Mexico
| | - Jorge Peón
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Ciudad de Mexico, 04510, Mexico
| | - Francisco F. De-Miguel
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México. Circuito Exterior, Ciudad Universitaria, 04510, D. F., Mexico
| |
Collapse
|
36
|
Josey BP, Heinrich F, Silin V, Lösche M. Association of Model Neurotransmitters with Lipid Bilayer Membranes. Biophys J 2020; 118:1044-1057. [PMID: 32032504 DOI: 10.1016/j.bpj.2020.01.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/08/2020] [Accepted: 01/15/2020] [Indexed: 11/15/2022] Open
Abstract
Aimed at reproducing the results of electrophysiological studies of synaptic signal transduction, conventional models of neurotransmission are based on the specific binding of neurotransmitters to ligand-gated receptor ion channels. However, the complex kinetic behavior observed in synaptic transmission cannot be reproduced in a standard kinetic model without the ad hoc postulation of additional conformational channel states. On the other hand, if one invokes unspecific neurotransmitter adsorption to the bilayer-a process not considered in the established models-the electrophysiological data can be rationalized with only the standard set of three conformational receptor states that also depend on this indirect coupling of neurotransmitters via their membrane interaction. Experimental verification has been difficult because binding affinities of neurotransmitters to the lipid bilayer are low. We quantify this interaction with surface plasmon resonance to measure equilibrium dissociation constants in neurotransmitter membrane association. Neutron reflection measurements on artificial membranes, so-called sparsely tethered bilayer lipid membranes, reveal the structural aspects of neurotransmitters' association with zwitterionic and anionic bilayers. We thus establish that serotonin interacts nonspecifically with the membrane at physiologically relevant concentrations, whereas γ-aminobutyric acid does not. Surface plasmon resonance shows that serotonin adsorbs with millimolar affinity, and neutron reflectometry shows that it penetrates the membrane deeply, whereas γ-aminobutyric is excluded from the bilayer.
Collapse
Affiliation(s)
- Brian P Josey
- Department of Physics, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Frank Heinrich
- Department of Physics, Carnegie Mellon University, Pittsburgh, Pennsylvania; National Institute of Standards and Technology, Center for Neutron Research, Gaithersburg, Maryland
| | - Vitalii Silin
- Institute for Bioscience and Biotechnology Research, Rockville, Maryland
| | - Mathias Lösche
- Department of Physics, Carnegie Mellon University, Pittsburgh, Pennsylvania; National Institute of Standards and Technology, Center for Neutron Research, Gaithersburg, Maryland; Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania.
| |
Collapse
|
37
|
Dyakonova TL, Sultanakhmetov GS, Mezheritskiy MI, Sakharov DA, Dyakonova VE. Storage and erasure of behavioural experiences at the single neuron level. Sci Rep 2019; 9:14733. [PMID: 31611611 PMCID: PMC6791831 DOI: 10.1038/s41598-019-51331-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/30/2019] [Indexed: 11/09/2022] Open
Abstract
Although predictions from the past about the future have been of major interest to current neuroscience, how past and present behavioral experience interacts at the level of a single neuron remains largely unknown. Using the pond snail Lymnaea stagnalis we found that recent experience of terrestrial locomotion (exercise) results in a long-term increase in the firing rate of serotonergic pedal (PeA) neurons. Isolation from the CNS preserved the "memory" about previous motor activity in the neurons even after the animals rested for two hours in deep water after the exercise. In contrast, in the CNS, no difference in the firing rate between the control and "exercise-rested" (ER) neurons was seen. ER snails, when placed again on a surface to exercise, nevertheless showed faster locomotor arousal. The difference in the firing rate between the control and ER isolated neurons disappeared when the neurons were placed in the microenvironment of their home ganglia. It is likely that an increased content of dopamine in the CNS masks an increased excitation of PeA neurons after rest: the dopamine receptor antagonist sulpiride produced sustained excitation in PeA neurons from ER snails but not in the control. Therefore, our data suggest the involvement of two mechanisms in the interplay of past and present experiences at the cellular level: intrinsic neuronal changes in the biophysical properties of the cell membrane and extrinsic modulatory environment of the ganglia.
