1
|
Wu Z, Shen Z, Xu Y, Chen S, Xiao S, Ye J, Zhang H, Ma X, Zhu Y, Zhu X, Jiang Y, Fang J, Liu B, He X, Gao S, Shao X, Liu J, Fang J. A neural circuit associated with anxiety-like behaviors induced by chronic inflammatory pain and the anxiolytic effects of electroacupuncture. CNS Neurosci Ther 2024; 30:e14520. [PMID: 38018559 PMCID: PMC11017463 DOI: 10.1111/cns.14520] [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: 07/21/2023] [Revised: 10/14/2023] [Accepted: 10/22/2023] [Indexed: 11/30/2023] Open
Abstract
AIMS Negative emotions induced by chronic pain are a serious clinical problem. Electroacupuncture (EA) is a clinically proven safe and effective method to manage pain-related negative emotions. However, the circuit mechanisms underlying the effect of EA treatment on negative emotions remain unclear. METHODS Plantar injection of complete Freund's adjuvant (CFA) was performed to establish a rat model of chronic inflammatory pain-induced anxiety-like behaviors. Adeno-associated virus (AAV) tracing was used to identify excitatory synaptic transmission from the rostral anterior cingulate cortex (rACC) to the dorsal raphe nucleus (DRN). Employing chemogenetic approaches, we examined the role of the rACC-DRN circuit in chronic pain-induced anxiety-like behaviors and investigated whether EA could reverse chronic pain-induced dysfunctions of the rACC-DRN circuit and anxiety-like behaviors. RESULTS We found that chemogenetic activation of the rACC-DRN circuit alleviated CFA-induced anxiety-like behaviors, while chemogenetic inhibition of the rACC-DRN circuit resulted in short-term CFA-induced anxiety-like behaviors. Further research revealed that the development of CFA-induced anxiety-like behaviors was attributed to the dysfunction of rACC CaMKII neurons projecting to DRN serotonergic neurons (rACCCaMKII-DRN5-HT neurons) but not rACC CaMKII neurons projecting to DRN GABAergic neurons (rACCCaMKII-DRNGABA neurons). This is supported by the findings that chemogenetic activation of the rACCCaMKII-DRN5-HT circuit alleviates anxiety-like behaviors in rats with chronic pain, whereas neither chemogenetic inhibition nor chemogenetic activation of the rACCCaMKII-DRNGABA circuit altered CFA chronic pain-evoked anxiety-like behaviors in rats. More importantly, we found that EA could reverse chronic pain-induced changes in the activity of rACC CaMKII neurons and DRN 5-HTergic neurons and that chemogenetic inhibition of the rACCCaMKII-DRN5-HT circuit blocked the therapeutic effects of EA on chronic pain-induced anxiety-like behaviors. CONCLUSIONS Our data suggest that the reversal of rACCCaMKII-DRN5-HT circuit dysfunction may be a mechanism underlying the therapeutic effect of EA on chronic pain-induced anxiety-like behaviors.
Collapse
Affiliation(s)
- Zemin Wu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
- Department of Acupuncture and Moxibustionthe First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Zui Shen
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yingling Xu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
| | - Shaozong Chen
- Institution of Acupuncture and Moxibustion, Shandong University of Traditional Chinese MedicineJinanChina
| | - Siqi Xiao
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Jiayu Ye
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Haiyan Zhang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Xinyi Ma
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yichen Zhu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Xixiao Zhu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yongliang Jiang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Junfan Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Boyi Liu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Xiaofen He
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Shuzhong Gao
- Institution of Acupuncture and Moxibustion, Shandong University of Traditional Chinese MedicineJinanChina
| | - Xiaomei Shao
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Jinggen Liu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
- National Key Laboratory of Drug ResearchShanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Jianqiao Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
- Department of Acupuncture and Moxibustionthe First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| |
Collapse
|
2
|
Siletti K, Hodge R, Mossi Albiach A, Lee KW, Ding SL, Hu L, Lönnerberg P, Bakken T, Casper T, Clark M, Dee N, Gloe J, Hirschstein D, Shapovalova NV, Keene CD, Nyhus J, Tung H, Yanny AM, Arenas E, Lein ES, Linnarsson S. Transcriptomic diversity of cell types across the adult human brain. Science 2023; 382:eadd7046. [PMID: 37824663 DOI: 10.1126/science.add7046] [Citation(s) in RCA: 132] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 09/07/2023] [Indexed: 10/14/2023]
Abstract
The human brain directs complex behaviors, ranging from fine motor skills to abstract intelligence, but the diversity of cell types that support these skills has not been fully described. In this work, we used single-nucleus RNA sequencing to systematically survey cells across the entire adult human brain. We sampled more than three million nuclei from approximately 100 dissections across the forebrain, midbrain, and hindbrain in three postmortem donors. Our analysis identified 461 clusters and 3313 subclusters organized largely according to developmental origins and revealing high diversity in midbrain and hindbrain neurons. Astrocytes and oligodendrocyte-lineage cells also exhibited regional diversity at multiple scales. The transcriptomic census of the entire human brain presented in this work provides a resource for understanding the molecular diversity of the human brain in health and disease.
Collapse
Affiliation(s)
| | - Rebecca Hodge
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Ka Wai Lee
- Karolinska Institute, 171 77 Stockholm, Sweden
| | - Song-Lin Ding
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Lijuan Hu
- Karolinska Institute, 171 77 Stockholm, Sweden
| | | | - Trygve Bakken
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Tamara Casper
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Michael Clark
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Julie Nyhus
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Herman Tung
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | |
Collapse
|
3
|
Zhang XL, Spencer WC, Tabuchi N, Kitt MM, Deneris ES. Reorganization of postmitotic neuronal chromatin accessibility for maturation of serotonergic identity. eLife 2022; 11:e75970. [PMID: 35471146 PMCID: PMC9098219 DOI: 10.7554/elife.75970] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/12/2022] [Indexed: 12/02/2022] Open
Abstract
Assembly of transcriptomes encoding unique neuronal identities requires selective accessibility of transcription factors to cis-regulatory sequences in nucleosome-embedded postmitotic chromatin. Yet, the mechanisms controlling postmitotic neuronal chromatin accessibility are poorly understood. Here, we show that unique distal enhancers define the Pet1 neuron lineage that generates serotonin (5-HT) neurons in mice. Heterogeneous single-cell chromatin landscapes are established early in postmitotic Pet1 neurons and reveal the putative regulatory programs driving Pet1 neuron subtype identities. Distal enhancer accessibility is highly dynamic as Pet1 neurons mature, suggesting the existence of regulatory factors that reorganize postmitotic neuronal chromatin. We find that Pet1 and Lmx1b control chromatin accessibility to select Pet1-lineage-specific enhancers for 5-HT neurotransmission. Additionally, these factors are required to maintain chromatin accessibility during early maturation suggesting that postmitotic neuronal open chromatin is unstable and requires continuous regulatory input. Together, our findings reveal postmitotic transcription factors that reorganize accessible chromatin for neuron specialization.
Collapse
Affiliation(s)
- Xinrui L Zhang
- Department of Neurosciences, Case Western Reserve UniversityClevelandUnited States
| | - William C Spencer
- Department of Neurosciences, Case Western Reserve UniversityClevelandUnited States
| | - Nobuko Tabuchi
- Department of Neurosciences, Case Western Reserve UniversityClevelandUnited States
| | - Meagan M Kitt
- Department of Neurosciences, Case Western Reserve UniversityClevelandUnited States
| | - Evan S Deneris
- Department of Neurosciences, Case Western Reserve UniversityClevelandUnited States
| |
Collapse
|
4
|
Kappa Opioid Receptor Mediated Differential Regulation of Serotonin and Dopamine Transporters in Mood and Substance Use Disorder. Handb Exp Pharmacol 2021; 271:97-112. [PMID: 34136961 DOI: 10.1007/164_2021_499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Dynorphin (DYN) is an endogenous neurosecretory peptide which exerts its activity by binding to the family of G protein-coupled receptors, namely the kappa opioid receptor (KOR). Opioids are associated with pain, analgesia, and drug abuse, which play a central role in mood disorders with monoamine neurotransmitter interactions. Growing evidence demonstrates the cellular signaling cascades linked to KOR-mediated monoamine transporters regulation in cell models and native brain tissues. This chapter will review DYN/KOR role in mood and addiction in relevance to dopaminergic and serotonergic neurotransmissions. Also, we discuss the recent findings on KOR-mediated differential regulation of serotonin and dopamine transporters (SERT and DAT). These findings led to a better understanding of the role of DYN/KOR system in aminergic neurotransmission via its modulatory effect on both amine release and clearance. Detailed knowledge of these processes at the molecular level enables designing novel pharmacological reagents to target transporter motifs to treat mood and addiction and reduce unwanted side effects such as aversion, dysphoria, sedation, and psychomimesis.
Collapse
|
5
|
Shin HY, Han KS, Park HW, Hong YH, Kim Y, Moon HE, Park KW, Park HR, Lee CJ, Lee K, Kim SJ, Heo MS, Park SH, Kim DG, Paek SH. Tumor Spheroids of an Aggressive Form of Central Neurocytoma Have Transit-Amplifying Progenitor Characteristics with Enhanced EGFR and Tumor Stem Cell Signaling. Exp Neurobiol 2021; 30:120-143. [PMID: 33972466 PMCID: PMC8118755 DOI: 10.5607/en21004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 11/19/2022] Open
Abstract
Central neurocytoma (CN) has been known as a benign neuronal tumor. In rare cases, CN undergoes malignant transformation to glioblastomas (GBM). Here we examined its cellular origin by characterizing differentiation potential and gene expression of CN-spheroids. First, we demonstrate that both CN tissue and cultured primary cells recapitulate the hierarchal cellular composition of subventricular zone (SVZ), which is comprised of neural stem cells (NSCs), transit amplifying progenitors (TAPs), and neuroblasts. We then derived spheroids from CN which displayed EGFR+/MASH+ TAP and BLBP+ radial glial cell (RGC) characteristic, and mitotic neurogenesis and gliogenesis by single spheroids were observed with cycling multipotential cells. CN-spheroids expressed increased levels of pluripotency and tumor stem cell genes such as KLF4 and TPD5L1, when compared to their differentiated cells and human NSCs. Importantly, Gene Set Enrichment Analysis showed that gene sets of GBM-Spheroids, EGFR Signaling, and Packaging of Telomere Ends are enriched in CN-spheroids in comparison with their differentiated cells. We speculate that CN tumor stem cells have TAP and RGC characteristics, and upregulation of EGFR signaling as well as downregulation of eph-ephrin signaling have critical roles in tumorigenesis of CN. And their ephemeral nature of TAPs destined to neuroblasts, might reflect benign nature of CN.
Collapse
Affiliation(s)
- Hye Young Shin
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Kyung-Seok Han
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Hyung Woo Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Yun Hwa Hong
- Department of Neurophysiology, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Yona Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Hyo Eun Moon
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Kwang Woo Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Hye Ran Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Kiyoung Lee
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Sang Jeong Kim
- Department of Neurophysiology, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Man Seung Heo
- Smart Healthcare Medical Device Research Center, Samsung Medical Center, Seoul 06351, Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Dong Gyu Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Sun Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea.,Ischemic/Hypoxic Disease Institute, Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea.,Clinical Research Institute, Seoul National University Hospital, Seoul 03082, Korea
| |
Collapse
|
6
|
STAT3 in the dorsal raphe gates behavioural reactivity and regulates gene networks associated with psychopathology. Mol Psychiatry 2021; 26:2886-2899. [PMID: 33046834 PMCID: PMC8505245 DOI: 10.1038/s41380-020-00904-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 01/02/2023]
Abstract
The signal transducer and activator of transcription 3 (STAT3) signalling pathway is activated through phosphorylation by Janus kinases in response to a diverse set of immunogenic and non-immunogenic triggers. Several distinct lines of evidence propose an intricate involvement of STAT3 in neural function relevant to behaviour in health and disease. However, in part due to the pleiotropic effects resulting from its DNA binding activity and the consequent regulation of expression of a variety of genes with context-dependent cellular consequences, the precise nature of STAT3 involvement in the neural mechanisms underlying psychopathology remains incompletely understood. Here, we focused on the midbrain serotonergic system, a central hub for the regulation of emotions, to examine the relevance of STAT3 signalling for emotional behaviour in mice by selectively knocking down raphe STAT3 expression using germline genetic (STAT3 KO) and viral-mediated approaches. Mice lacking serotonergic STAT3 presented with reduced negative behavioural reactivity and a blunted response to the sensitising effects of amphetamine, alongside alterations in midbrain neuronal firing activity of serotonergic neurons and transcriptional control of gene networks relevant for neuropsychiatric disorders. Viral knockdown of dorsal raphe (DR) STAT3 phenocopied the behavioural alterations of STAT3 KO mice, excluding a developmentally determined effect and suggesting that disruption of STAT3 signalling in the DR of adult mice is sufficient for the manifestation of behavioural traits relevant to psychopathology. Collectively, these results suggest DR STAT3 as a molecular gate for the control of behavioural reactivity, constituting a mechanistic link between the upstream activators of STAT3, serotonergic neurotransmission and psychopathology.
Collapse
|
7
|
Black JB, McCutcheon SR, Dube S, Barrera A, Klann TS, Rice GA, Adkar SS, Soderling SH, Reddy TE, Gersbach CA. Master Regulators and Cofactors of Human Neuronal Cell Fate Specification Identified by CRISPR Gene Activation Screens. Cell Rep 2020; 33:108460. [PMID: 33264623 PMCID: PMC7730023 DOI: 10.1016/j.celrep.2020.108460] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 08/02/2020] [Accepted: 11/09/2020] [Indexed: 01/06/2023] Open
Abstract
Technologies to reprogram cell-type specification have revolutionized the fields of regenerative medicine and disease modeling. Currently, the selection of fate-determining factors for cell reprogramming applications is typically a laborious and low-throughput process. Therefore, we use high-throughput pooled CRISPR activation (CRISPRa) screens to systematically map human neuronal cell fate regulators. We utilize deactivated Cas9 (dCas9)-based gene activation to target 1,496 putative transcription factors (TFs) in the human genome. Using a reporter of neuronal commitment, we profile the neurogenic activity of these factors in human pluripotent stem cells (PSCs), leading to a curated set of pro-neuronal factors. Activation of pairs of TFs reveals neuronal cofactors, including E2F7, RUNX3, and LHX8, that improve conversion efficiency, subtype specificity, and maturation of neuronal cell types. Finally, using multiplexed gene regulation with orthogonal CRISPR systems, we demonstrate improved neuronal differentiation with concurrent activation and repression of target genes, underscoring the power of CRISPR-based gene regulation for programming complex cellular phenotypes.
