1
|
Fiorenzano A, Storm P, Sozzi E, Bruzelius A, Corsi S, Kajtez J, Mudannayake J, Nelander J, Mattsson B, Åkerblom M, Björklund T, Björklund A, Parmar M. TARGET-seq: Linking single-cell transcriptomics of human dopaminergic neurons with their target specificity. Proc Natl Acad Sci U S A 2024; 121:e2410331121. [PMID: 39541349 DOI: 10.1073/pnas.2410331121] [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: 05/27/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Dopaminergic (DA) neurons exhibit significant diversity characterized by differences in morphology, anatomical location, axonal projection pattern, and selective vulnerability to disease. More recently, scRNAseq has been used to map DA neuron diversity at the level of gene expression. These studies have revealed a higher than expected molecular diversity in both mouse and human DA neurons. However, whether different molecular expression profiles correlate with specific functions of different DA neurons or with their classical division into mesolimbic (A10) and nigrostriatal (A9) neurons, remains to be determined. To address this, we have developed an approach termed TARGET-seq (Tagging projections by AAV-mediated RetroGrade Enrichment of Transcriptomes) that links the transcriptional profile of the DA neurons with their innervation of specific target structures in the forebrain. Leveraging this technology, we identify molecularly distinct subclusters of human DA neurons with a clear link between transcriptome and axonal target-specificity, offering the possibility to infer neuroanatomical-based classification to molecular identity and target-specific connectivity. We subsequently used this dataset to identify candidate transcription factors along DA developmental trajectories that may control subtype identity, thus providing broad avenues that can be further explored in the design of next-generation A9 and A10 enriched DA-neurons for drug screening or A9 enriched DA cells for clinical stem cell-based therapies.
Collapse
Affiliation(s)
- Alessandro Fiorenzano
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne 223 62, Sweden
| | - Petter Storm
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne 223 62, Sweden
| | - Edoardo Sozzi
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne 223 62, Sweden
| | - Andreas Bruzelius
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne 223 62, Sweden
| | - Sara Corsi
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne 223 62, Sweden
| | - Janko Kajtez
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne 223 62, Sweden
| | - Janitha Mudannayake
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne 223 62, Sweden
| | - Jenny Nelander
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne 223 62, Sweden
| | - Bengt Mattsson
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne 223 62, Sweden
| | - Malin Åkerblom
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne 223 62, Sweden
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund Skåne 223 62, Sweden
| | - Tomas Björklund
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund Skåne 223 62, Sweden
| | - Anders Björklund
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne 223 62, Sweden
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne 223 62, Sweden
| |
Collapse
|
2
|
Gaertner Z, Oram C, Schneeweis A, Schonfeld E, Bolduc C, Chen C, Dombeck D, Parisiadou L, Poulin JF, Awatramani R. Molecular and spatial transcriptomic classification of midbrain dopamine neurons and their alterations in a LRRK2 G2019S model of Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597807. [PMID: 38895448 PMCID: PMC11185743 DOI: 10.1101/2024.06.06.597807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Several studies have revealed that midbrain dopamine (DA) neurons, even within a single neuroanatomical area, display heterogeneous properties. In parallel, studies using single cell profiling techniques have begun to cluster DA neurons into subtypes based on their molecular signatures. Recent work has shown that molecularly defined DA subtypes within the substantia nigra (SNc) display distinctive anatomic and functional properties, and differential vulnerability in Parkinson's disease (PD). Based on these provocative results, a granular understanding of these putative subtypes and their alterations in PD models, is imperative. We developed an optimized pipeline for single-nuclear RNA sequencing (snRNA-seq) and generated a high-resolution hierarchically organized map revealing 20 molecularly distinct DA neuron subtypes belonging to three main families. We integrated this data with spatial MERFISH technology to map, with high definition, the location of these subtypes in the mouse midbrain, revealing heterogeneity even within neuroanatomical sub-structures. Finally, we demonstrate that in the preclinical LRRK2G2019S knock-in mouse model of PD, subtype organization and proportions are preserved. Transcriptional alterations occur in many subtypes including those localized to the ventral tier SNc, where differential expression is observed in synaptic pathways, which might account for previously described DA release deficits in this model. Our work provides an advancement of current taxonomic schemes of the mouse midbrain DA neuron subtypes, a high-resolution view of their spatial locations, and their alterations in a prodromal mouse model of PD.