Collapse
Affiliation(s)
- T L Dyakonova
- Koltzov Institute of Developmental Biology RAS, Vavilov St. 26, 119334, Moscow, Russia
| | - G S Sultanakhmetov
- Koltzov Institute of Developmental Biology RAS, Vavilov St. 26, 119334, Moscow, Russia
| | - M I Mezheritskiy
- Koltzov Institute of Developmental Biology RAS, Vavilov St. 26, 119334, Moscow, Russia
| | - D A Sakharov
- Koltzov Institute of Developmental Biology RAS, Vavilov St. 26, 119334, Moscow, Russia
| | - V E Dyakonova
- Koltzov Institute of Developmental Biology RAS, Vavilov St. 26, 119334, Moscow, Russia.
| |
Collapse
|
38
|
Serotonin regulates mitochondrial biogenesis and function in rodent cortical neurons via the 5-HT 2A receptor and SIRT1-PGC-1α axis. Proc Natl Acad Sci U S A 2019; 116:11028-11037. [PMID: 31072928 PMCID: PMC6561197 DOI: 10.1073/pnas.1821332116] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neuronal mitochondria are crucial organelles that regulate bioenergetics and also modulate survival and function under environmental challenges. Here, we show that the neurotransmitter serotonin (5-HT) plays an important role in the making of new mitochondria (mitochondrial biogenesis) in cortical neurons, through the 5-HT2A receptor and via master regulators of mitochondrial biogenesis, SIRT1 and PGC-1α. Mitochondrial function is also enhanced by 5-HT, increasing cellular respiration and ATP, the energy currency of the cell. We found 5-HT reduces cellular reactive oxygen species and exerts potent neuroprotective action in neurons challenged with stress, an effect that requires SIRT1. These findings highlight a role for the mitochondrial effects of 5-HT in the facilitation of stress adaptation and identify drug targets to ameliorate mitochondrial dysfunction in neurons. Mitochondria in neurons, in addition to their primary role in bioenergetics, also contribute to specialized functions, including regulation of synaptic transmission, Ca2+ homeostasis, neuronal excitability, and stress adaptation. However, the factors that influence mitochondrial biogenesis and function in neurons remain poorly elucidated. Here, we identify an important role for serotonin (5-HT) as a regulator of mitochondrial biogenesis and function in rodent cortical neurons, via a 5-HT2A receptor-mediated recruitment of the SIRT1–PGC-1α axis, which is relevant to the neuroprotective action of 5-HT. We found that 5-HT increased mitochondrial biogenesis, reflected through enhanced mtDNA levels, mitotracker staining, and expression of mitochondrial components. This resulted in higher mitochondrial respiratory capacity, oxidative phosphorylation (OXPHOS) efficiency, and a consequential increase in cellular ATP levels. Mechanistically, the effects of 5-HT were mediated via the 5-HT2A receptor and master modulators of mitochondrial biogenesis, SIRT1 and PGC-1α. SIRT1 was required to mediate the effects of 5-HT on mitochondrial biogenesis and function in cortical neurons. In vivo studies revealed that 5-HT2A receptor stimulation increased cortical mtDNA and ATP levels in a SIRT1-dependent manner. Direct infusion of 5-HT into the neocortex and chemogenetic activation of 5-HT neurons also resulted in enhanced mitochondrial biogenesis and function in vivo. In cortical neurons, 5-HT enhanced expression of antioxidant enzymes, decreased cellular reactive oxygen species, and exhibited neuroprotection against excitotoxic and oxidative stress, an effect that required SIRT1. These findings identify 5-HT as an upstream regulator of mitochondrial biogenesis and function in cortical neurons and implicate the mitochondrial effects of 5-HT in its neuroprotective action.