Collapse
Affiliation(s)
- Joshua B Black
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Sean R McCutcheon
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Shataakshi Dube
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Alejandro Barrera
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA; Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC 27710, USA
| | - Tyler S Klann
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Grayson A Rice
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Shaunak S Adkar
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Scott H Soderling
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Timothy E Reddy
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA; Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC 27710, USA; Graduate Program in Computational Biology and Bioinformatics, Duke University, Durham, NC 27708, USA; University Program in Genetics and Genomics, Duke University, Durham, NC 27708, USA; Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708, USA
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Graduate Program in Computational Biology and Bioinformatics, Duke University, Durham, NC 27708, USA; University Program in Genetics and Genomics, Duke University, Durham, NC 27708, USA; Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
8
|
Baskin BM, Mai JJ, Dymecki SM, Kantak KM. Cocaine reward and memory after chemogenetic inhibition of distinct serotonin neuron subtypes in mice. Psychopharmacology (Berl) 2020; 237:2633-2648. [PMID: 32494973 PMCID: PMC8057056 DOI: 10.1007/s00213-020-05560-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/18/2020] [Indexed: 01/23/2023]
Abstract
RATIONALE We probed serotonin neurons, those denoted by their developmental gene expression as r2Hoxa2-Pet1 (experiment 1) and Drd1a-Pet1 (experiment 2), for differential modulation of cocaine reward and memory as revealed by the expression and development of conditioned place preference (CPP) in transgenic mice. OBJECTIVES To query roles in CPP, we inhibited neurons cell autonomously in vivo by activating the transgenically expressed, synthetic DREADD receptor hM4Di (Di) with the exogenous ligand clozapine-N-oxide (CNO). METHODS To examine CPP expression, mice were conditioned using behaviorally active doses of cocaine (10.0 or 17.8 mg/kg) vs. saline followed by CPP assessment, first without neuron inhibition (post-conditioning session 1), and then with CNO-mediated neuron inhibition (post-conditioning session 2), followed by 4 more post-conditioning sessions. To examine CPP development, we administered CNO during conditioning sessions and then assayed CPP across 6 post-conditioning sessions. RESULTS In r2Hoxa2-Pet1-Di mice, post-conditioning CNO administration did not impact cocaine CPP expression, but after CNO administration during conditioning, cocaine CPP (17.8 mg/kg) persisted across post-conditioning sessions compared with that in controls, suggesting a deficit in extinguishing cocaine memory. Drd1a-Pet1-Di mice, prior to CNO-Di-triggered neuronal inhibition, unexpectedly expressed heightened cocaine CPP (10.0 and 17.8 mg/kg) compared with controls, and this basal phenotype was transiently blocked by acute post-conditioning CNO administration and persistently blocked by repeated CNO administration during conditioning. CONCLUSION Cocaine reward and memory likely map to distinct serotonergic Pet1 neuron subtypes. r2Hoxa2-Pet1 neurons normally may limit the durability of cocaine memory, without impacting initial cocaine reward magnitude. Drd1a-Pet1 neurons normally may help to promote cocaine reward.
Collapse
Affiliation(s)
- Britahny M. Baskin
- Department of Psychological and Brain Sciences, Boston University, 64 Cummington Mall, Boston, MA 02215, USA
| | - Jia Jia Mai
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, USA
| | - Susan M. Dymecki
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, USA
| | - Kathleen M. Kantak
- Department of Psychological and Brain Sciences, Boston University, 64 Cummington Mall, Boston, MA 02215, USA
| |
Collapse
|
9
|
Coates KE, Calle-Schuler SA, Helmick LM, Knotts VL, Martik BN, Salman F, Warner LT, Valla SV, Bock DD, Dacks AM. The Wiring Logic of an Identified Serotonergic Neuron That Spans Sensory Networks. J Neurosci 2020; 40:6309-6327. [PMID: 32641403 PMCID: PMC7424878 DOI: 10.1523/jneurosci.0552-20.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/16/2020] [Accepted: 06/25/2020] [Indexed: 12/21/2022] Open
Abstract
Serotonergic neurons project widely throughout the brain to modulate diverse physiological and behavioral processes. However, a single-cell resolution understanding of the connectivity of serotonergic neurons is currently lacking. Using a whole-brain EM dataset of a female Drosophila, we comprehensively determine the wiring logic of a broadly projecting serotonergic neuron (the CSDn) that spans several olfactory regions. Within the antennal lobe, the CSDn differentially innervates each glomerulus, yet surprisingly, this variability reflects a diverse set of presynaptic partners, rather than glomerulus-specific differences in synaptic output, which is predominately to local interneurons. Moreover, the CSDn has distinct connectivity relationships with specific local interneuron subtypes, suggesting that the CSDn influences distinct aspects of local network processing. Across olfactory regions, the CSDn has different patterns of connectivity, even having different connectivity with individual projection neurons that also span these regions. Whereas the CSDn targets inhibitory local neurons in the antennal lobe, the CSDn has more distributed connectivity in the LH, preferentially synapsing with principal neuron types based on transmitter content. Last, we identify individual novel synaptic partners associated with other sensory domains that provide strong, top-down input to the CSDn. Together, our study reveals the complex connectivity of serotonergic neurons, which combine the integration of local and extrinsic synaptic input in a nuanced, region-specific manner.SIGNIFICANCE STATEMENT All sensory systems receive serotonergic modulatory input. However, a comprehensive understanding of the synaptic connectivity of individual serotonergic neurons is lacking. In this study, we use a whole-brain EM microscopy dataset to comprehensively determine the wiring logic of a broadly projecting serotonergic neuron in the olfactory system of Drosophila Collectively, our study demonstrates, at a single-cell level, the complex connectivity of serotonergic neurons within their target networks, identifies specific cell classes heavily targeted for serotonergic modulation in the olfactory system, and reveals novel extrinsic neurons that provide strong input to this serotonergic system outside of the context of olfaction. Elucidating the connectivity logic of individual modulatory neurons provides a ground plan for the seemingly heterogeneous effects of modulatory systems.
Collapse
Affiliation(s)
- Kaylynn E Coates
- Department of Biology, West Virginia University, Morgantown, West Virginia 26506
| | | | - Levi M Helmick
- Department of Biology, West Virginia University, Morgantown, West Virginia 26506
| | - Victoria L Knotts
- Department of Biology, West Virginia University, Morgantown, West Virginia 26506
| | - Brennah N Martik
- Department of Biology, West Virginia University, Morgantown, West Virginia 26506
| | - Farzaan Salman
- Department of Biology, West Virginia University, Morgantown, West Virginia 26506
| | - Lauren T Warner
- Department of Biology, West Virginia University, Morgantown, West Virginia 26506
| | - Sophia V Valla
- Department of Biology, West Virginia University, Morgantown, West Virginia 26506
| | - Davi D Bock
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia 20147
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405
| | - Andrew M Dacks
- Department of Biology, West Virginia University, Morgantown, West Virginia 26506
- Department of Neuroscience, West Virginia University, Morgantown, West Virginia 26506
| |
Collapse
|
10
|
Kinney HC, Haynes RL. The Serotonin Brainstem Hypothesis for the Sudden Infant Death Syndrome. J Neuropathol Exp Neurol 2020; 78:765-779. [PMID: 31397480 DOI: 10.1093/jnen/nlz062] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/28/2019] [Accepted: 06/25/2019] [Indexed: 01/04/2023] Open
Abstract
The sudden infant death syndrome (SIDS) is the leading cause of postneonatal infant mortality in the United States today, with an overall rate of 0.39/1000 live births. It is defined as the sudden and unexpected death of an infant <12 months of age that remains unexplained after a complete autopsy, death scene investigation, and review of the clinical history. The serotonin brainstem hypothesis has been a leading hypothesis for SIDS over the last 2 decades. Our laboratory has studied this hypothesis over time with a variety of tissue techniques, including tissue receptor autoradiography, high performance liquid chromatography, Western blot analysis, immunocytochemistry, and proteomics. The purpose of this article is to review the progress in our laboratory toward supporting this hypothesis. We conclude that an important subset of SIDS infants has serotonergic abnormalities resulting from a "core lesion" in the medullary reticular formation comprised of nuclei that contain serotonin neurons. This lesion could lead to a failure of protective brainstem responses to homeostatic challenges during sleep in a critical developmental period which cause sleep-related sudden death.
Collapse
Affiliation(s)
- Hannah C Kinney
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Robin L Haynes
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
11
|
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
|
12
|
Ren J, Isakova A, Friedmann D, Zeng J, Grutzner SM, Pun A, Zhao GQ, Kolluru SS, Wang R, Lin R, Li P, Li A, Raymond JL, Luo Q, Luo M, Quake SR, Luo L. Single-cell transcriptomes and whole-brain projections of serotonin neurons in the mouse dorsal and median raphe nuclei. eLife 2019; 8:e49424. [PMID: 31647409 PMCID: PMC6812963 DOI: 10.7554/elife.49424] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/12/2019] [Indexed: 12/11/2022] Open
Abstract
Serotonin neurons of the dorsal and median raphe nuclei (DR, MR) collectively innervate the entire forebrain and midbrain, modulating diverse physiology and behavior. To gain a fundamental understanding of their molecular heterogeneity, we used plate-based single-cell RNA-sequencing to generate a comprehensive dataset comprising eleven transcriptomically distinct serotonin neuron clusters. Systematic in situ hybridization mapped specific clusters to the principal DR, caudal DR, or MR. These transcriptomic clusters differentially express a rich repertoire of neuropeptides, receptors, ion channels, and transcription factors. We generated novel intersectional viral-genetic tools to access specific subpopulations. Whole-brain axonal projection mapping revealed that DR serotonin neurons co-expressing vesicular glutamate transporter-3 preferentially innervate the cortex, whereas those co-expressing thyrotropin-releasing hormone innervate subcortical regions in particular the hypothalamus. Reconstruction of 50 individual DR serotonin neurons revealed diverse and segregated axonal projection patterns at the single-cell level. Together, these results provide a molecular foundation of the heterogenous serotonin neuronal phenotypes.
Collapse
Affiliation(s)
- Jing Ren
- Department of Biology and Howard Hughes Medical InstituteStanford UniversityStanfordUnited States
| | - Alina Isakova
- Department of BioengineeringStanford UniversityStanfordUnited States
- Department of Applied PhysicsStanford UniversityStanfordUnited States
| | - Drew Friedmann
- Department of Biology and Howard Hughes Medical InstituteStanford UniversityStanfordUnited States
| | - Jiawei Zeng
- National Institute of Biological ScienceBeijingChina
| | - Sophie M Grutzner
- Department of Biology and Howard Hughes Medical InstituteStanford UniversityStanfordUnited States
| | - Albert Pun
- Department of Biology and Howard Hughes Medical InstituteStanford UniversityStanfordUnited States
| | - Grace Q Zhao
- Department of NeurobiologyStanford University School of MedicineStanfordUnited States
| | - Sai Saroja Kolluru
- Department of BioengineeringStanford UniversityStanfordUnited States
- Department of Applied PhysicsStanford UniversityStanfordUnited States
| | - Ruiyu Wang
- National Institute of Biological ScienceBeijingChina
| | - Rui Lin
- National Institute of Biological ScienceBeijingChina
| | - Pengcheng Li
- Britton Chance Center for Biomedical PhotonicsWuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology (HUST)WuhanChina
- HUST-Suzhou Institute for Brainsmatics, JITRI Institute for BrainsmaticsSuzhouChina
| | - Anan Li
- Britton Chance Center for Biomedical PhotonicsWuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology (HUST)WuhanChina
- HUST-Suzhou Institute for Brainsmatics, JITRI Institute for BrainsmaticsSuzhouChina
| | - Jennifer L Raymond
- Department of NeurobiologyStanford University School of MedicineStanfordUnited States
| | - Qingming Luo
- Britton Chance Center for Biomedical PhotonicsWuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology (HUST)WuhanChina
| | - Minmin Luo
- National Institute of Biological ScienceBeijingChina
- School of Life ScienceTsinghua UniversityBeijingChina
| | - Stephen R Quake
- Department of BioengineeringStanford UniversityStanfordUnited States
- Department of Applied PhysicsStanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Liqun Luo
- Department of Biology and Howard Hughes Medical InstituteStanford UniversityStanfordUnited States
| |
Collapse
|
13
|
Chleilat E, Mallmann R, Spanagel R, Klugbauer N, Krieglstein K, Roussa E. Spatiotemporal Role of Transforming Growth Factor Beta 2 in Developing and Mature Mouse Hindbrain Serotonergic Neurons. Front Cell Neurosci 2019; 13:427. [PMID: 31619968 PMCID: PMC6763588 DOI: 10.3389/fncel.2019.00427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/06/2019] [Indexed: 12/31/2022] Open
Abstract
Transforming growth factor betas are integral molecular components of the signalling cascades defining development and survival of several neuronal groups. Among TGF-β ligands, TGF-β2 has been considered as relatively more important during development. We have generated a conditional knockout mouse of the Tgf-β2 gene with knock-in of an EGFP reporter and subsequently a mouse line with cell-type specific deletion of TGF-β2 ligand from Krox20 expressing cells (i.e., in cells from rhombomeres r3 and r5). We performed a phenotypic analysis of the hindbrain serotonergic system during development and in adulthood, determined the neurochemical profile in hindbrain and forebrain, and assessed behavioural performance of wild type and mutant mice. Mutant mice revealed significantly decreased number of caudal 5-HT neurons at embryonic day (E) 14, and impaired development of caudal dorsal raphe, median raphe, raphe magnus, and raphe obscurus neurons at E18, a phenotype that was largely restored and even overshot in dorsal raphe of mutant adult mice. Serotonin levels were decreased in hindbrain but significantly increased in cortex of adult mutant mice, though without any behavioural consequences. These results highlight differential and temporal dependency of developing and adult neurons on TGF-β2. The results also indicate TGF-β2 being directly or indirectly potent to modulate neurotransmitter synthesis and metabolism. The novel floxed TGF-β2 mouse model is a suitable tool for analysing the in vivo functions of TGF-β2 during development and in adulthood in many organs.
Collapse
Affiliation(s)
- Enaam Chleilat
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Robert Mallmann
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health (ZI), Heidelberg University, Mannheim, Germany
| | - Norbert Klugbauer
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Kerstin Krieglstein
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Eleni Roussa
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, Albert Ludwigs University Freiburg, Freiburg, Germany
| |
Collapse
|
14
|
Mark MD, Wollenweber P, Gesk A, Kösters K, Batzke K, Janoschka C, Maejima T, Han J, Deneris ES, Herlitze S. RGS2 drives male aggression in mice via the serotonergic system. Commun Biol 2019; 2:373. [PMID: 31633064 PMCID: PMC6789038 DOI: 10.1038/s42003-019-0622-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 09/19/2019] [Indexed: 12/23/2022] Open
Abstract
Aggressive behavior in our modern, civilized society is often counterproductive and destructive. Identifying specific proteins involved in the disease can serve as therapeutic targets for treating aggression. Here, we found that overexpression of RGS2 in explicitly serotonergic neurons augments male aggression in control mice and rescues male aggression in Rgs2-/- mice, while anxiety is not affected. The aggressive behavior is directly correlated to the immediate early gene c-fos induction in the dorsal raphe nuclei and ventrolateral part of the ventromedial nucleus hypothalamus, to an increase in spontaneous firing in serotonergic neurons and to a reduction in the modulatory action of Gi/o and Gq/11 coupled 5HT and adrenergic receptors in serotonergic neurons of Rgs2-expressing mice. Collectively, these findings specifically identify that RGS2 expression in serotonergic neurons is sufficient to drive male aggression in mice and as a potential therapeutic target for treating aggression.