Collapse
Affiliation(s)
- Zachary Gaertner
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Cameron Oram
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and Neurosurgery, Montreal (QC), Canada
| | - Amanda Schneeweis
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Elan Schonfeld
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
| | - Cyril Bolduc
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and Neurosurgery, Montreal (QC), Canada
| | - Chuyu Chen
- Northwestern University Feinberg School of Medicine, Dept of Pharmacology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Daniel Dombeck
- Northwestern University, Dept of Neurobiology, Evanston, IL 60201
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Loukia Parisiadou
- Northwestern University Feinberg School of Medicine, Dept of Pharmacology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Jean Francois Poulin
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and Neurosurgery, Montreal (QC), Canada
| | - Rajeshwar Awatramani
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
3
|
Lee RS, Sagiv Y, Engelhard B, Witten IB, Daw ND. A feature-specific prediction error model explains dopaminergic heterogeneity. Nat Neurosci 2024; 27:1574-1586. [PMID: 38961229 DOI: 10.1038/s41593-024-01689-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/22/2024] [Indexed: 07/05/2024]
Abstract
The hypothesis that midbrain dopamine (DA) neurons broadcast a reward prediction error (RPE) is among the great successes of computational neuroscience. However, recent results contradict a core aspect of this theory: specifically that the neurons convey a scalar, homogeneous signal. While the predominant family of extensions to the RPE model replicates the classic model in multiple parallel circuits, we argue that these models are ill suited to explain reports of heterogeneity in task variable encoding across DA neurons. Instead, we introduce a complementary 'feature-specific RPE' model, positing that individual ventral tegmental area DA neurons report RPEs for different aspects of an animal's moment-to-moment situation. Further, we show how our framework can be extended to explain patterns of heterogeneity in action responses reported among substantia nigra pars compacta DA neurons. This theory reconciles new observations of DA heterogeneity with classic ideas about RPE coding while also providing a new perspective of how the brain performs reinforcement learning in high-dimensional environments.
Collapse
Affiliation(s)
- Rachel S Lee
- Princeton Neuroscience Institute, Princeton, NJ, USA
| | - Yotam Sagiv
- Princeton Neuroscience Institute, Princeton, NJ, USA
| | - Ben Engelhard
- Princeton Neuroscience Institute, Princeton, NJ, USA
| | | | - Nathaniel D Daw
- Princeton Neuroscience Institute, Princeton, NJ, USA.
- Department of Psychology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
4
|
Apuschkin M, Burm HB, Schmidt JH, Skov LJ, Andersen RC, Bowin CF, Støier JF, Jensen KL, Posselt LP, Dmytriyeva O, Sørensen AT, Egerod KL, Holst B, Rickhag M, Schwartz TW, Gether U. An atlas of GPCRs in dopamine neurons: Identification of the free fatty acid receptor 4 as a regulator of food and water intake. Cell Rep 2024; 43:114509. [PMID: 39003735 DOI: 10.1016/j.celrep.2024.114509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 04/03/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Midbrain dopaminergic neurons (DANs) are subject to extensive metabotropic regulation, but the repertoire of G protein-coupled receptors (GPCRs) present in these neurons has not been mapped. Here, we isolate DANs from Dat-eGFP mice to generate a GPCR atlas by unbiased qPCR array expression analysis of 377 GPCRs. Combined with data mining of scRNA-seq databases, we identify multiple receptors in DAN subpopulations with 38 of these receptors representing the majority of transcripts. We identify 41 receptors expressed in midbrain DANs but not in non-DAN midbrain cells, including the free fatty acid receptor 4 (FFAR4). Functional expression of FFAR4 is validated by ex vivo Ca2+ imaging, and in vivo experiments support that FFAR4 negatively regulates food and water intake and bodyweight. In addition to providing a critical framework for understanding metabotropic DAN regulation, our data suggest fatty acid sensing by FFAR4 as a mechanism linking high-energy intake to the dopamine-reward pathway.