Collapse
|
39
|
Weese ME, Krevh RA, Li Y, Alvarez NT, Ross AE. Defect Sites Modulate Fouling Resistance on Carbon-Nanotube Fiber Electrodes. ACS Sens 2019; 4:1001-1007. [PMID: 30920207 DOI: 10.1021/acssensors.9b00161] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Carbon nanotube (CNT) fiber electrodes have become increasingly popular electrode materials for neurotransmitter detection with fast-scan cyclic voltammetry (FSCV). The unique properties of CNT fiber electrodes like increased electron transfer, sensitivity, waveform application frequency independence, and resistance to fouling make them ideal biological sensors for FSCV. In particular, their resistance to fouling has been observed for several years, but the specific physical properties which aid in fouling resistance have been debated. Here, we investigate the extent to which the presence of defect sites on the surface attenuate both chemical and biological fouling with FSCV. We compared traditional carbon-fiber microelectrodes (CFMEs) to pristine CNTs and functionalized CNTs. CFMEs and functionalized CNTs are highly disordered with a great deal of defect sites on the surface. The pristine CNTs have fewer defects compared to the purposefully functionalized CNTs and CFMEs. All electrode surfaces were characterized by a combination of scanning electron microscopy (SEM), Raman spectroscopy, and energy dispersive spectroscopy (EDS). Chemical fouling was studied using serotonin, a popular neurotransmitter notoriously known for electrode fouling. To assess biological fouling, electrodes were implanted in brain tissue for 2 h. Defect sites on the carbon were shown to resist biofouling compared to pristine CNTs but were detrimental for serotonin detection. Overall, we provide insight into the extent to which the electrode surface dictates fouling resistance with FSCV. This work provides evidence that careful considerations of the surface of the CNT material are needed when designing sensors for fouling resistance.
Collapse
Affiliation(s)
- Moriah E. Weese
- Department of Chemistry, University of Cincinnati, 404 Crosley Tower, 312 College Dr., Cincinnati, Ohio 45221-0172, United States
| | - Rachel A. Krevh
- Department of Chemistry, University of Cincinnati, 404 Crosley Tower, 312 College Dr., Cincinnati, Ohio 45221-0172, United States
| | - Yuxin Li
- Department of Chemistry, University of Cincinnati, 404 Crosley Tower, 312 College Dr., Cincinnati, Ohio 45221-0172, United States
| | - Noe T. Alvarez
- Department of Chemistry, University of Cincinnati, 404 Crosley Tower, 312 College Dr., Cincinnati, Ohio 45221-0172, United States
| | - Ashley E. Ross
- Department of Chemistry, University of Cincinnati, 404 Crosley Tower, 312 College Dr., Cincinnati, Ohio 45221-0172, United States
| |
Collapse
|
40
|
Dorsal raphe serotonin neurons inhibit operant responding for reward via inputs to the ventral tegmental area but not the nucleus accumbens: evidence from studies combining optogenetic stimulation and serotonin reuptake inhibition. Neuropsychopharmacology 2019; 44:793-804. [PMID: 30420603 PMCID: PMC6372654 DOI: 10.1038/s41386-018-0271-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/25/2018] [Accepted: 11/02/2018] [Indexed: 01/16/2023]
Abstract
The monoamine neurotransmitter serotonin (5-hydroxytryptamine; 5-HT) exerts an inhibitory influence over motivation, but the circuits mediating this are unknown. Here, we used an optogenetic approach to isolate the contribution of dorsal raphe nucleus (DRN) 5-HT neurons and 5-HT innervation of the mesolimbic dopamine (DA) system to motivated behavior in mice. We found that optogenetic stimulation of DRN 5-HT neurons enhanced downstream 5-HT release, but this was not sufficient to inhibit operant responding for saccharin, a measure of motivated behavior. However, combining optogenetic stimulation of DRN 5-HT neurons with a low dose of the selective serotonin reuptake inhibitor (SSRI) citalopram synergistically reduced operant responding. We then examined whether these effects could be recapitulated if optogenetic stimulation specifically targeted 5-HT terminals in the ventral tegmental area (VTA) or nucleus accumbens (NAc) of the mesolimbic DA system. Optogenetic stimulation of 5-HT input to the VTA combined with citalopram treatment produced a synergistic decrease in responding for saccharin, resembling the changes produced by targeting 5-HT neurons in the DRN. However, this effect was not observed when optogenetic stimulation targeted 5-HT terminals in the NAc. Taken together, these results suggest that DRN 5-HT neurons exert an inhibitory influence over operant responding for reward through a direct interaction with the mesolimbic DA system at the level of the VTA. These studies support an oppositional interaction between 5-HT and DA systems in controlling motivation and goal-directed behavior, and have important implications for the development and refinement of treatment strategies for psychiatric disorders such as depression and addiction.