Collapse
Affiliation(s)
- Melanie D. Mark
- Department of General Zoology and Neurobiology, Ruhr-University Bochum, 44780 Bochum, Germany
| | - Patric Wollenweber
- Department of General Zoology and Neurobiology, Ruhr-University Bochum, 44780 Bochum, Germany
| | - Annika Gesk
- Department of General Zoology and Neurobiology, Ruhr-University Bochum, 44780 Bochum, Germany
| | - Katja Kösters
- Department of General Zoology and Neurobiology, Ruhr-University Bochum, 44780 Bochum, Germany
| | - Katharina Batzke
- Department of General Zoology and Neurobiology, Ruhr-University Bochum, 44780 Bochum, Germany
| | - Claudia Janoschka
- Department of General Zoology and Neurobiology, Ruhr-University Bochum, 44780 Bochum, Germany
| | - Takashi Maejima
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-8640 Japan
| | - Jing Han
- Institute for Applied Cancer Science, University of Texas, MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Evan S. Deneris
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH USA
| | - Stefan Herlitze
- Department of General Zoology and Neurobiology, Ruhr-University Bochum, 44780 Bochum, Germany
| |
Collapse
|
15
|
Arnold MR, Williams PH, McArthur JA, Archuleta AR, O'Neill CE, Hassell JE, Smith DG, Bachtell RK, Lowry CA. Effects of chronic caffeine exposure during adolescence and subsequent acute caffeine challenge during adulthood on rat brain serotonergic systems. Neuropharmacology 2019; 148:257-271. [PMID: 30579884 PMCID: PMC6438184 DOI: 10.1016/j.neuropharm.2018.12.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 01/09/2023]
Abstract
Caffeine is the most commonly used drug in the world. However, animal studies suggest that chronic consumption of caffeine during adolescence can result in enhanced anxiety-like behavioral responses during adulthood. One mechanism through which chronic caffeine administration may influence subsequent anxiety-like responses is through actions on brainstem serotonergic systems. In order to explore potential effects of chronic caffeine consumption on brainstem serotonergic systems, we evaluated the effects of a 28-day exposure to chronic caffeine (0.3 g/L; postnatal day 28-56) or vehicle administration in the drinking water, followed by 24 h caffeine withdrawal, and subsequent challenge with caffeine (30 mg/kg; s.c.) or vehicle in adolescent male rats. In Experiment 1, acute caffeine challenge induced a widespread activation of serotonergic neurons throughout the dorsal raphe nucleus (DR); this effect was attenuated in rats that had been exposed to chronic caffeine consumption. In Experiment 2, acute caffeine administration profoundly decreased tph2 and slc22a3 mRNA expression throughout the DR, with no effects on htr1a or slc6a4 mRNA expression. Chronic caffeine exposure for four weeks during adolescence was sufficient to decrease tph2 mRNA expression in the DR measured 28 h after caffeine withdrawal. Chronic caffeine administration during adolescence did not impact the ability of acute caffeine to decrease tph2 or slc22a3 mRNA expression. Together, these data suggest that both chronic caffeine administration during adolescence and acute caffeine challenge during adulthood are important determinants of serotonergic function and serotonergic gene expression, effects that may contribute to chronic effects of caffeine on anxiety-like responses.
Collapse
Affiliation(s)
- M R Arnold
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - P H Williams
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - J A McArthur
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - A R Archuleta
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA; Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - C E O'Neill
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA; Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - J E Hassell
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - D G Smith
- Department of Chemistry and Biochemistry, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - R K Bachtell
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA; Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - C A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA; Department of Physical Medicine & Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA; Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center, Denver Veterans Affairs Medical Center (VAMC), Denver, CO, 80220, USA; Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Denver, CO, 80220, USA.
| |
Collapse
|
16
|
Loupy KM, Arnold MR, Hassell JE, Lieb MW, Milton LN, Cler KE, Fox JH, Siebler PH, Schmidt D, Noronha SISR, Day HEW, Lowry CA. Evidence that preimmunization with a heat-killed preparation of Mycobacterium vaccae reduces corticotropin-releasing hormone mRNA expression in the extended amygdala in a fear-potentiated startle paradigm. Brain Behav Immun 2019; 77:127-140. [PMID: 30597198 DOI: 10.1016/j.bbi.2018.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 12/21/2018] [Accepted: 12/22/2018] [Indexed: 01/16/2023] Open
Abstract
Posttraumatic stress disorder (PTSD) is a trauma and stressor-related disorder that is characterized by dysregulation of glucocorticoid signaling, chronic low-grade inflammation, and impairment in the ability to extinguish learned fear. Corticotropin-releasing hormone (Crh) is a stress- and immune-responsive neuropeptide secreted from the paraventricular nucleus of the hypothalamus (PVN) to stimulate the hypothalamic-pituitary-adrenal (HPA) axis; however, extra-hypothalamic sources of Crh from the central nucleus of the amygdala (CeA) and bed nucleus of the stria terminalis (BNST) govern specific fear- and anxiety-related defensive behavioral responses. We previously reported that preimmunization with a heat-killed preparation of the immunoregulatory environmental bacterium Mycobacterium vaccae NCTC 11659 enhances fear extinction in a fear-potentiated startle (FPS) paradigm. In this follow-up study, we utilized an in situ hybridization histochemistry technique to investigate Crh, Crhr1, and Crhr2 mRNA expression in the CeA, BNST, and PVN of the same rats from the original study [Fox et al., 2017, Brain, Behavior, and Immunity, 66: 70-84]. Here, we demonstrate that preimmunization with M. vaccae NCTC 11659 decreases Crh mRNA expression in the CeA and BNST of rats exposed to the FPS paradigm, and, further, that Crh mRNA expression in these regions is correlated with fear behavior during extinction training. These data are consistent with the hypothesis that M. vaccae promotes stress-resilience by attenuating Crh production in fear- and anxiety-related circuits. These data suggest that immunization with M. vaccae may be an effective strategy for prevention of fear- and anxiety-related disorders.
Collapse
Affiliation(s)
- Kelsey M Loupy
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Mathew R Arnold
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - James E Hassell
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Margaret W Lieb
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Lauren N Milton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Kristin E Cler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - James H Fox
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Philip H Siebler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Dominic Schmidt
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Sylvana I S R Noronha
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Heidi E W Day
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Department of Physical Medicine & Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center, Denver Veterans Affairs Medical Center (VAMC), Denver, CO 80045, USA; Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Denver, CO 80045, USA.
| |
Collapse
|
17
|
Cham KL, Soga T, Parhar IS. Expression of RING Finger Protein 38 in Serotonergic Neurons in the Brain of Nile Tilapia, Oreochromis niloticus. Front Neuroanat 2018; 12:109. [PMID: 30574074 PMCID: PMC6292424 DOI: 10.3389/fnana.2018.00109] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/21/2018] [Indexed: 11/13/2022] Open
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is one of the major neurotransmitters, modulating diverse behaviours and physiological functions. Really interesting new gene (RING) finger protein 38 (RNF38) is an E3 ubiquitin ligase whose function remains unclear. A recent study has shown a possible regulatory relationship between RNF38 and the 5-HT system. Therefore, to gain insight into the role of RNF38 in the central 5-HT system, we identified the neuroanatomical location of 5-HT positive cells and investigated the relationship between RNF38 and the 5-HT system in the brain of the Nile tilapia, Oreochromis niloticus. Immunocytochemistry revealed three neuronal populations of 5-HT in the brain of tilapia; the paraventricular organ (PVO), the dorsal and ventral periventricular pretectal nuclei (PPd and PPv), and, the superior and inferior raphe (SR and IR). The 5-HT neuronal number was highest in the raphe (90.4 in SR, 284.6 in IR), followed by the pretectal area (22.3 in PPd, 209.8 in PPv). Double-label immunocytochemistry showed that the majority of 5-HT neurons express RNF38 nuclear proteins (66.5% in PPd; 77.9% in PPv; 35.7% in SR; 49.1% in IR). These findings suggest that RNF38 could be involved in E3 ubiquitination in the central 5-HT system.
Collapse
Affiliation(s)
- Kai Lin Cham
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Tomoko Soga
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Ishwar S Parhar
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
18
|
Hernández HG, Sandoval-Hernández AG, Garrido-Gil P, Labandeira-Garcia JL, Zelaya MV, Bayon GF, Fernández AF, Fraga MF, Arboleda G, Arboleda H. Alzheimer's disease DNA methylome of pyramidal layers in frontal cortex: laser-assisted microdissection study. Epigenomics 2018; 10:1365-1382. [PMID: 30324800 DOI: 10.2217/epi-2017-0160] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE To study DNA methylation patterns of cortical pyramidal layers susceptible to late-onset Alzheimer's disease (LOAD) neurodegeneration. METHODS Laser-assisted microdissection to select pyramidal layers' cells in frontal cortex of 32 human brains (18 LOAD) and Infinium DNA Methylation 450K analysis were performed to find differential methylated positions and regions, in addition to the corresponding gene set functional enrichment analyses. RESULTS Differential hypermethylation in several genomic regions and genes mainly in HOXA3, GSTP1, CXXC1-3 and BIN1. The functional enrichment analysis revealed genes significantly related to oxidative-stress and synapsis. CONCLUSION The present results indicate the differentially methylated genes related to neural projections, synapsis, oxidative stress and epigenetic regulator genes and represent the first epigenome of cortical pyramidal layers in LOAD.
Collapse
Affiliation(s)
- Hernán Guillermo Hernández
- PhD Program in Dentistry, Universidad Santo Tomás, Bucaramanga, Colombia.,Research Unity, Universidad Manuela Beltrán, Bucaramanga, Colombia
| | - Adrián Gabriel Sandoval-Hernández
- Grupo de Neurociencias y muerte Celular, Facultad de Medicina e instituto de Genética, Universidad Nacional de Colombia, Colombia.,Área de Bioquímica, Departamento de Química Universidad Nacional de Colombia, Colombia
| | - Pablo Garrido-Gil
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - José Luis Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - María Victoria Zelaya
- Navarrabiomed Brain Bank, Navarra Institute for Health Research, Pamplona, Navarra, Spain
| | - Gustavo F Bayon
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA), Universidad de Oviedo, Principado de Asturias, Spain
| | - Agustín F Fernández
- Fundación para la Investigación Biosanitaria de Asturias (FINBA), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Principado de Asturias, Spain
| | - Mario F Fraga
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo, Principado de Asturias, Spain
| | - Gonzalo Arboleda
- Grupo de Neurociencias y muerte Celular, Facultad de Medicina e instituto de Genética, Universidad Nacional de Colombia, Colombia.,Área de Bioquímica, Departamento de Química Universidad Nacional de Colombia, Colombia
| | - Humberto Arboleda
- Grupo de Neurociencias y muerte Celular, Facultad de Medicina e instituto de Genética, Universidad Nacional de Colombia, Colombia
| |
Collapse
|
19
|
Chleilat E, Skatulla L, Rahhal B, Hussein MT, Feuerstein M, Krieglstein K, Roussa E. TGF-β Signaling Regulates Development of Midbrain Dopaminergic and Hindbrain Serotonergic Neuron Subgroups. Neuroscience 2018; 381:124-137. [PMID: 29689292 DOI: 10.1016/j.neuroscience.2018.04.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/27/2018] [Accepted: 04/15/2018] [Indexed: 10/17/2022]
Abstract
Molecular and functional diversity within midbrain dopaminergic (mDA) and hindbrain serotonergic (5-HT) neurons has emerged as a relevant feature that could underlie selective vulnerability of neurons in clinical disorders. We have investigated the role of transforming growth factor beta (TGF-β) during development of mDA and 5-HT subgroups. We have generated TβRIIflox/flox::En1cre/+ mice where type II TGF-β receptor is conditionally deleted from engrailed 1-expressing cells and have investigated the hindbrain serotonergic system of these mice together with Tgf-β2-/- mice. The results show a significant decrease in the number of 5-HT neurons in TGF-β2-deficient mice at embryonic day (E) 12 and a selective significant decrease in the hindbrain paramedian raphe 5-HT neurons at E18, compared to wild type. Moreover, conditional deletion of TGF-β signaling from midbrain and rhombomere 1 leads to inactive TGF-β signaling in cre-expressing cells, impaired development of mouse mDA neuron subgroups and of dorsal raphe 5-HT neuron subgroups in a temporal manner. These results highlight a selective growth factor dependency of individual rostral hindbrain serotonergic subpopulations, emphasize the impact of TGF-β signaling during development of mDA and 5-HT subgroups, and suggest TGF-βs as potent candidates to establish diversity within the hindbrain serotonergic system. Thus, the data contribute to a better understanding of development and degeneration of mDA neurons and 5-HT-associated clinical disorders.
Collapse
Affiliation(s)
- Enaam Chleilat
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Lena Skatulla
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Belal Rahhal
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; School of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine.
| | - Manal T Hussein
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Melanie Feuerstein
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Kerstin Krieglstein
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Eleni Roussa
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
20
|
Grünewald L, Becker N, Camphausen A, O'Leary A, Lesch KP, Freudenberg F, Reif A. Expression of the ADHD candidate gene Diras2 in the brain. J Neural Transm (Vienna) 2018; 125:913-923. [PMID: 29488099 DOI: 10.1007/s00702-018-1867-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 02/21/2018] [Indexed: 12/21/2022]
Abstract
The distinct subgroup of the Ras family member 2 (DIRAS2) gene has been found to be associated with attention-deficit/hyperactivity disorder (ADHD) in one of our previous studies. This gene is coding for a small Ras GTPase with unknown function. DIRAS2 is highly expressed in the brain. However, the exact neural expression pattern of this gene was unknown so far. Therefore, we investigated the expressional profile of DIRAS2 in the human and murine brain. In the present study, qPCR analyses in the human and in the developing mouse brain, immunocytological double staining on murine hippocampal primary cells and RNA in situ hybridization (ISH) on brain sections of C57BL/6J wild-type mice, have been used to reveal the expression pattern of DIRAS2 in the brain. We could show that DIRAS2 expression in the human brain is the highest in the hippocampus and the cerebral cortex, which is in line with the ISH results in the mouse brain. During mouse brain development, Diras2 levels strongly increase from prenatal to late postnatal stages. Co-expression studies indicate Diras2 expression in glutamatergic and catecholaminergic neurons. Our findings support the idea of DIRAS2 as a candidate gene for ADHD as the timeline of its expression as well as the brain regions and cell types that show Diras2 expression correspond to those assumed to underlie the pathomechanisms of the disease.