Collapse
Affiliation(s)
- Mia Apuschkin
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Hayley B Burm
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jan H Schmidt
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Louise J Skov
- Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Rita C Andersen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Carl-Fredrik Bowin
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jonatan F Støier
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kathrine L Jensen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Leonie P Posselt
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Oksana Dmytriyeva
- Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Andreas T Sørensen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kristoffer L Egerod
- Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Birgitte Holst
- Department of Biomedical Sciences, Laboratory for Molecular Pharmacology and Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Mattias Rickhag
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; Danish Research Centre for Magnetic Resonance (DRCMR), Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Thue W Schwartz
- Novo Nordic Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ulrik Gether
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
5
|
Islam KUS, Blaess S. The impact of the mesoprefrontal dopaminergic system on the maturation of interneurons in the murine prefrontal cortex. Front Neurosci 2024; 18:1403402. [PMID: 39035778 PMCID: PMC11257905 DOI: 10.3389/fnins.2024.1403402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/20/2024] [Indexed: 07/23/2024] Open
Abstract
The prefrontal cortex (PFC) undergoes a protracted maturation process. This is true both for local interneurons and for innervation from midbrain dopaminergic (mDA) neurons. In the striatum, dopaminergic (DA) neurotransmission is required for the maturation of medium spiny neurons during a critical developmental period. To investigate whether DA innervation influences the maturation of interneurons in the PFC, we used a conditional knockout (cKO) mouse model in which innervation from mDA neurons to the mPFC (mesoprefrontal innnervation) is not established during development. In this mouse model, the maturation of parvalbumin (PV) and calbindin (CB) interneuron populations in the PFC is dysregulated during a critical period in adolescence with changes persisting into adulthood. PV interneurons are particularly vulnerable to lack of mesoprefrontal input, showing an inability to maintain adequate PV expression with a concomitant decrease in Gad1 expression levels. Interestingly, lack of mesoprefrontal innervation does not appear to induce compensatory changes such as upregulation of DA receptor expression in PFC neurons or increased innervation density of other neuromodulatory (serotonergic and noradrenergic) innervation. In conclusion, our study shows that adolescence is a sensitive period during which mesoprefrontal input plays a critical role in promoting the maturation of specific interneuron subgroups. The results of this study will help to understand how a dysregulated mesoprefrontal DA system contributes to the pathophysiology of neurodevelopmental disorders.
Collapse
Affiliation(s)
| | - Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
6
|
Hose L, Langenhagen AK, Kefalakes E, Schweitzer T, Kubinski S, Barak S, Pich A, Grothe C. A dual-omics approach on the effects of fibroblast growth factor-2 (FGF-2) on ventral tegmental area dopaminergic neurons in response to alcohol consumption in mice. Eur J Neurosci 2024; 59:1519-1535. [PMID: 38185886 DOI: 10.1111/ejn.16234] [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: 07/31/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 01/09/2024]
Abstract
Harmful alcohol consumption is a major socioeconomic burden to the health system, as it can be the cause of mortality of heavy alcohol drinkers. The dopaminergic (DAergic) system is thought to play an important role in the pathogenesis of alcohol drinking behaviour; however, its exact role remains elusive. Fibroblast growth factor 2 (FGF-2), a neurotrophic factor, associated with both the DAergic system and alcohol consumption, may play an important role in DAergic neuroadaptations during alcohol abuse. Within this study, we aimed to clarify the role of endogenous FGF-2 on the DAergic system and whether there is a possible link to alcohol consumption. We found that lack of FGF-2 reduces the alcohol intake of mice. Transcriptome analysis of DAergic neurons revealed that FGF-2 knockout (FGF-2 KO) shifts the molecular fingerprint of midbrain dopaminergic (mDA) neurons to DA subtypes of the ventral tegmental area (VTA). In line with this, proteomic changes predominantly appear also in the VTA. Interestingly, these changes led to an altered regulation of the FGF-2 signalling cascades and DAergic pathways in a region-specific manner, which was only marginally affected by voluntary alcohol consumption. Thus, lack of FGF-2 not only affects the gene expression but also the proteome of specific brain regions of mDA neurons. Our study provides new insights into the neuroadaptations of the DAergic system during alcohol abuse and, therefore, comprises novel targets for future pharmacological interventions.