Collapse
|
41
|
Shen M, Qu Z, DesLaurier J, Welle TM, Sweedler JV, Chen R. Single Synaptic Observation of Cholinergic Neurotransmission on Living Neurons: Concentration and Dynamics. J Am Chem Soc 2018; 140:7764-7768. [PMID: 29883110 DOI: 10.1021/jacs.8b01989] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Acetylcholine, the first neurotransmitter identified more than a century ago, plays critical roles in human activities and health; however, its synaptic concentration dynamics have remained unknown. Here, we demonstrate the in situ simultaneous measurements of synaptic cholinergic transmitter concentration and release dynamics. We used nanoscale electroanalytical methods: nanoITIES electrode of 15 nm in radius and nanoresolved scanning electrochemical microscopy (SECM). Time-resolved in situ measurements unveiled information on synaptic acetylcholine concentration and release dynamics of living Aplysia neurons. The measuring technique enabled the quantitative sensing of acetylcholine with negligible interference of other ionic and redox-active species. We measured cholinergic transmitter concentrations very close to the synapse, with values as high as 2.4 mM. We observed diverse synaptic transmitter concentration dynamics consisting of singlet, doublet and multiplet events with a signal-to-noise ratio of 6 to 130. The unprecedented details about synaptic neurotransmission unveiled are instrumental for understanding brain communication and diseases in a way distinctive from extra-synaptic studies.
Collapse
Affiliation(s)
- Mei Shen
- Department of Chemistry , University of Illinois at Urbana-Champaign , 600 South Matthews Avenue , Urbana , Illinois 61801 , United States
| | - Zizheng Qu
- Department of Chemistry , University of Illinois at Urbana-Champaign , 600 South Matthews Avenue , Urbana , Illinois 61801 , United States
| | - Justin DesLaurier
- Department of Chemistry , University of Illinois at Urbana-Champaign , 600 South Matthews Avenue , Urbana , Illinois 61801 , United States
| | - Theresa M Welle
- Department of Chemistry , University of Illinois at Urbana-Champaign , 600 South Matthews Avenue , Urbana , Illinois 61801 , United States
| | - Jonathan V Sweedler
- Department of Chemistry , University of Illinois at Urbana-Champaign , 600 South Matthews Avenue , Urbana , Illinois 61801 , United States
| | - Ran Chen
- Department of Chemistry , University of Illinois at Urbana-Champaign , 600 South Matthews Avenue , Urbana , Illinois 61801 , United States
| |
Collapse
|
42
|
Petersen CL, Hurley LM. Putting it in Context: Linking Auditory Processing with Social Behavior Circuits in the Vertebrate Brain. Integr Comp Biol 2018; 57:865-877. [PMID: 28985384 DOI: 10.1093/icb/icx055] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Context is critical to the adaptive value of communication. Sensory systems such as the auditory system represent an important juncture at which information on physiological state or social valence can be added to communicative information. However, the neural pathways that convey context to the auditory system are not well understood. The serotonergic system offers an excellent model to address these types of questions. Serotonin fluctuates in the mouse inferior colliculus (IC), an auditory midbrain region important for species-specific vocalizations, during specific social and non-social contexts. Furthermore, serotonin is an indicator of the valence of event-based changes within individual social interactions. We propose a model in which the brain's social behavior network serves as an afferent effector of the serotonergic dorsal raphe nucleus in order to gate contextual release of serotonin in the IC. Specifically, discrete vasopressinergic nuclei within the hypothalamus and extended amygdala that project to the dorsal raphe are functionally engaged during contexts in which serotonin fluctuates in the IC. Since serotonin strongly influences the responses of IC neurons to social vocalizations, this pathway could serve as a feedback loop whereby integrative social centers modulate their own sources of input. The end result of this feedback would be to produce a process that is geared, from sensory input to motor output, toward responding appropriately to a dynamic external world.