Collapse
Affiliation(s)
- Lena Grünewald
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt, Germany.
| | - Nils Becker
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Annika Camphausen
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Aet O'Leary
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt, Germany
| | - Klaus-Peter Lesch
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Florian Freudenberg
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt, Germany
| |
Collapse
|
21
|
Deneris E, Gaspar P. Serotonin neuron development: shaping molecular and structural identities. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2018; 7:10.1002/wdev.301. [PMID: 29072810 PMCID: PMC5746461 DOI: 10.1002/wdev.301] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/03/2017] [Accepted: 08/24/2017] [Indexed: 02/03/2023]
Abstract
The continuing fascination with serotonin (5-hydroxytryptamine, 5-HT) as a nervous system chemical messenger began with its discovery in the brains of mammals in 1953. Among the many reasons for this decades-long interest is that the small numbers of neurons that make 5-HT influence the excitability of neural circuits in nearly every region of the brain and spinal cord. A further reason is that 5-HT dysfunction has been linked to a range of psychiatric and neurological disorders many of which have a neurodevelopmental component. This has led to intense interest in understanding 5-HT neuron development with the aim of determining whether early alterations in their generation lead to brain disease susceptibility. Here, we present an overview of the neuroanatomical organization of vertebrate 5-HT neurons, their neurogenesis, and prodigious axonal architectures, which enables the expansive reach of 5-HT neuromodulation in the central nervous system. We review recent findings that have revealed the molecular basis for the tremendous diversity of 5-HT neuron subtypes, the impact of environmental factors on 5-HT neuron development, and how 5-HT axons are topographically organized through disparate signaling pathways. We summarize studies of the gene regulatory networks that control the differentiation, maturation, and maintenance of 5-HT neurons. These studies show that the regulatory factors controlling acquisition of 5-HT-type transmitter identity continue to play critical roles in the functional maturation and the maintenance of 5-HT neurons. New insights are presented into how continuously expressed 5-HT regulatory factors control 5-HT neurons at different stages of life and how the regulatory networks themselves are maintained. WIREs Dev Biol 2018, 7:e301. doi: 10.1002/wdev.301 This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Gene Networks and Genomics Gene Expression and Transcriptional Hierarchies > Cellular Differentiation Nervous System Development > Secondary: Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Evan Deneris
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Patricia Gaspar
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S839, Paris, France
- Sorbonne Université, Paris, France
- Institut du Fer à Moulin, Campus Jussieu, Paris, France
| |
Collapse
|
22
|
Lee SJ, Kwon S, Gatti JR, Korcari E, Gresser TE, Felix PC, Keep SG, Pasquale KC, Bai T, Blanchett-Anderson SA, Wu NW, Obeng-Nyarko C, Senagbe KM, Young KC, Maripudi S, Yalavarthi BC, Korcari D, Liu AY, Schaffler BC, Keep RF, Wang MM. Large-scale identification of human cerebrovascular proteins: Inter-tissue and intracerebral vascular protein diversity. PLoS One 2017; 12:e0188540. [PMID: 29190776 PMCID: PMC5708641 DOI: 10.1371/journal.pone.0188540] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 11/08/2017] [Indexed: 12/21/2022] Open
Abstract
The human cerebrovascular system is responsible for regulating demand-dependent perfusion and maintaining the blood-brain barrier (BBB). In addition, defects in the human cerebrovasculature lead to stroke, intracerebral hemorrhage, vascular malformations, and vascular cognitive impairment. The objective of this study was to discover new proteins of the human cerebrovascular system using expression data from the Human Protein Atlas, a large-scale project which allows public access to immunohistochemical analysis of human tissues. We screened 20,158 proteins in the HPA and identified 346 expression patterns correlating to blood vessels in human brain. Independent experiments showed that 51/52 of these distributions could be experimentally replicated across different brain samples. Some proteins (40%) demonstrated endothelial cell (EC)-enriched expression, while others were expressed primarily in vascular smooth muscle cells (VSMC; 18%); 39% of these proteins were expressed in both cell types. Most brain EC markers were tissue oligospecific; that is, they were expressed in endothelia in an average of 4.8 out of 9 organs examined. Although most markers expressed in endothelial cells of the brain were present in all cerebral capillaries, a significant number (21%) were expressed only in a fraction of brain capillaries within each brain sample. Among proteins found in cerebral VSMC, virtually all were also expressed in peripheral VSMC and in non-vascular smooth muscle cells (SMC). Only one was potentially brain specific: VHL (Von Hippel-Lindau tumor suppressor). HRC (histidine rich calcium binding protein) and VHL were restricted to VSMC and not found in non-vascular tissues such as uterus or gut. In conclusion, we define a set of brain vascular proteins that could be relevant to understanding the unique physiology and pathophysiology of the human cerebrovasculature. This set of proteins defines inter-organ molecular differences in the vasculature and confirms the broad heterogeneity of vascular cells within the brain.
Collapse
Affiliation(s)
- Soo Jung Lee
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Soonhyung Kwon
- School of Social Work, University of Michigan, Ann Arbor, Michigan, United States of America
| | - John R. Gatti
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ejona Korcari
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ty E. Gresser
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Princess C. Felix
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Simon G. Keep
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kevin C. Pasquale
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Tongxu Bai
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | | | - Nancy W. Wu
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Charissa Obeng-Nyarko
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kossi M. Senagbe
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kathy C. Young
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Snehaa Maripudi
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Bharath C. Yalavarthi
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Dajana Korcari
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Andre Y. Liu
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Benjamin C. Schaffler
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Richard F. Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Michael M. Wang
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
- Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
23
|
Fox JH, Hassell JE, Siebler PH, Arnold MR, Lamb AK, Smith DG, Day HEW, Smith TM, Simmerman EM, Outzen AA, Holmes KS, Brazell CJ, Lowry CA. Preimmunization with a heat-killed preparation of Mycobacterium vaccae enhances fear extinction in the fear-potentiated startle paradigm. Brain Behav Immun 2017; 66:70-84. [PMID: 28888667 DOI: 10.1016/j.bbi.2017.08.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/02/2017] [Accepted: 08/15/2017] [Indexed: 12/31/2022] Open
Abstract
The hygiene hypothesis or "Old Friends" hypothesis proposes that inflammatory diseases are increasing in modern urban societies, due in part to reduced exposure to microorganisms that drive immunoregulatory circuits, and a failure to terminate inappropriate inflammatory responses. Inappropriate inflammation is also emerging as a risk factor for trauma-related, anxiety, and affective disorders, including posttraumatic stress disorder (PTSD), which is characterized as persistent re-experiencing of the trauma after a traumatic experience. Traumatic experiences can lead to long-lasting fear memories and exaggerated fear potentiation of the acoustic startle reflex. The acoustic startle reflex is an ethologically relevant reflex and can be potentiated in both humans and rats through Pavlovian conditioning. Mycobacterium vaccae NCTC 11659 is a soil-derived bacterium with immunoregulatory and anti-inflammatory properties that has been demonstrated to confer stress resilience in mice. Here we immunized adult male Sprague Dawley rats 3×, once per week, with a heat-killed preparation of M. vaccae NCTC 11659 (0.1mg, s.c., in 100µl borate-buffered saline) or vehicle, and, then, 3weeks following the final immunization, tested them in the fear-potentiated startle paradigm; controls were maintained under home cage control conditions throughout the experiment (n=11-12 per group). Rats were tested on days 1 and 2 for baseline acoustic startle, received fear conditioning on days 3 and 4, and underwent fear extinction training on days 5-10. Rats were euthanized on day 11 and brain tissue was sectioned for analysis of mRNA expression for genes important in control of brain serotonergic signaling, including tph2, htr1a, slc6a4, and slc22a3, throughout the brainstem dorsal and median raphe nuclei. Immunization with M. vaccae had no effect on baseline acoustic startle or fear expression on day 5. However, M. vaccae-immunized rats showed enhanced between-session and within-session extinction on day 6, relative to vehicle-immunized controls. Immunization with M. vaccae and fear-potentiated startle altered serotonergic gene expression in a gene- and subregion-specific manner. These data are consistent with the hypothesis that immunoregulatory strategies, such as preimmunization with M. vaccae, have potential for prevention of stress- and trauma-related psychiatric disorders.
Collapse
Affiliation(s)
- James H Fox
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - James E Hassell
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Philip H Siebler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Mathew R Arnold
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Andrew K Lamb
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - David G Smith
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Heidi E W Day
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Tessa M Smith
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Emma M Simmerman
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Alexander A Outzen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Kaley S Holmes
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Christopher J Brazell
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA; Department of Physical Medicine & Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Rocky Mountain Mental Illness Research Education and Clinical Center, Denver, CO 80220, USA; Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Denver, CO 80220, USA.
| |
Collapse
|
24
|
Forero A, Rivero O, Wäldchen S, Ku HP, Kiser DP, Gärtner Y, Pennington LS, Waider J, Gaspar P, Jansch C, Edenhofer F, Resink TJ, Blum R, Sauer M, Lesch KP. Cadherin-13 Deficiency Increases Dorsal Raphe 5-HT Neuron Density and Prefrontal Cortex Innervation in the Mouse Brain. Front Cell Neurosci 2017; 11:307. [PMID: 29018333 PMCID: PMC5623013 DOI: 10.3389/fncel.2017.00307] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 09/15/2017] [Indexed: 01/29/2023] Open
Abstract
Background: During early prenatal stages of brain development, serotonin (5-HT)-specific neurons migrate through somal translocation to form the raphe nuclei and subsequently begin to project to their target regions. The rostral cluster of cells, comprising the median and dorsal raphe (DR), innervates anterior regions of the brain, including the prefrontal cortex. Differential analysis of the mouse 5-HT system transcriptome identified enrichment of cell adhesion molecules in 5-HT neurons of the DR. One of these molecules, cadherin-13 (Cdh13) has been shown to play a role in cell migration, axon pathfinding, and synaptogenesis. This study aimed to investigate the contribution of Cdh13 to the development of the murine brain 5-HT system. Methods: For detection of Cdh13 and components of the 5-HT system at different embryonic developmental stages of the mouse brain, we employed immunofluorescence protocols and imaging techniques, including epifluorescence, confocal and structured illumination microscopy. The consequence of CDH13 loss-of-function mutations on brain 5-HT system development was explored in a mouse model of Cdh13 deficiency. Results: Our data show that in murine embryonic brain Cdh13 is strongly expressed on 5-HT specific neurons of the DR and in radial glial cells (RGCs), which are critically involved in regulation of neuronal migration. We observed that 5-HT neurons are intertwined with these RGCs, suggesting that these neurons undergo RGC-guided migration. Cdh13 is present at points of intersection between these two cell types. Compared to wildtype controls, Cdh13-deficient mice display increased cell densities in the DR at embryonic stages E13.5, E17.5, and adulthood, and higher serotonergic innervation of the prefrontal cortex at E17.5. Conclusion: Our findings provide evidence for a role of CDH13 in the development of the serotonergic system in early embryonic stages. Specifically, we indicate that Cdh13 deficiency affects the cell density of the developing DR and the posterior innervation of the prefrontal cortex (PFC), and therefore might be involved in the migration, axonal outgrowth and terminal target finding of DR 5-HT neurons. Dysregulation of CDH13 expression may thus contribute to alterations in this system of neurotransmission, impacting cognitive function, which is frequently impaired in neurodevelopmental disorders including attention-deficit/hyperactivity and autism spectrum disorders.
Collapse
Affiliation(s)
- Andrea Forero
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Olga Rivero
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Sina Wäldchen
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Hsing-Ping Ku
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Dominik P Kiser
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Yvonne Gärtner
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Laura S Pennington
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Jonas Waider
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Patricia Gaspar
- Institut du Fer á Moulin, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S839, Paris, France
| | - Charline Jansch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Frank Edenhofer
- Department of Genomics, Stem Cell Biology and Regenerative Medicine, Institute of Molecular Biology and Center for Molecular Biosciences Innsbruck (CMBI), Leopold-Franzens-University Innsbruck, Innsbruck, Austria.,Stem Cell Biology and Regenerative Medicine Group, Institute of Anatomy and Cell Biology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Thérèse J Resink
- Laboratory for Signal Transduction, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Robert Blum
- Department of Clinical Neurobiology, University of Würzburg, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany.,Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I. M. Sechenov First Moscow State Medical University, Moscow, Russia.,Department of Translational Neuroscience, School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
25
|
Kamitakahara A, Wu HH, Levitt P. Distinct projection targets define subpopulations of mouse brainstem vagal neurons that express the autism-associated MET receptor tyrosine kinase. J Comp Neurol 2017; 525:3787-3808. [PMID: 28758209 DOI: 10.1002/cne.24294] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 07/14/2017] [Accepted: 07/17/2017] [Indexed: 12/11/2022]
Abstract
Detailed anatomical tracing and mapping of the viscerotopic organization of the vagal motor nuclei has provided insight into autonomic function in health and disease. To further define specific cellular identities, we paired information based on visceral connectivity with a cell-type specific marker of a subpopulation of neurons in the dorsal motor nucleus of the vagus (DMV) and nucleus ambiguus (nAmb) that express the autism-associated MET receptor tyrosine kinase. As gastrointestinal disturbances are common in children with autism spectrum disorder (ASD), we sought to define the relationship between MET-expressing (MET+) neurons in the DMV and nAmb, and the gastrointestinal tract. Using wholemount tissue staining and clearing, or retrograde tracing in a METEGFP transgenic mouse, we identify three novel subpopulations of EGFP+ vagal brainstem neurons: (a) EGFP+ neurons in the nAmb projecting to the esophagus or laryngeal muscles, (b) EGFP+ neurons in the medial DMV projecting to the stomach, and (b) EGFP+ neurons in the lateral DMV projecting to the cecum and/or proximal colon. Expression of the MET ligand, hepatocyte growth factor (HGF), by tissues innervated by vagal motor neurons during fetal development reveal potential sites of HGF-MET interaction. Furthermore, similar cellular expression patterns of MET in the brainstem of both the mouse and nonhuman primate suggests that MET expression at these sites is evolutionarily conserved. Together, the data suggest that MET+ neurons in the brainstem vagal motor nuclei are anatomically positioned to regulate distinct portions of the gastrointestinal tract, with implications for the pathophysiology of gastrointestinal comorbidities of ASD.
Collapse
Affiliation(s)
- Anna Kamitakahara
- Program in Developmental Neurogenetics, Institute for the Developing Mind, The Saban Resarch Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Hsiao-Huei Wu
- Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Pat Levitt
- Program in Developmental Neurogenetics, Institute for the Developing Mind, The Saban Resarch Institute, Children's Hospital Los Angeles, Los Angeles, California.,Department of Pediatrics, Keck School of Medicine of University of Southern California, Los Angeles, California.,University of Southern California Program in Neuroscience, Los Angeles, California
| |
Collapse
|
26
|
Spencer WC, Deneris ES. Regulatory Mechanisms Controlling Maturation of Serotonin Neuron Identity and Function. Front Cell Neurosci 2017; 11:215. [PMID: 28769770 PMCID: PMC5515867 DOI: 10.3389/fncel.2017.00215] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/05/2017] [Indexed: 11/29/2022] Open
Abstract
The brain serotonin (5-hydroxytryptamine; 5-HT) system has been extensively studied for its role in normal physiology and behavior, as well as, neuropsychiatric disorders. The broad influence of 5-HT on brain function, is in part due to the vast connectivity pattern of 5-HT-producing neurons throughout the CNS. 5-HT neurons are born and terminally specified midway through embryogenesis, then enter a protracted period of maturation, where they functionally integrate into CNS circuitry and then are maintained throughout life. The transcriptional regulatory networks controlling progenitor cell generation and terminal specification of 5-HT neurons are relatively well-understood, yet the factors controlling 5-HT neuron maturation are only recently coming to light. In this review, we first provide an update on the regulatory network controlling 5-HT neuron development, then delve deeper into the properties and regulatory strategies governing 5-HT neuron maturation. In particular, we discuss the role of the 5-HT neuron terminal selector transcription factor (TF) Pet-1 as a key regulator of 5-HT neuron maturation. Pet-1 was originally shown to positively regulate genes needed for 5-HT synthesis, reuptake and vesicular transport, hence 5-HT neuron-type transmitter identity. It has now been shown to regulate, both positively and negatively, many other categories of genes in 5-HT neurons including ion channels, GPCRs, transporters, neuropeptides, and other transcription factors. Its function as a terminal selector results in the maturation of 5-HT neuron excitability, firing characteristics, and synaptic modulation by several neurotransmitters. Furthermore, there is a temporal requirement for Pet-1 in the control of postmitotic gene expression trajectories thus indicating a direct role in 5-HT neuron maturation. Proper regulation of the maturation of cellular identity is critical for normal neuronal functioning and perturbations in the gene regulatory networks controlling these processes may result in long-lasting changes in brain function in adulthood. Further study of 5-HT neuron gene regulatory networks is likely to provide additional insight into how neurons acquire their mature identities and how terminal selector-type TFs function in postmitotic vertebrate neurons.
Collapse
Affiliation(s)
- William C Spencer
- Department of Neurosciences, Case Western Reserve UniversityCleveland, OH, United States
| | - Evan S Deneris
- Department of Neurosciences, Case Western Reserve UniversityCleveland, OH, United States
| |
Collapse
|
27
|
Identified Serotonergic Modulatory Neurons Have Heterogeneous Synaptic Connectivity within the Olfactory System of Drosophila. J Neurosci 2017; 37:7318-7331. [PMID: 28659283 DOI: 10.1523/jneurosci.0192-17.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 05/20/2017] [Accepted: 06/19/2017] [Indexed: 11/21/2022] Open
Abstract
Modulatory neurons project widely throughout the brain, dynamically altering network processing based on an animal's physiological state. The connectivity of individual modulatory neurons can be complex, as they often receive input from a variety of sources and are diverse in their physiology, structure, and gene expression profiles. To establish basic principles about the connectivity of individual modulatory neurons, we examined a pair of identified neurons, the "contralaterally projecting, serotonin-immunoreactive deutocerebral neurons" (CSDns), within the olfactory system of Drosophila Specifically, we determined the neuronal classes providing synaptic input to the CSDns within the antennal lobe (AL), an olfactory network targeted by the CSDns, and the degree to which CSDn active zones are uniformly distributed across the AL. Using anatomical techniques, we found that the CSDns received glomerulus-specific input from olfactory receptor neurons (ORNs) and projection neurons (PNs), and networkwide input from local interneurons (LNs). Furthermore, we quantified the number of CSDn active zones in each glomerulus and found that CSDn output is not uniform, but rather heterogeneous, across glomeruli and stereotyped from animal to animal. Finally, we demonstrate that the CSDns synapse broadly onto LNs and PNs throughout the AL but do not synapse upon ORNs. Our results demonstrate that modulatory neurons do not necessarily provide purely top-down input but rather receive neuron class-specific input from the networks that they target, and that even a two cell modulatory network has highly heterogeneous, yet stereotyped, pattern of connectivity.SIGNIFICANCE STATEMENT Modulatory neurons often project broadly throughout the brain to alter processing based on physiological state. However, the connectivity of individual modulatory neurons to their target networks is not well understood, as modulatory neuron populations are heterogeneous in their physiology, morphology, and gene expression. In this study, we use a pair of identified serotonergic neurons within the Drosophila olfactory system as a model to establish a framework for modulatory neuron connectivity. We demonstrate that individual modulatory neurons can integrate neuron class-specific input from their target network, which is often nonreciprocal. Additionally, modulatory neuron output can be stereotyped, yet nonuniform, across network regions. Our results provide new insight into the synaptic relationships that underlie network function of modulatory neurons.
Collapse
|
28
|
Oliveira MAP, Balling R, Smidt MP, Fleming RMT. Embryonic development of selectively vulnerable neurons in Parkinson's disease. NPJ Parkinsons Dis 2017; 3:21. [PMID: 28685157 PMCID: PMC5484687 DOI: 10.1038/s41531-017-0022-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 05/24/2017] [Accepted: 06/01/2017] [Indexed: 02/07/2023] Open
Abstract
A specific set of brainstem nuclei are susceptible to degeneration in Parkinson's disease. We hypothesise that neuronal vulnerability reflects shared phenotypic characteristics that confer selective vulnerability to degeneration. Neuronal phenotypic specification is mainly the cumulative result of a transcriptional regulatory program that is active during the development. By manual curation of the developmental biology literature, we comprehensively reconstructed an anatomically resolved cellular developmental lineage for the adult neurons in five brainstem regions that are selectively vulnerable to degeneration in prodromal or early Parkinson's disease. We synthesised the literature on transcription factors that are required to be active, or required to be inactive, in the development of each of these five brainstem regions, and at least two differentially vulnerable nuclei within each region. Certain transcription factors, e.g., Ascl1 and Lmx1b, seem to be required for specification of many brainstem regions that are susceptible to degeneration in early Parkinson's disease. Some transcription factors can even distinguish between differentially vulnerable nuclei within the same brain region, e.g., Pitx3 is required for specification of the substantia nigra pars compacta, but not the ventral tegmental area. We do not suggest that Parkinson's disease is a developmental disorder. In contrast, we consider identification of shared developmental trajectories as part of a broader effort to identify the molecular mechanisms that underlie the phenotypic features that are shared by selectively vulnerable neurons. Systematic in vivo assessment of fate determining transcription factors should be completed for all neuronal populations vulnerable to degeneration in early Parkinson's disease.
Collapse
Affiliation(s)
- Miguel A. P. Oliveira
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, Belvaux, L-4362 Luxembourg
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, Belvaux, L-4362 Luxembourg
| | - Marten P. Smidt
- Department of Molecular Neuroscience, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Sciencepark 904, 1098 XH Amsterdam, The Netherlands
| | - Ronan M. T. Fleming
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, Belvaux, L-4362 Luxembourg
| |
Collapse
|
29
|
Teissier A, Soiza-Reilly M, Gaspar P. Refining the Role of 5-HT in Postnatal Development of Brain Circuits. Front Cell Neurosci 2017; 11:139. [PMID: 28588453 PMCID: PMC5440475 DOI: 10.3389/fncel.2017.00139] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 04/26/2017] [Indexed: 11/30/2022] Open
Abstract
Changing serotonin (5-hydroxytryptamine, 5-HT) brain levels during critical periods in development has long-lasting effects on brain function, particularly on later anxiety/depression-related behaviors in adulthood. A large part of the known developmental effects of 5-HT occur during critical periods of postnatal life, when activity-dependent mechanisms remodel neural circuits. This was first demonstrated for the maturation of sensory brain maps in the barrel cortex and the visual system. More recently this has been extended to the 5-HT raphe circuits themselves and to limbic circuits. Recent studies overviewed here used new genetic models in mice and rats and combined physiological and structural approaches to provide new insights on the cellular and molecular mechanisms controlled by 5-HT during late stages of neural circuit maturation in the raphe projections, the somatosensory cortex and the visual system. Similar mechanisms appear to be also involved in the maturation of limbic circuits such as prefrontal circuits. The latter are of particular relevance to understand the impact of transient 5-HT dysfunction during postnatal life on psychiatric illnesses and emotional disorders in adult life.
Collapse
Affiliation(s)
- Anne Teissier
- Institut du Fer à Moulin, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S839Paris, France.,Université Pierre et Marie CurieParis, France.,Institut du Fer à MoulinParis, France
| | - Mariano Soiza-Reilly
- Institut du Fer à Moulin, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S839Paris, France.,Université Pierre et Marie CurieParis, France.,Institut du Fer à MoulinParis, France
| | - Patricia Gaspar
- Institut du Fer à Moulin, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S839Paris, France.,Université Pierre et Marie CurieParis, France.,Institut du Fer à MoulinParis, France
| |
Collapse
|
30
|
Puissant MM, Mouradian GC, Liu P, Hodges MR. Identifying Candidate Genes that Underlie Cellular pH Sensitivity in Serotonin Neurons Using Transcriptomics: A Potential Role for Kir5.1 Channels. Front Cell Neurosci 2017; 11:34. [PMID: 28270749 PMCID: PMC5318415 DOI: 10.3389/fncel.2017.00034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/06/2017] [Indexed: 11/30/2022] Open
Abstract
Ventilation is continuously adjusted by a neural network to maintain blood gases and pH. Acute CO2 and/or pH regulation requires neural feedback from brainstem cells that encode CO2/pH to modulate ventilation, including but not limited to brainstem serotonin (5-HT) neurons. Brainstem 5-HT neurons modulate ventilation and are stimulated by hypercapnic acidosis, the sensitivity of which increases with increasing postnatal age. The proper function of brainstem 5-HT neurons, particularly during post-natal development is critical given that multiple abnormalities in the 5-HT system have been identified in victims of Sudden Infant Death Syndrome. Here, we tested the hypothesis that there are age-dependent increases in expression of pH-sensitive ion channels in brainstem 5-HT neurons, which may underlie their cellular CO2/pH sensitivity. Midline raphe neurons were acutely dissociated from neonatal and mature transgenic SSePet-eGFP rats [which have enhanced green fluorescent protein (eGFP) expression in all 5-HT neurons] and sorted with fluorescence-activated cell sorting (FACS) into 5-HT-enriched and non-5-HT cell pools for subsequent RNA extraction, cDNA library preparation and RNA sequencing. Overlapping differential expression analyses pointed to age-dependent shifts in multiple ion channels, including but not limited to the pH-sensitive potassium ion (K+) channel genes kcnj10 (Kir4.1), kcnj16 (Kir5.1), kcnk1 (TWIK-1), kcnk3 (TASK-1) and kcnk9 (TASK-3). Intracellular contents isolated from single adult eGFP+ 5-HT neurons confirmed gene expression of Kir4.1, Kir5.1 and other K+ channels, but also showed heterogeneity in the expression of multiple genes. 5-HT neuron-enriched cell pools from selected post-natal ages showed increases in Kir4.1, Kir5.1, and TWIK-1, fitting with age-dependent increases in Kir4.1 and Kir5.1 protein expression in raphe tissue samples. Immunofluorescence imaging confirmed Kir5.1 protein was co-localized to brainstem neurons and glia including 5-HT neurons as expected. However, Kir4.1 protein expression was restricted to glia, suggesting that it may not contribute to 5-HT neuron pH sensitivity. Although there are caveats to this approach, the data suggest that pH-sensitive Kir5.1 channels may underlie cellular CO2/pH chemosensitivity in brainstem 5-HT neurons.
Collapse
Affiliation(s)
- Madeleine M Puissant
- Department of Physiology, Medical College of Wisconsin, MilwaukeeWI, USA; Neuroscience Research Center, Medical College of Wisconsin, MilwaukeeWI, USA
| | - Gary C Mouradian
- Department of Physiology, Medical College of Wisconsin, MilwaukeeWI, USA; Center for Systems Molecular Medicine, Medical College of Wisconsin, MilwaukeeWI, USA
| | - Pengyuan Liu
- Department of Physiology, Medical College of Wisconsin, MilwaukeeWI, USA; Center for Systems Molecular Medicine, Medical College of Wisconsin, MilwaukeeWI, USA; Cancer Research Center, Medical College of Wisconsin, MilwaukeeWI, USA
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin, MilwaukeeWI, USA; Neuroscience Research Center, Medical College of Wisconsin, MilwaukeeWI, USA; Center for Systems Molecular Medicine, Medical College of Wisconsin, MilwaukeeWI, USA
| |
Collapse
|
31
|
EphrinA5 Signaling Is Required for the Distinctive Targeting of Raphe Serotonin Neurons in the Forebrain. eNeuro 2017; 4:eN-NWR-0327-16. [PMID: 28197551 PMCID: PMC5292598 DOI: 10.1523/eneuro.0327-16.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 01/06/2017] [Indexed: 12/13/2022] Open
Abstract
Serotonin (5-HT) neurotransmission in the brain relies on a widespread axon terminal network originating from the hindbrain raphe nuclei. These projections are topographically organized such that the dorsal (DR), and median raphe (MnR) nuclei have different brain targets. However, the guidance molecules involved in this selective targeting in development are unknown. Here, we show the implication of ephrinA5 signaling in this process. We find that the EphA5 gene is selectively expressed in a subset of 5-HT neurons during embryonic and postnatal development. Highest coexpression of EphA5 and the 5-HT marker Tph2 is found in the DR, with lower coexpression in the MnR, and hardly any colocalization of the caudal raphe in the medulla. Accordingly, ephrinA induced a dose-dependent collapse response of 5-HT growth cones cultured from rostral but not caudal raphe. Ectopic expression of ephrinA3, after in utero electroporation in the amygdala and piriform cortex, repelled 5-HT raphe fiber ingrowth. Conversely, misplaced DR 5-HT axons were found in ephrin A5 knockout mice in brain regions that are normally only targeted by MnR 5-HT axons. This causes an overall increase in the density of 5-HT innervation in the ventromedial hypothalamus, the suprachiasmatic nucleus, and the olfactory bulb. All these brain areas have high expression of ephrinAs at the time of 5-HT fiber ingrowth. Present results show for the first time the role of a guidance molecule for the region-specific targeting of raphe neurons. This has important implications to understand how functional parsing of central 5-HT neurons is established during development.
Collapse
|
32
|
Functional Impact of An ADHD-Associated DIRAS2 Promoter Polymorphism. Neuropsychopharmacology 2016; 41:3025-3031. [PMID: 27364329 PMCID: PMC5101550 DOI: 10.1038/npp.2016.113] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 06/20/2016] [Accepted: 06/24/2016] [Indexed: 12/17/2022]
Abstract
The DIRAS2 gene is coding for a small Ras GTPase with so far unknown function. In a previous study, we described the association of DIRAS2 rs1412005, as well as a haplotype containing this polymorphism and located in the promoter region of this gene with attention-deficit/hyperactivity disorder (ADHD). In the present study, we searched for rare variants within or near the DIRAS2 gene that might be associated with ADHD using next-generation sequencing. As we were not able to detect any rare variants associated with the disease, we sought to establish a functional role of DIRAS2 rs1412005 on the molecular or systems level. First, we investigated whether it has an influence on gene expression by means of a luciferase-based promoter assay. We could demonstrate that the minor risk allele goes along with the increased expression of the reporter gene. Next, we aimed to identify differences in response inhibition between risk-allele and non-risk allele carriers in children suffering from ADHD and healthy control individuals by analyzing event-related potentials in the electroencephalogram during a Go/NoGo task. Risk-allele carriers showed a changed NoGo anteriorization. Therefore, our results suggest an impact of the investigated polymorphism on the prefrontal response control in children with ADHD. These results imply that the promoter polymorphism is indeed the associated as well as in itself a causal variant. Further research is thus warranted to clarify the mechanisms linking DIRAS2 to ADHD.
Collapse
|
33
|
Niederkofler V, Asher TE, Okaty BW, Rood BD, Narayan A, Hwa LS, Beck SG, Miczek KA, Dymecki SM. Identification of Serotonergic Neuronal Modules that Affect Aggressive Behavior. Cell Rep 2016; 17:1934-1949. [PMID: 27851959 PMCID: PMC5156533 DOI: 10.1016/j.celrep.2016.10.063] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 09/16/2016] [Accepted: 10/17/2016] [Indexed: 11/19/2022] Open
Abstract
Escalated aggression can have devastating societal consequences, yet underlying neurobiological mechanisms are poorly understood. Here, we show significantly increased inter-male mouse aggression when neurotransmission is constitutively blocked from either of two subsets of serotonergic, Pet1+ neurons: one identified by dopamine receptor D1(Drd1a)::cre-driven activity perinatally, and the other by Drd2::cre from pre-adolescence onward. Blocking neurotransmission from other Pet1+ neuron subsets of similar size and/or overlapping anatomical domains had no effect on aggression compared with controls, suggesting subtype-specific serotonergic neuron influences on aggression. Using established and novel intersectional genetic tools, we further characterized these subtypes across multiple parameters, showing both overlapping and distinct features in axonal projection targets, gene expression, electrophysiological properties, and effects on non-aggressive behaviors. Notably, Drd2::cre marked 5-HT neurons exhibited D2-dependent inhibitory responses to dopamine in slices, suggesting direct and specific interplay between inhibitory dopaminergic signaling and a serotonergic subpopulation. Thus, we identify specific serotonergic modules that shape aggression.