Collapse
Affiliation(s)
| | - Laura M Hurley
- Department of Biology, Indiana University, Bloomington, 47405 IN, USA
| |
Collapse
|
43
|
Guidolin D, Marcoli M, Maura G, Agnati LF. New dimensions of connectomics and network plasticity in the central nervous system. Rev Neurosci 2018; 28:113-132. [PMID: 28030363 DOI: 10.1515/revneuro-2016-0051] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/20/2016] [Indexed: 12/24/2022]
Abstract
Cellular network architecture plays a crucial role as the structural substrate for the brain functions. Therefore, it represents the main rationale for the emerging field of connectomics, defined as the comprehensive study of all aspects of central nervous system connectivity. Accordingly, in the present paper the main emphasis will be on the communication processes in the brain, namely wiring transmission (WT), i.e. the mapping of the communication channels made by cell components such as axons and synapses, and volume transmission (VT), i.e. the chemical signal diffusion along the interstitial brain fluid pathways. Considering both processes can further expand the connectomics concept, since both WT-connectomics and VT-connectomics contribute to the structure of the brain connectome. A consensus exists that such a structure follows a hierarchical or nested architecture, and macro-, meso- and microscales have been defined. In this respect, however, several lines of evidence indicate that a nanoscale (nano-connectomics) should also be considered to capture direct protein-protein allosteric interactions such as those occurring, for example, in receptor-receptor interactions at the plasma membrane level. In addition, emerging evidence points to novel mechanisms likely playing a significant role in the modulation of intercellular connectivity, increasing the plasticity of the system and adding complexity to its structure. In particular, the roamer type of VT (i.e. the intercellular transfer of RNA, proteins and receptors by extracellular vesicles) will be discussed since it allowed us to introduce a new concept of 'transient changes of cell phenotype', that is the transient acquisition of new signal release capabilities and/or new recognition/decoding apparatuses.
Collapse
|
44
|
Rojas P, Aguayo F, Neira D, Tejos M, Aliaga E, Muñoz J, Parra C, Fiedler J. Dual effect of serotonin on the dendritic growth of cultured hippocampal neurons: Involvement of 5-HT1A and 5-HT7 receptors. Mol Cell Neurosci 2017; 85:148-161. [DOI: 10.1016/j.mcn.2017.09.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 09/04/2017] [Accepted: 09/29/2017] [Indexed: 01/11/2023] Open
|
45
|
Domínguez-Soto Á, Usategui A, Casas-Engel MDL, Simón-Fuentes M, Nieto C, Cuevas VD, Vega MA, Luis Pablos J, Corbí ÁL. Serotonin drives the acquisition of a profibrotic and anti-inflammatory gene profile through the 5-HT7R-PKA signaling axis. Sci Rep 2017; 7:14761. [PMID: 29116249 PMCID: PMC5676747 DOI: 10.1038/s41598-017-15348-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 10/23/2017] [Indexed: 12/19/2022] Open
Abstract
Peripheral serotonin (5-hydroxytryptamine, 5-HT) regulates cell growth and differentiation in numerous cell types through engagement of seven types of cell surface receptors (HTR1-7). Deregulated 5-HT/HTR levels contribute to pathology in chronic inflammatory diseases, with macrophages being relevant targets for the physio-pathological effects of 5-HT. In fact, 5-HT skews human macrophage polarization through engagement of 5-HT2BR and 5-HT7R receptors. We now report that 5-HT primes macrophages for reduced pro-inflammatory cytokine production and IFN type I-mediated signaling, and promotes an anti-inflammatory and pro-fibrotic gene signature in human macrophages. The acquisition of the 5-HT-dependent gene profile primarily depends on the 5-HT7R receptor and 5-HT7R-initiated PKA-dependent signaling. In line with the transcriptional results, 5-HT upregulates TGFβ1 production by human macrophages in an HTR7- and PKA-dependent manner, whereas the absence of Htr7 in vivo results in diminished macrophage infiltration and collagen deposition in a mouse model of skin fibrosis. Our results indicate that the anti-inflammatory and pro-fibrotic activity of 5-HT is primarily mediated through the 5-HT7R-PKA axis, and that 5-HT7R contributes to pathology in fibrotic diseases.