Collapse
Affiliation(s)
- Vera Niederkofler
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Tedi E Asher
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Benjamin W Okaty
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Benjamin D Rood
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Ankita Narayan
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Lara S Hwa
- Department of Psychology, Tufts University, 530 Boston Avenue, Medford, MA 02155, USA
| | - Sheryl G Beck
- Departments of Anesthesiology and Critical Care and Pharmacology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104; Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Klaus A Miczek
- Department of Psychology, Tufts University, 530 Boston Avenue, Medford, MA 02155, USA
| | - Susan M Dymecki
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| |
Collapse
|
34
|
Haugas M, Tikker L, Achim K, Salminen M, Partanen J. Gata2 and Gata3 regulate the differentiation of serotonergic and glutamatergic neuron subtypes of the dorsal raphe. Development 2016; 143:4495-4508. [PMID: 27789623 DOI: 10.1242/dev.136614] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 10/18/2016] [Indexed: 12/18/2022]
Abstract
Serotonergic and glutamatergic neurons of the dorsal raphe regulate many brain functions and are important for mental health. Their functional diversity is based on molecularly distinct subtypes; however, the development of this heterogeneity is poorly understood. We show that the ventral neuroepithelium of mouse anterior hindbrain is divided into specific subdomains giving rise to serotonergic neurons as well as other types of neurons and glia. The newly born serotonergic precursors are segregated into distinct subpopulations expressing vesicular glutamate transporter 3 (Vglut3) or serotonin transporter (Sert). These populations differ in their requirements for transcription factors Gata2 and Gata3, which are activated in the post-mitotic precursors. Gata2 operates upstream of Gata3 as a cell fate selector in both populations, whereas Gata3 is important for the differentiation of the Sert+ precursors and for the serotonergic identity of the Vglut3+ precursors. Similar to the serotonergic neurons, the Vglut3-expressing glutamatergic neurons, located in the central dorsal raphe, are derived from neural progenitors in the ventral hindbrain and express Pet1 Furthermore, both Gata2 and Gata3 are redundantly required for their differentiation. Our study demonstrates lineage relationships of the dorsal raphe neurons and suggests that functionally significant heterogeneity of these neurons is established early during their differentiation.
Collapse
Affiliation(s)
- Maarja Haugas
- Department of Biosciences, P.O. Box 56, Viikinkaari 9, FIN00014-University of Helsinki, Helsinki, Finland
| | - Laura Tikker
- Department of Biosciences, P.O. Box 56, Viikinkaari 9, FIN00014-University of Helsinki, Helsinki, Finland
| | - Kaia Achim
- EMBL Developmental Biology Unit, Meyerhofstrasse 1, Heidelberg 69117, Germany
| | - Marjo Salminen
- Department of Veterinary Biosciences, P.O. Box 66, Agnes Sjobergin katu 2, FIN00014-University of Helsinki, Helsinki, Finland
| | - Juha Partanen
- Department of Biosciences, P.O. Box 56, Viikinkaari 9, FIN00014-University of Helsinki, Helsinki, Finland
| |
Collapse
|
35
|
Mlinar B, Montalbano A, Piszczek L, Gross C, Corradetti R. Firing Properties of Genetically Identified Dorsal Raphe Serotonergic Neurons in Brain Slices. Front Cell Neurosci 2016; 10:195. [PMID: 27536220 PMCID: PMC4971071 DOI: 10.3389/fncel.2016.00195] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 07/22/2016] [Indexed: 11/13/2022] Open
Abstract
Tonic spiking of serotonergic neurons establishes serotonin levels in the brain. Since the first observations, slow regular spiking has been considered as a defining feature of serotonergic neurons. Recent studies, however, have revealed the heterogeneity of serotonergic neurons at multiple levels, comprising their electrophysiological properties, suggesting the existence of functionally distinct cellular subpopulations. In order to examine in an unbiased manner whether serotonergic neurons of the dorsal raphe nucleus (DRN) are heterogeneous, we used a non-invasive loose-seal cell-attached method to record α1 adrenergic receptor-stimulated spiking of a large sample of neurons in brain slices obtained from transgenic mice lines that express fluorescent marker proteins under the control of serotonergic system-specific Tph2 and Pet-1 promoters. We found wide homogeneous distribution of firing rates, well fitted by a single Gaussian function (r (2) = 0.93) and independent of anatomical location (P = 0.45), suggesting that in terms of intrinsic firing properties, serotonergic neurons in the DRN represent a single cellular population. Characterization of the population in terms of spiking regularity was hindered by its dependence on the firing rate. For instance, the coefficient of variation of the interspike intervals (ISI), a common measure of spiking irregularity, is of limited usefulness since it correlates negatively with the firing rate (r = -0.33, P < 0.0001). Nevertheless, the majority of neurons exhibited regular, pacemaker-like activity, with coefficient of variance of the ISI lower than 0.5 in ~97% of cases. Unexpectedly, a small percentage of neurons (~1%) exhibited a particular spiking pattern, characterized by low frequency (~0.02-0.1 Hz) oscillations in the firing rate. Transitions between regular and oscillatory firing were observed, suggesting that the oscillatory firing is an alternative firing pattern of serotonergic neurons.
Collapse
Affiliation(s)
- Boris Mlinar
- Department of Neuroscience, Psychology, Drug Research and Children's Health, University of Florence Florence, Italy
| | - Alberto Montalbano
- Department of Neuroscience, Psychology, Drug Research and Children's Health, University of Florence Florence, Italy
| | - Lukasz Piszczek
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| | - Cornelius Gross
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| | - Renato Corradetti
- Department of Neuroscience, Psychology, Drug Research and Children's Health, University of Florence Florence, Italy
| |
Collapse
|
36
|
Pet-1 Switches Transcriptional Targets Postnatally to Regulate Maturation of Serotonin Neuron Excitability. J Neurosci 2016; 36:1758-74. [PMID: 26843655 DOI: 10.1523/jneurosci.3798-15.2016] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
UNLABELLED Newborn neurons enter an extended maturation stage, during which they acquire excitability characteristics crucial for development of presynaptic and postsynaptic connectivity. In contrast to earlier specification programs, little is known about the regulatory mechanisms that control neuronal maturation. The Pet-1 ETS (E26 transformation-specific) factor is continuously expressed in serotonin (5-HT) neurons and initially acts in postmitotic precursors to control acquisition of 5-HT transmitter identity. Using a combination of RNA sequencing, electrophysiology, and conditional targeting approaches, we determined gene expression patterns in maturing flow-sorted 5-HT neurons and the temporal requirements for Pet-1 in shaping these patterns for functional maturation of mouse 5-HT neurons. We report a profound disruption of postmitotic expression trajectories in Pet-1(-/-) neurons, which prevented postnatal maturation of 5-HT neuron passive and active intrinsic membrane properties, G-protein signaling, and synaptic responses to glutamatergic, lysophosphatidic, and adrenergic agonists. Unexpectedly, conditional targeting revealed a postnatal stage-specific switch in Pet-1 targets from 5-HT synthesis genes to transmitter receptor genes required for afferent modulation of 5-HT neuron excitability. Five-HT1a autoreceptor expression depended transiently on Pet-1, thus revealing an early postnatal sensitive period for control of 5-HT excitability genes. Chromatin immunoprecipitation followed by sequencing revealed that Pet-1 regulates 5-HT neuron maturation through direct gene activation and repression. Moreover, Pet-1 directly regulates the 5-HT neuron maturation factor Engrailed 1, which suggests Pet-1 orchestrates maturation through secondary postmitotic regulatory factors. The early postnatal switch in Pet-1 targets uncovers a distinct neonatal stage-specific function for Pet-1, during which it promotes maturation of 5-HT neuron excitability. SIGNIFICANCE STATEMENT The regulatory mechanisms that control functional maturation of neurons are poorly understood. We show that in addition to inducing brain serotonin (5-HT) synthesis and reuptake, the Pet-1 ETS (E26 transformation-specific) factor subsequently globally coordinates postmitotic expression trajectories of genes necessary for maturation of 5-HT neuron excitability. Further, Pet-1 switches its transcriptional targets as 5-HT neurons mature from 5-HT synthesis genes to G-protein-coupled receptors, which are necessary for afferent synaptic modulation of 5-HT neuron excitability. Our findings uncover gene-specific switching of downstream targets as a previously unrecognized regulatory strategy through which continuously expressed transcription factors control acquisition of neuronal identity at different stages of development.
Collapse
|
37
|
LoVerso PR, Cui F. Cell type-specific transcriptome profiling in mammalian brains. Front Biosci (Landmark Ed) 2016; 21:973-85. [PMID: 27100485 DOI: 10.2741/4434] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A mammalian brain contains numerous types of cells. Advances in neuroscience in the past decade allow us to identify and isolate neural cells of interest from mammalian brains. Recent developments in high-throughput technologies, such as microarrays and next-generation sequencing (NGS), provide detailed information on gene expression in pooled cells on a genomic scale. As a result, many novel genes have been found critical in cell type-specific transcriptional regulation. These differentially expressed genes can be used as molecular signatures, unique to a particular class of neural cells. Use of this gene expression-based approach can further differentiate neural cell types into subtypes, potentially linking some of them with neurological diseases. In this article, experimental techniques used to purify neural cells are described, followed by a review on recent microarray- or NGS-based transcriptomic studies of common neural cell types. The future prospects of cell type-specific research are also discussed.
Collapse
Affiliation(s)
- Peter R LoVerso
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, One Lomb Memorial Dr., Rochester, NY 14623
| | - Feng Cui
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, One Lomb Memorial Dr., Rochester, NY 14623,
| |
Collapse
|
38
|
Kouwenhoven WM, Veenvliet JV, van Hooft JA, van der Heide LP, Smidt MP. Engrailed 1 shapes the dopaminergic and serotonergic landscape through proper isthmic organizer maintenance and function. Biol Open 2016; 5:279-88. [PMID: 26879466 PMCID: PMC4810741 DOI: 10.1242/bio.015032] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The isthmic organizer (IsO) is a signaling center that specifies the correct and distinct embryonic development of the dopaminergic midbrain and serotonergic hindbrain. The IsO is a linear boundary between the two brain regions, emerging at around embryonic day 7-8 of murine embryonic development, that shapes its surroundings through the expression of instructive signals such as Wnt and growth factors. Homeobox transcription factor engrailed 1 (En1) is present in midbrain and rostral hindbrain (i.e. rhombomere 1, R1). Its expression spans the IsO, and it is known to be an important survival factor for both dopaminergic and serotonergic neurons. Erroneous composition of dopaminergic neurons in the midbrain or serotonergic neurons in the hindbrain is associated with severe pathologies such as Parkinson's disease, depression or autism. Here we investigated the role of En1 in early mid-hindbrain development, using multiple En1-ablated mouse models as well as lineage-tracing techniques, and observed the appearance of ectopic dopaminergic neurons, indistinguishable from midbrain dopaminergic neurons based on molecular profile and intrinsic electrophysiological properties. We propose that this change is the direct result of a caudal relocation of the IsO as represented by ectopic presence of Fgf8, Otx2, Wnt1 and canonical Wnt-signalling. Our work suggests a newly-discovered role for En1: the repression of Otx2, Wnt1 and canonical Wnt-signaling in R1. Overall, our results suggest that En1 is essential for proper IsO maintenance and function. Summary: Local molecular coding under the influence of En1 is essential for proper spatiotemporal expression of key factors involved in the maintenance and function of the isthmic organizer.
Collapse
Affiliation(s)
- Willemieke M Kouwenhoven
- Swammerdam Institute for Life Sciences, University of Amsterdam, P.O. Box 94215, 1090 GE Amsterdam, The Netherlands
| | - Jesse V Veenvliet
- Swammerdam Institute for Life Sciences, University of Amsterdam, P.O. Box 94215, 1090 GE Amsterdam, The Netherlands
| | - Johannes A van Hooft
- Swammerdam Institute for Life Sciences, University of Amsterdam, P.O. Box 94215, 1090 GE Amsterdam, The Netherlands
| | - L P van der Heide
- Swammerdam Institute for Life Sciences, University of Amsterdam, P.O. Box 94215, 1090 GE Amsterdam, The Netherlands
| | - Marten P Smidt
- Swammerdam Institute for Life Sciences, University of Amsterdam, P.O. Box 94215, 1090 GE Amsterdam, The Netherlands
| |
Collapse
|
39
|
Abstract
Serotonergic (5HT) neurons exert diverse and widespread functions in the brain. Dysfunction of the serotonergic system gives rise to a variety of mental illnesses including depression, anxiety, obsessive compulsive disorder, autism and eating disorders. Here we show that human primary fibroblasts were directly converted to induced serotonergic (i5HT) neurons by the expression of Ascl1, Foxa2, Lmx1b and FEV. The transdifferentiation was enhanced by p53 knockdown and appropriate culture conditions including hypoxia. The i5HT neurons expressed markers for mature serotonergic neurons, had Ca(2+)-dependent 5HT release and selective 5HT uptake, exhibited spontaneous action potentials and spontaneous excitatory postsynaptic currents. Application of serotonin significantly increased the firing rate of spontaneous action potentials, demonstrating the functional utility of i5HT neurons for studying serotonergic neurotransmission. The availability of human i5HT neurons will be very useful for research and drug discovery on many serotonin-related mental disorders.
Collapse
Affiliation(s)
- Z Xu
- Department of Neurology and Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - H Jiang
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - P Zhong
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Z Yan
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - S Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Neurology and Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China. E-mail:
| | - J Feng
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA,Department of Physiology and Biophysics, State University of New York at Buffalo, 124 Sherman Hall, Buffalo, NY 14214, USA E-mail:
| |
Collapse
|
40
|
Hu H, Haas SA, Chelly J, Van Esch H, Raynaud M, de Brouwer APM, Weinert S, Froyen G, Frints SGM, Laumonnier F, Zemojtel T, Love MI, Richard H, Emde AK, Bienek M, Jensen C, Hambrock M, Fischer U, Langnick C, Feldkamp M, Wissink-Lindhout W, Lebrun N, Castelnau L, Rucci J, Montjean R, Dorseuil O, Billuart P, Stuhlmann T, Shaw M, Corbett MA, Gardner A, Willis-Owen S, Tan C, Friend KL, Belet S, van Roozendaal KEP, Jimenez-Pocquet M, Moizard MP, Ronce N, Sun R, O'Keeffe S, Chenna R, van Bömmel A, Göke J, Hackett A, Field M, Christie L, Boyle J, Haan E, Nelson J, Turner G, Baynam G, Gillessen-Kaesbach G, Müller U, Steinberger D, Budny B, Badura-Stronka M, Latos-Bieleńska A, Ousager LB, Wieacker P, Rodríguez Criado G, Bondeson ML, Annerén G, Dufke A, Cohen M, Van Maldergem L, Vincent-Delorme C, Echenne B, Simon-Bouy B, Kleefstra T, Willemsen M, Fryns JP, Devriendt K, Ullmann R, Vingron M, Wrogemann K, Wienker TF, Tzschach A, van Bokhoven H, Gecz J, Jentsch TJ, Chen W, Ropers HH, Kalscheuer VM. X-exome sequencing of 405 unresolved families identifies seven novel intellectual disability genes. Mol Psychiatry 2016; 21:133-48. [PMID: 25644381 PMCID: PMC5414091 DOI: 10.1038/mp.2014.193] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 11/17/2014] [Accepted: 12/08/2014] [Indexed: 12/27/2022]
Abstract
X-linked intellectual disability (XLID) is a clinically and genetically heterogeneous disorder. During the past two decades in excess of 100 X-chromosome ID genes have been identified. Yet, a large number of families mapping to the X-chromosome remained unresolved suggesting that more XLID genes or loci are yet to be identified. Here, we have investigated 405 unresolved families with XLID. We employed massively parallel sequencing of all X-chromosome exons in the index males. The majority of these males were previously tested negative for copy number variations and for mutations in a subset of known XLID genes by Sanger sequencing. In total, 745 X-chromosomal genes were screened. After stringent filtering, a total of 1297 non-recurrent exonic variants remained for prioritization. Co-segregation analysis of potential clinically relevant changes revealed that 80 families (20%) carried pathogenic variants in established XLID genes. In 19 families, we detected likely causative protein truncating and missense variants in 7 novel and validated XLID genes (CLCN4, CNKSR2, FRMPD4, KLHL15, LAS1L, RLIM and USP27X) and potentially deleterious variants in 2 novel candidate XLID genes (CDK16 and TAF1). We show that the CLCN4 and CNKSR2 variants impair protein functions as indicated by electrophysiological studies and altered differentiation of cultured primary neurons from Clcn4(-/-) mice or after mRNA knock-down. The newly identified and candidate XLID proteins belong to pathways and networks with established roles in cognitive function and intellectual disability in particular. We suggest that systematic sequencing of all X-chromosomal genes in a cohort of patients with genetic evidence for X-chromosome locus involvement may resolve up to 58% of Fragile X-negative cases.
Collapse
Affiliation(s)
- H Hu
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - S A Haas
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - J Chelly
- University Paris Descartes, Paris, France,Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Institut National de la Santé et de la Recherche Médicale Unité 1016, Institut Cochin, Paris, France
| | - H Van Esch
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - M Raynaud
- Inserm U930 ‘Imaging and Brain', Tours, France,University François-Rabelais, Tours, France,Centre Hospitalier Régional Universitaire, Service de Génétique, Tours, France
| | - A P M de Brouwer
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - S Weinert
- Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany,Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - G Froyen
- Human Genome Laboratory, VIB Center for the Biology of Disease, Leuven, Belgium,Human Genome Laboratory, Department of Human Genetics, K.U. Leuven, Leuven, Belgium
| | - S G M Frints
- Department of Clinical Genetics, Maastricht University Medical Center, azM, Maastricht, The Netherlands,School for Oncology and Developmental Biology, GROW, Maastricht University, Maastricht, The Netherlands
| | - F Laumonnier
- Inserm U930 ‘Imaging and Brain', Tours, France,University François-Rabelais, Tours, France
| | - T Zemojtel
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - M I Love
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - H Richard
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - A-K Emde
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - M Bienek
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - C Jensen
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - M Hambrock
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - U Fischer
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - C Langnick
- Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany
| | - M Feldkamp
- Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany
| | - W Wissink-Lindhout
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - N Lebrun
- University Paris Descartes, Paris, France,Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Institut National de la Santé et de la Recherche Médicale Unité 1016, Institut Cochin, Paris, France
| | - L Castelnau
- University Paris Descartes, Paris, France,Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Institut National de la Santé et de la Recherche Médicale Unité 1016, Institut Cochin, Paris, France
| | - J Rucci
- University Paris Descartes, Paris, France,Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Institut National de la Santé et de la Recherche Médicale Unité 1016, Institut Cochin, Paris, France
| | - R Montjean
- University Paris Descartes, Paris, France,Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Institut National de la Santé et de la Recherche Médicale Unité 1016, Institut Cochin, Paris, France
| | - O Dorseuil
- University Paris Descartes, Paris, France,Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Institut National de la Santé et de la Recherche Médicale Unité 1016, Institut Cochin, Paris, France
| | - P Billuart
- University Paris Descartes, Paris, France,Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Institut National de la Santé et de la Recherche Médicale Unité 1016, Institut Cochin, Paris, France
| | - T Stuhlmann
- Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany,Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - M Shaw
- School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, SA, Australia,Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - M A Corbett
- School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, SA, Australia,Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - A Gardner
- School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, SA, Australia,Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - S Willis-Owen
- School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, SA, Australia,National Heart and Lung Institute, Imperial College London, London, UK
| | - C Tan
- School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, SA, Australia
| | - K L Friend
- SA Pathology, Women's and Children's Hospital, Adelaide, SA, Australia
| | - S Belet
- Human Genome Laboratory, VIB Center for the Biology of Disease, Leuven, Belgium,Human Genome Laboratory, Department of Human Genetics, K.U. Leuven, Leuven, Belgium
| | - K E P van Roozendaal
- Department of Clinical Genetics, Maastricht University Medical Center, azM, Maastricht, The Netherlands,School for Oncology and Developmental Biology, GROW, Maastricht University, Maastricht, The Netherlands
| | - M Jimenez-Pocquet
- Centre Hospitalier Régional Universitaire, Service de Génétique, Tours, France
| | - M-P Moizard
- Inserm U930 ‘Imaging and Brain', Tours, France,University François-Rabelais, Tours, France,Centre Hospitalier Régional Universitaire, Service de Génétique, Tours, France
| | - N Ronce
- Inserm U930 ‘Imaging and Brain', Tours, France,University François-Rabelais, Tours, France,Centre Hospitalier Régional Universitaire, Service de Génétique, Tours, France
| | - R Sun
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - S O'Keeffe
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - R Chenna
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - A van Bömmel
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - J Göke
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - A Hackett
- Genetics of Learning and Disability Service, Hunter Genetics, Waratah, NSW, Australia
| | - M Field
- Genetics of Learning and Disability Service, Hunter Genetics, Waratah, NSW, Australia
| | - L Christie
- Genetics of Learning and Disability Service, Hunter Genetics, Waratah, NSW, Australia
| | - J Boyle
- Genetics of Learning and Disability Service, Hunter Genetics, Waratah, NSW, Australia
| | - E Haan
- School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, SA, Australia,SA Pathology, Women's and Children's Hospital, Adelaide, SA, Australia
| | - J Nelson
- Genetic Services of Western Australia, King Edward Memorial Hospital, Perth, WA, Australia
| | - G Turner
- Genetics of Learning and Disability Service, Hunter Genetics, Waratah, NSW, Australia
| | - G Baynam
- Genetic Services of Western Australia, King Edward Memorial Hospital, Perth, WA, Australia,School of Paediatrics and Child Health, University of Western Australia, Perth, WA, Australia,Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia,Telethon Kids Institute, Perth, WA, Australia
| | | | - U Müller
- Institut für Humangenetik, Justus-Liebig-Universität Giessen, Giessen, Germany,bio.logis Center for Human Genetics, Frankfurt a. M., Germany
| | - D Steinberger
- Institut für Humangenetik, Justus-Liebig-Universität Giessen, Giessen, Germany,bio.logis Center for Human Genetics, Frankfurt a. M., Germany
| | - B Budny
- Chair and Department of Endocrinology, Metabolism and Internal Diseases, Ponzan University of Medical Sciences, Poznan, Poland
| | - M Badura-Stronka
- Chair and Department of Medical Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | - A Latos-Bieleńska
- Chair and Department of Medical Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | - L B Ousager
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - P Wieacker
- Institut für Humangenetik, Universitätsklinikum Münster, Muenster, Germany
| | | | - M-L Bondeson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - G Annerén
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - A Dufke
- Institut für Medizinische Genetik und Angewandte Genomik, Tübingen, Germany
| | - M Cohen
- Kinderzentrum München, München, Germany
| | - L Van Maldergem
- Centre de Génétique Humaine, Université de Franche-Comté, Besançon, France
| | - C Vincent-Delorme
- Service de Génétique, Hôpital Jeanne de Flandre CHRU de Lilles, Lille, France
| | - B Echenne
- Service de Neuro-Pédiatrie, CHU Montpellier, Montpellier, France
| | - B Simon-Bouy
- Laboratoire SESEP, Centre hospitalier de Versailles, Le Chesnay, France
| | - T Kleefstra
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - M Willemsen
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - J-P Fryns
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - K Devriendt
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - R Ullmann
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - M Vingron
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - K Wrogemann
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany,Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | - T F Wienker
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - A Tzschach
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - H van Bokhoven
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - J Gecz
- School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, SA, Australia,Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - T J Jentsch
- Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany,Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - W Chen
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany,Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany
| | - H-H Ropers
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - V M Kalscheuer
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany,Max Planck Institute for Molecular Genetics, Ihnestrasse 73, Berlin 14195, Germany. E-mail:
| |
Collapse
|
41
|
Griggs JL, Sinnayah P, Mathai ML. Prader–Willi syndrome: From genetics to behaviour, with special focus on appetite treatments. Neurosci Biobehav Rev 2015; 59:155-72. [DOI: 10.1016/j.neubiorev.2015.10.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 09/30/2015] [Accepted: 10/12/2015] [Indexed: 12/22/2022]
|
42
|
Fernandez SP, Cauli B, Cabezas C, Muzerelle A, Poncer JC, Gaspar P. Multiscale single-cell analysis reveals unique phenotypes of raphe 5-HT neurons projecting to the forebrain. Brain Struct Funct 2015; 221:4007-4025. [PMID: 26608830 DOI: 10.1007/s00429-015-1142-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/02/2015] [Indexed: 11/28/2022]
Abstract
Serotonergic neurons of the raphe nuclei exhibit anatomical, neurochemical and elecrophysiological heterogeneity that likely underpins their specific role in multiple behaviors. However, the precise organization of serotonin (5-HT) neurons to orchestrate 5-HT release patterns throughout the brain is not well understood. We compared the electrophysiological and neurochemical properties of dorsal and median raphe 5-HT neurons projecting to the medial prefrontal cortex (mPFC), amygdala (BLA) and dorsal hippocampus (dHP), combining retrograde tract tracing with brain slice electrophysiology and single-cell RT-PCR in Pet1-EGFP mice. Our results show that 5-HT neurons projecting to the dHP and the mPFC and the BLA form largely non-overlapping populations and that BLA-projecting neurons have characteristic excitability and membrane properties. In addition, using an unbiased clustering method that correlates anatomical, molecular and electrophysiological phenotypes, we find that 5-HT neurons with projections to the mPFC and the dHP segregate from those projecting to the BLA. Single-cell gene profiling showed a restricted expression of the peptide galanin in the population of 5-HT neurons projecting to the mPFC. Finally, cluster analysis allowed identifying an atypical subtype of 5-HT neuron with low excitability, long firing delays and preferential expression of the vesicular glutamate transporter type 3. Overall, these findings allow to define correlated anatomical and physiological identities of serotonin raphe neurons that help understanding how discrete raphe cells subpopulations account for the heterogeneous activities of the midbrain serotonergic system.
Collapse
Affiliation(s)
- Sebastian Pablo Fernandez
- Institut du Fer à Moulin, INSERM U839, 17 rue du Fer à Moulin, 75005, Paris, France. .,Université Pierre et Marie Curie, Paris, France. .,Institut du Fer a Moulin, Paris, France.
| | - Bruno Cauli
- Université Pierre et Marie Curie, Paris, France.,CNRS, UMR 8246, Neuroscience Paris Seine, 75005, Paris, France.,Inserm UMR-S 1130, Neuroscience Paris Seine, 75005, Paris, France
| | - Carolina Cabezas
- Institut du Fer à Moulin, INSERM U839, 17 rue du Fer à Moulin, 75005, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Institut du Fer a Moulin, Paris, France
| | - Aude Muzerelle
- Institut du Fer à Moulin, INSERM U839, 17 rue du Fer à Moulin, 75005, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Institut du Fer a Moulin, Paris, France
| | - Jean-Christophe Poncer
- Institut du Fer à Moulin, INSERM U839, 17 rue du Fer à Moulin, 75005, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Institut du Fer a Moulin, Paris, France
| | - Patricia Gaspar
- Institut du Fer à Moulin, INSERM U839, 17 rue du Fer à Moulin, 75005, Paris, France. .,Université Pierre et Marie Curie, Paris, France. .,Institut du Fer a Moulin, Paris, France.
| |
Collapse
|
43
|
Okaty BW, Freret ME, Rood BD, Brust RD, Hennessy ML, deBairos D, Kim JC, Cook MN, Dymecki SM. Multi-Scale Molecular Deconstruction of the Serotonin Neuron System. Neuron 2015; 88:774-91. [PMID: 26549332 DOI: 10.1016/j.neuron.2015.10.007] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 08/06/2015] [Accepted: 09/28/2015] [Indexed: 02/01/2023]
Abstract
Serotonergic (5HT) neurons modulate diverse behaviors and physiology and are implicated in distinct clinical disorders. Corresponding diversity in 5HT neuronal phenotypes is becoming apparent and is likely rooted in molecular differences, yet a comprehensive approach characterizing molecular variation across the 5HT system is lacking, as is concomitant linkage to cellular phenotypes. Here we combine intersectional fate mapping, neuron sorting, and genome-wide RNA-seq to deconstruct the mouse 5HT system at multiple levels of granularity-from anatomy, to genetic sublineages, to single neurons. Our unbiased analyses reveal principles underlying system organization, 5HT neuron subtypes, constellations of differentially expressed genes distinguishing subtypes, and predictions of subtype-specific functions. Using electrophysiology, subtype-specific neuron silencing, and conditional gene knockout, we show that these molecularly defined 5HT neuron subtypes are functionally distinct. Collectively, this resource classifies molecular diversity across the 5HT system and discovers sertonergic subtypes, markers, organizing principles, and subtype-specific functions with potential disease relevance.
Collapse
Affiliation(s)
- Benjamin W Okaty
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Morgan E Freret
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Benjamin D Rood
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Rachael D Brust
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Morgan L Hennessy
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Danielle deBairos
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Jun Chul Kim
- Psychology Department, University of Toronto, 100 St. George Street, Toronto ON, M5S 3G3, Canada
| | - Melloni N Cook
- Department of Psychology, University of Memphis, 400 Innovation Drive, Memphis, TN 38152, USA
| | - Susan M Dymecki
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| |
Collapse
|
44
|
Andrade R, Huereca D, Lyons JG, Andrade EM, McGregor KM. 5-HT1A Receptor-Mediated Autoinhibition and the Control of Serotonergic Cell Firing. ACS Chem Neurosci 2015; 6:1110-5. [PMID: 25913021 DOI: 10.1021/acschemneuro.5b00034] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The idea that serotonergic synaptic transmission plays an essential role in the control of mood and the pharmacotherapy of anxiety and depression is one of the cornerstones of modern biological psychiatry. As a result, there is intense interest in understanding the mechanisms controlling the activity of serotonin-synthesizing (serotonergic) neurons. One of the oldest and most durable ideas emerging from this work is that serotonergic neurons are capable of autonomously regulating their own basal firing rate. Serotonergic neurons express on their surface 5-HT1A receptors (autoreceptors) that, when activated, induce the opening of potassium channels that hyperpolarize and thereby inhibit cell firing. Activity-dependent release of serotonin within serotonergic nuclei is thought to activate these autoreceptors, thus completing an autoinhibitory feedback loop. This concept, which was originally proposed in the 1970s, has proven to be enormously fruitful and has guided the interpretation of a broad range of clinical and preclinical work. Yet, remarkably, electrophysiological studies seeking to directly demonstrate this phenomenon, especially in in vitro brain slices, have produced mixed results. Here, we critically review this work with a focus on electrophysiological studies, which directly assess neuronal activity. We also highlight recent work suggesting that 5-HT1A receptor-mediated autoinhibition may play other roles in the control of firing besides acting as a feedback regulator for the pacemaker-like firing rate of serotonergic neurons.