Collapse
Affiliation(s)
| | - Alicia Usategui
- Servicio de Reumatología, Instituto de Investigación Hospital 12 de octubre, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Miriam Simón-Fuentes
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Concha Nieto
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Víctor D Cuevas
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Miguel A Vega
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - José Luis Pablos
- Servicio de Reumatología, Instituto de Investigación Hospital 12 de octubre, Universidad Complutense de Madrid, Madrid, Spain
| | - Ángel L Corbí
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| |
Collapse
|
46
|
Manciu FS, Ciubuc JD, Sundin EM, Qiu C, Bennet KE. Analysis of Serotonin Molecules on Silver Nanocolloids-A Raman Computational and Experimental Study. SENSORS 2017. [PMID: 28640186 PMCID: PMC5539499 DOI: 10.3390/s17071471] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Combined theoretical and experimental analysis of serotonin by quantum chemical density functional calculations and surface-enhanced Raman spectroscopy, respectively, is presented in this work to better understand phenomena related to this neurotransmitter’s detection and monitoring at very low concentrations specific to physiological levels. In addition to the successful ultrasensitive analyte detection on silver nanoparticles for concentrations as low as 10−11 molar, the relatively good agreement between the simulated and experimentally determined results indicates the presence of all serotonin molecular forms, such as neutral, ionic, and those oxidized through redox reactions. Obvious structural molecular deformations such as bending of lateral amino chains are observed for both ionic and oxidized forms. Not only does this combined approach reveal more probable adsorption of serotonin into the silver surface through hydroxyl/oxygen sites than through NH/nitrogen sites, but also that it does so predominantly in its neutral (reduced) form, somewhat less so in its ionic forms, and much less in its oxidized forms. If the development of opto-voltammetric biosensors and their effective implementation is envisioned for the future, this study provides some needed scientific background for comprehending changes in the vibrational signatures of this important neurotransmitter.
Collapse
Affiliation(s)
- Felicia S Manciu
- Department of Physics, University of Texas at El Paso, El Paso, TX 79968, USA.
- Department of Biomedical Engineering, University of Texas at El Paso, El Paso, TX 79968, USA.
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA.
| | - John D Ciubuc
- Department of Physics, University of Texas at El Paso, El Paso, TX 79968, USA.
- Department of Biomedical Engineering, University of Texas at El Paso, El Paso, TX 79968, USA.
| | - Emma M Sundin
- Department of Physics, University of Texas at El Paso, El Paso, TX 79968, USA.
| | - Chao Qiu
- Department of Physics, University of Texas at El Paso, El Paso, TX 79968, USA.
| | - Kevin E Bennet
- Division of Engineering, Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
47
|
Tao R, Shokry IM, Callanan JJ. Environment Influencing Serotonin Syndrome Induced by Ecstasy Abuse. ANNALS OF FORENSIC RESEARCH AND ANALYSIS 2017; 4:1039. [PMID: 29732414 PMCID: PMC5931730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Ecstasy is a recreational drug containing 3,4-methylenedioxymethamphetamine (MDMA). In the U.S., there are several millions of lifetime users, and millions each year added to the list as new users. Only several thousand every year show signs of severe toxicity and require emergency intervention. The illness is known as serotonin (5-HT) syndrome, which can be mild, moderate or severe. The relationship between mild, moderate and severe syndromes appears to be interchangeable, but the severe syndrome is life-threatening. The serotonergic mechanisms of how the mild or moderate syndrome becomes severe and life-threatening have attracted considerable attention in the last few years as an effort to explore new treatments potentially to manage illness and prevent death of patients. High levels of extracellular 5-HT in the brain produced by large doses of MDMA are not always necessary to cause a severe serotonin syndrome. Additional mechanisms may be more important. Recent research has demonstrated that environmental conditions (i.e., non-drug factors) are more critical in determining the severity of MDMA-induced serotonin syndrome than the drug dose. The purpose of the current article was to review available evidence regarding the effect of non-drug factors on serotonergic extrasynaptic receptor responsivity and the severity of MDMA-induced serotonin syndrome.