Collapse
Affiliation(s)
- Rodrigo Andrade
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Daniel Huereca
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Joseph G. Lyons
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Elaine M. Andrade
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Kelly M. McGregor
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| |
Collapse
|
45
|
Hainer C, Mosienko V, Koutsikou S, Crook JJ, Gloss B, Kasparov S, Lumb BM, Alenina N. Beyond Gene Inactivation: Evolution of Tools for Analysis of Serotonergic Circuitry. ACS Chem Neurosci 2015; 6:1116-29. [PMID: 26132472 DOI: 10.1021/acschemneuro.5b00045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In the brain, serotonin (5-hydroxytryptamine, 5-HT) controls a multitude of physiological and behavioral functions. Serotonergic neurons in the raphe nuclei give rise to a complex and extensive network of axonal projections throughout the whole brain. A major challenge in the analysis of these circuits is to understand how the serotonergic networks are linked to the numerous functions of this neurotransmitter. In the past, many studies employed approaches to inactivate different genes involved in serotonergic neuron formation, 5-HT transmission, or 5-HT metabolism. Although these approaches have contributed significantly to our understanding of serotonergic circuits, they usually result in life-long gene inactivation. As a consequence, compensatory changes in serotonergic and other neurotransmitter systems may occur and complicate the interpretation of the observed phenotypes. To dissect the complexity of the serotonergic system with greater precision, approaches to reversibly manipulate subpopulations of serotonergic neurons are required. In this review, we summarize findings on genetic animal models that enable control of 5-HT neuronal activity or mapping of the serotonergic system. This includes a comparative analysis of several mouse and rat lines expressing Cre or Flp recombinases under Tph2, Sert, or Pet1 promoters with a focus on specificity and recombination efficiency. We further introduce applications for Cre-mediated cell-type specific gene expression to optimize spatial and temporal precision for the manipulation of serotonergic neurons. Finally, we discuss other temporally regulated systems, such as optogenetics and designer receptors exclusively activated by designer drugs (DREADD) approaches to control 5-HT neuron activity.
Collapse
Affiliation(s)
- Cornelia Hainer
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin 13125, Germany
| | | | | | | | - Bernd Gloss
- National Institute of Environmental Health Science, Durham, North Carolina 27709, United States
| | | | | | - Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin 13125, Germany
- Institute
of Translational Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia
| |
Collapse
|
46
|
Wyler SC, Donovan LJ, Yeager M, Deneris E. Pet-1 Controls Tetrahydrobiopterin Pathway and Slc22a3 Transporter Genes in Serotonin Neurons. ACS Chem Neurosci 2015; 6:1198-205. [PMID: 25642596 PMCID: PMC4504805 DOI: 10.1021/cn500331z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Coordinated serotonin (5-HT) synthesis and reuptake depends on coexpression of Tph2, Aadc (Ddc), and Sert (Slc6a4) in brain 5-HT neurons. However, other gene products play critical roles in brain 5-HT synthesis and transport. For example, 5-HT synthesis depends on coexpression of genes encoding the enzymatic machinery necessary for the production and regeneration of tetrahydrobiopterin (BH4). In addition, the organic cation transporter 3 (Oct3, Slc22a3) functions as a low affinity, high capacity 5-HT reuptake protein in 5-HT neurons. The regulatory strategies controlling BH4 and Oct3 gene expression in 5-HT neurons have not been investigated. Our previous studies showed that Pet-1 is a critical transcription factor in a regulatory program that controls coexpression of Tph2, Aadc, and Sert in 5-HT neurons. Here, we investigate whether a common regulatory program determines global 5-HT synthesis and reuptake through coordinate transcriptional control. We show with comparative microarray profiling of flow sorted YFP(+) Pet-1(-/-) and wild type 5-HT neurons that Pet-1 regulates BH4 pathway genes, Gch1, Gchfr, and Qdpr. Thus, Pet-1 coordinates expression of all rate-limiting enzymatic (Tph2, Gch1) and post-translational regulatory (Gchfr) steps that determine the level of mammalian brain 5-HT synthesis. Moreover, Pet-1 globally controls acquisition of 5-HT reuptake in dorsal raphe 5-HT neurons by coordinating expression of Slc6a4 and Slc22a3. In situ hybridizations revealed that virtually all 5-HT neurons in the dorsal raphe depend on Pet-1 for Slc22a3 expression; similar results were obtained for Htr1a. Therefore, few if any 5-HT neurons in the dorsal raphe are resistant to loss of Pet-1 for their full neuron-type identity.
Collapse
Affiliation(s)
| | | | - Mia Yeager
- Department of Neurosciences, Case Western Reserve University Cleveland, Ohio, 44106, United States
| | - Evan Deneris
- Department of Neurosciences, Case Western Reserve University Cleveland, Ohio, 44106, United States
| |
Collapse
|
47
|
Jochems J, Teegarden SL, Chen Y, Boulden J, Challis C, Ben-Dor GA, Kim SF, Berton O. Enhancement of stress resilience through histone deacetylase 6-mediated regulation of glucocorticoid receptor chaperone dynamics. Biol Psychiatry 2015; 77:345-55. [PMID: 25442004 PMCID: PMC4297530 DOI: 10.1016/j.biopsych.2014.07.036] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 07/29/2014] [Accepted: 07/29/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Acetylation of heat shock protein 90 (Hsp90) regulates downstream hormone signaling via the glucocorticoid receptor (GR), but the role of this molecular mechanism in stress homeostasis is poorly understood. We tested whether acetylation of Hsp90 in the brain predicts and modulates the behavioral sequelae of a mouse model of social stress. METHODS Mice subjected to chronic social defeat stress were stratified into resilient and vulnerable subpopulations. Hypothalamic-pituitary-adrenal axis function was probed using a dexamethasone/corticotropin-releasing factor test. Measurements of Hsp90 acetylation, Hsp90-GR interactions, and GR translocation were performed in the dorsal raphe nucleus. To manipulate Hsp90 acetylation, we pharmacologically inhibited histone deacetylase 6, a known deacetylase of Hsp90, or overexpressed a point mutant that mimics the hyperacetylated state of Hsp90 at lysine K294. RESULTS Lower acetylated Hsp90, higher GR-Hsp90 association, and enhanced GR translocation were observed in dorsal raphe nucleus of vulnerable mice after chronic social defeat stress. Administration of ACY-738, a histone deacetylase 6-selective inhibitor, led to Hsp90 hyperacetylation in brain and in neuronal culture. In cell-based assays, ACY-738 increased the relative association of Hsp90 with FK506 binding protein 51 versus FK506 binding protein 52 and inhibited hormone-induced GR translocation. This effect was replicated by overexpressing the acetylation-mimic point mutant of Hsp90. In vivo, ACY-738 promoted resilience to chronic social defeat stress, and serotonin-selective viral overexpression of the acetylation-mimic mutant of Hsp90 in raphe neurons reproduced the behavioral effect of ACY-738. CONCLUSIONS Hyperacetylation of Hsp90 is a predictor and causal molecular determinant of stress resilience in mice. Brain-penetrant histone deacetylase 6 inhibitors increase Hsp90 acetylation and modulate GR chaperone dynamics offering a promising strategy to curtail deleterious socioaffective effects of stress and glucocorticoids.
Collapse
Affiliation(s)
- Jeanine Jochems
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia PA 19104-3403
| | - Sarah L Teegarden
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia PA 19104-3403
| | - Yong Chen
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia PA 19104-3403
| | - Janette Boulden
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia PA 19104-3403
| | - Collin Challis
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia PA 19104-3403
| | - Gabriel A Ben-Dor
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia PA 19104-3403
| | - Sangwon F Kim
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia PA 19104-3403
| | - Olivier Berton
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia Pennsylvania..
| |
Collapse
|
48
|
Rogers SM, Ott SR. Differential activation of serotonergic neurons during short- and long-term gregarization of desert locusts. Proc Biol Sci 2015; 282:20142062. [PMID: 25520357 PMCID: PMC4298206 DOI: 10.1098/rspb.2014.2062] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Serotonin is a neurochemical with evolutionarily conserved roles in orchestrating nervous system function and behavioural plasticity. A dramatic example is the rapid transformation of desert locusts from cryptic asocial animals into gregarious crop pests that occurs when drought forces them to accumulate on dwindling resources, triggering a profound alteration of behaviour within just a few hours. The onset of crowding induces a surge in serotonin within their thoracic ganglia that is sufficient and necessary to induce the switch from solitarious to gregarious behaviour. To identify the neurons responsible, we have analysed how acute exposure to three gregarizing stimuli--crowding, touching the hind legs or seeing and smelling other locusts--and prolonged group living affect the expression of serotonin in individual neurons in the thoracic ganglia. Quantitative analysis of cell body immunofluorescence revealed three classes of neurons with distinct expressional responses. All ganglia contained neurons that responded to multiple gregarizing stimuli with increased expression. A second class showed increased expression only in response to intense visual and olfactory stimuli from conspecifics. Prolonged group living affected a third and entirely different set of neurons, revealing a two-tiered role of the serotonergic system as both initiator and substrate of socially induced plasticity. This demonstrates the critical importance of ontogenetic time for understanding the function of serotonin in the reorganization of behaviour.
Collapse
Affiliation(s)
- Stephen M Rogers
- School of Biological Sciences, University of Sydney, A08 Heydon-Laurence Building, New South Wales 2006, Australia
| | - Swidbert R Ott
- Department of Biology, University of Leicester, Adrian Building, University Road, Leicester LE1 7RH, UK
| |
Collapse
|
49
|
Chiu IM, Barrett LB, Williams EK, Strochlic DE, Lee S, Weyer AD, Lou S, Bryman GS, Roberson DP, Ghasemlou N, Piccoli C, Ahat E, Wang V, Cobos EJ, Stucky CL, Ma Q, Liberles SD, Woolf CJ. Transcriptional profiling at whole population and single cell levels reveals somatosensory neuron molecular diversity. eLife 2014; 3. [PMID: 25525749 PMCID: PMC4383053 DOI: 10.7554/elife.04660] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/18/2014] [Indexed: 12/17/2022] Open
Abstract
The somatosensory nervous system is critical for the organism's ability to respond to
mechanical, thermal, and nociceptive stimuli. Somatosensory neurons are functionally
and anatomically diverse but their molecular profiles are not well-defined. Here, we
used transcriptional profiling to analyze the detailed molecular signatures of dorsal
root ganglion (DRG) sensory neurons. We used two mouse reporter lines and surface IB4
labeling to purify three major non-overlapping classes of neurons: 1)
IB4+SNS-Cre/TdTomato+, 2)
IB4−SNS-Cre/TdTomato+, and 3)
Parv-Cre/TdTomato+ cells, encompassing the majority of
nociceptive, pruriceptive, and proprioceptive neurons. These neurons displayed
distinct expression patterns of ion channels, transcription factors, and GPCRs.
Highly parallel qRT-PCR analysis of 334 single neurons selected by membership of the
three populations demonstrated further diversity, with unbiased clustering analysis
identifying six distinct subgroups. These data significantly increase our knowledge
of the molecular identities of known DRG populations and uncover potentially novel
subsets, revealing the complexity and diversity of those neurons underlying
somatosensation. DOI:http://dx.doi.org/10.7554/eLife.04660.001 In the nervous system, a network of specialized neurons—known as the
somatosensory system—carries information about sensations including touch,
muscle position, temperature and pain. Distinct sets of somatosensory neurons are
thought to carry information about the different types of sensations. In young
animals, the precise switching on, or ‘expression’, of genes controls
the formation of the network of neurons. However, it is not known exactly which genes
are expressed in what types of neurons, where, or when. Here, Chiu et al. used a technique called flow cytometry using different fluorescent
markers to isolate a group of cells called Dorsal Root Ganglion (DRG) neurons in
mice. These neurons have long thread-like fibers that extend from the spinal cord to
the skin, muscles and joints all over the body. These fibers carry sensory
information to the spinal cord, where it can be relayed to the brain and processed.
The experiments compared three distinct types of DRG neuron and found that they
differed in their ability to send information to other cells. Chiu et al. analyzed the expression of all the genes in the three types of DRG
neurons. Each type of neuron had distinct groups of genes that were being expressed.
Also, several genes that are known to be important for sensation were expressed at
different levels in the different types of cells. Next, large numbers of single cells
were analyzed to find out the finer details about the three types of neuron. These
findings made it possible to further divide the DRG neurons into six distinct subsets
that matched previously known groups of somatosensory neurons, and also identified
new ones. Chiu et al.'s findings reveal the complexity and diversity of the neurons involved in
carrying information about sensations towards the brain. This is an important step in
classifying the nervous system, and uncovers many genes previously not linked to
sensation. The next challenges lie in understanding how the expression of these genes
in each type of neuron relates to their unique roles. DOI:http://dx.doi.org/10.7554/eLife.04660.002
Collapse
Affiliation(s)
- Isaac M Chiu
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States
| | - Lee B Barrett
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States
| | - Erika K Williams
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - David E Strochlic
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Seungkyu Lee
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States
| | - Andy D Weyer
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, United States
| | - Shan Lou
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, United States
| | - Gregory S Bryman
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States
| | - David P Roberson
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States
| | - Nader Ghasemlou
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States
| | - Cara Piccoli
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States
| | - Ezgi Ahat
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States
| | - Victor Wang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States
| | - Enrique J Cobos
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States
| | - Cheryl L Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, United States
| | - Qiufu Ma
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, United States
| | - Stephen D Liberles
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States
| |
Collapse
|
50
|
Functional and developmental identification of a molecular subtype of brain serotonergic neuron specialized to regulate breathing dynamics. Cell Rep 2014; 9:2152-65. [PMID: 25497093 DOI: 10.1016/j.celrep.2014.11.027] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 09/23/2014] [Accepted: 11/18/2014] [Indexed: 11/22/2022] Open
Abstract
Serotonergic neurons modulate behavioral and physiological responses from aggression and anxiety to breathing and thermoregulation. Disorders involving serotonin (5HT) dysregulation are commensurately heterogeneous and numerous. We hypothesized that this breadth in functionality derives in part from a developmentally determined substructure of distinct subtypes of 5HT neurons each specialized to modulate specific behaviors. By manipulating developmentally defined subgroups one by one chemogenetically, we find that the Egr2-Pet1 subgroup is specialized to drive increased ventilation in response to carbon dioxide elevation and acidosis. Furthermore, this subtype exhibits intrinsic chemosensitivity and modality-specific projections-increasing firing during hypercapnic acidosis and selectively projecting to respiratory chemosensory but not motor centers, respectively. These findings show that serotonergic regulation of the respiratory chemoreflex is mediated by a specialized molecular subtype of 5HT neuron harboring unique physiological, biophysical, and hodological properties specified developmentally and demonstrate that the serotonergic system contains specialized modules contributing to its collective functional breadth.
Collapse
|