Collapse
Affiliation(s)
- Rui Tao
- Charles E. Schmidt College of Medicine, Florida Atlantic University, USA
| | - Ibrahim M. Shokry
- Charles E. Schmidt College of Medicine, Florida Atlantic University, USA
- Ross University School of Veterinary Medicine, West Indies
| | | |
Collapse
|
48
|
SSRI Facilitated Crack Dancing. Case Rep Neurol Med 2017; 2017:4318450. [PMID: 28487792 PMCID: PMC5405355 DOI: 10.1155/2017/4318450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 03/31/2017] [Accepted: 04/06/2017] [Indexed: 11/18/2022] Open
Abstract
Choreoathetoid movement secondary to cocaine use is a well-documented phenomenon better known as “crack dancing.” It consists of uncontrolled writhing movements secondary to excess dopamine from cocaine use. We present a 32-year-old male who had been using cocaine for many years and was recently started on paroxetine, a selective serotonin reuptake inhibitor (SSRI) for worsening depression four weeks before presentation. He had been doing cocaine every 2 weeks for the last three years and had never “crack danced” before this episode. The authors have conducted a thorough literature review and cited studies that suggest “crack dancing” is associated with excess dopamine. There has never been a documented case report of an SSRI being linked with “crack dancing.” The authors propose that the excess dopaminergic effect of the SSRI lowered the dopamine threshold for “crack dancing.” There is a communication with the Raphe Nucleus and the Substantia Nigra, which explains how the SSRI increases dopamine levels. This is the first documented case of an SSRI facilitating the “crack dance.”
Collapse
|
49
|
Hai A, Cai LX, Lee T, Lelyveld VS, Jasanoff A. Molecular fMRI of Serotonin Transport. Neuron 2016; 92:754-765. [PMID: 27773583 DOI: 10.1016/j.neuron.2016.09.048] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 06/29/2016] [Accepted: 09/20/2016] [Indexed: 01/03/2023]
Abstract
Reuptake of neurotransmitters from the brain interstitium shapes chemical signaling processes and is disrupted in several pathologies. Serotonin reuptake in particular is important for mood regulation and is inhibited by first-line drugs for treatment of depression. Here we introduce a molecular-level fMRI technique for micron-scale mapping of serotonin transport in live animals. Intracranial injection of an MRI-detectable serotonin sensor complexed with serotonin, together with serial imaging and compartmental analysis, permits neurotransmitter transport to be quantified as serotonin dissociates from the probe. Application of this strategy to much of the striatum and surrounding areas reveals widespread nonsaturating serotonin removal with maximal rates in the lateral septum. The serotonin reuptake inhibitor fluoxetine selectively suppresses serotonin removal in septal subregions, whereas both fluoxetine and a dopamine transporter blocker depress reuptake in striatum. These results highlight promiscuous pharmacological influences on the serotonergic system and demonstrate the utility of molecular fMRI for characterization of neurochemical dynamics.
Collapse
Affiliation(s)
- Aviad Hai
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Lili X Cai
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Taekwan Lee
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Victor S Lelyveld
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Alan Jasanoff
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Department of Nuclear Science & Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
50
|
Monoaminergic control of brain states and sensory processing: Existing knowledge and recent insights obtained with optogenetics. Prog Neurobiol 2016; 151:237-253. [PMID: 27634227 DOI: 10.1016/j.pneurobio.2016.09.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/18/2016] [Accepted: 09/10/2016] [Indexed: 01/18/2023]
Abstract
Monoamines are key neuromodulators involved in a variety of physiological and pathological brain functions. Classical studies using physiological and pharmacological tools have revealed several essential aspects of monoaminergic involvement in regulating the sleep-wake cycle and influencing sensory responses but many features have remained elusive due to technical limitations. The application of optogenetic tools led to the ability of monitoring and controlling neuronal populations with unprecedented temporal precision and neurochemical specificity. Here, we focus on recent advances in revealing the roles of some monoamines in brain state control and sensory information processing. We summarize the central position of monoamines in integrating sensory processing across sleep-wake states with an emphasis on research conducted using optogenetic techniques. Finally, we discuss the limitations and perspectives of new integrated experimental approaches in understanding the modulatory mechanisms of monoaminergic systems in the mammalian brain.
Collapse
|