Collapse
Affiliation(s)
- Leonie Hose
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Alina Katharina Langenhagen
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Hannover, Germany
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Ekaterini Kefalakes
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Hannover, Germany
| | - Theresa Schweitzer
- Institute of Toxicology, Hannover, Germany
- Core Facility Proteomics, Institute of Toxicology, Hannover, Germany
| | - Sabrina Kubinski
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Hannover, Germany
| | - Segev Barak
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Andreas Pich
- Institute of Toxicology, Hannover, Germany
- Core Facility Proteomics, Institute of Toxicology, Hannover, Germany
| | - Claudia Grothe
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
7
|
Yang L, Kim TW, Han Y, Nair MS, Harschnitz O, Zhu J, Wang P, Koo SY, Lacko LA, Chandar V, Bram Y, Zhang T, Zhang W, He F, Pan C, Wu J, Huang Y, Evans T, van der Valk P, Titulaer MJ, Spoor JKH, Furler O'Brien RL, Bugiani M, D J Van de Berg W, Schwartz RE, Ho DD, Studer L, Chen S. SARS-CoV-2 infection causes dopaminergic neuron senescence. Cell Stem Cell 2024; 31:196-211.e6. [PMID: 38237586 PMCID: PMC10843182 DOI: 10.1016/j.stem.2023.12.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 01/30/2024]
Abstract
COVID-19 patients commonly present with signs of central nervous system and/or peripheral nervous system dysfunction. Here, we show that midbrain dopamine (DA) neurons derived from human pluripotent stem cells (hPSCs) are selectively susceptible and permissive to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. SARS-CoV-2 infection of DA neurons triggers an inflammatory and cellular senescence response. High-throughput screening in hPSC-derived DA neurons identified several FDA-approved drugs that can rescue the cellular senescence phenotype by preventing SARS-CoV-2 infection. We also identified the inflammatory and cellular senescence signature and low levels of SARS-CoV-2 transcripts in human substantia nigra tissue of COVID-19 patients. Furthermore, we observed reduced numbers of neuromelanin+ and tyrosine-hydroxylase (TH)+ DA neurons and fibers in a cohort of severe COVID-19 patients. Our findings demonstrate that hPSC-derived DA neurons are susceptible to SARS-CoV-2, identify candidate neuroprotective drugs for COVID-19 patients, and suggest the need for careful, long-term monitoring of neurological problems in COVID-19 patients.
Collapse
Affiliation(s)
- Liuliu Yang
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Tae Wan Kim
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA; Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Yuling Han
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Manoj S Nair
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | | | - Jiajun Zhu
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Pengfei Wang
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - So Yeon Koo
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA; Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Neuroscience Graduate Program of Weill Cornell Graduate School of Biomedical Sciences, New York, NY, USA
| | - Lauretta A Lacko
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Vasuretha Chandar
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA; Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Yaron Bram
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA; Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Tuo Zhang
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Wei Zhang
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Feng He
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Chendong Pan
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Junjie Wu
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Yaoxing Huang
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Paul van der Valk
- Department of Pathology, Amsterdam University Medical Center, VU University Amsterdam, Amsterdam, the Netherlands
| | - Maarten J Titulaer
- Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jochem K H Spoor
- Department of Neurosurgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Robert L Furler O'Brien
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Marianna Bugiani
- Amsterdam UMC, Location Vrije Universiteit Amsterdam, Department of Pathology, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Wilma D J Van de Berg
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands; Amsterdam UMC, Location Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Robert E Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA; Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA.
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA.
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA; Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA.
| |
Collapse
|
8
|
Gomez Ramos B, Ohnmacht J, de Lange N, Valceschini E, Ginolhac A, Catillon M, Ferrante D, Rakovic A, Halder R, Massart F, Arena G, Antony P, Bolognin S, Klein C, Krause R, Schulz MH, Sauter T, Krüger R, Sinkkonen L. Multiomics analysis identifies novel facilitators of human dopaminergic neuron differentiation. EMBO Rep 2024; 25:254-285. [PMID: 38177910 PMCID: PMC10897179 DOI: 10.1038/s44319-023-00024-2] [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: 04/17/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 01/06/2024] Open
Abstract
Midbrain dopaminergic neurons (mDANs) control voluntary movement, cognition, and reward behavior under physiological conditions and are implicated in human diseases such as Parkinson's disease (PD). Many transcription factors (TFs) controlling human mDAN differentiation during development have been described, but much of the regulatory landscape remains undefined. Using a tyrosine hydroxylase (TH) human iPSC reporter line, we here generate time series transcriptomic and epigenomic profiles of purified mDANs during differentiation. Integrative analysis predicts novel regulators of mDAN differentiation and super-enhancers are used to identify key TFs. We find LBX1, NHLH1 and NR2F1/2 to promote mDAN differentiation and show that overexpression of either LBX1 or NHLH1 can also improve mDAN specification. A more detailed investigation of TF targets reveals that NHLH1 promotes the induction of neuronal miR-124, LBX1 regulates cholesterol biosynthesis, and NR2F1/2 controls neuronal activity.
Collapse
Affiliation(s)
- Borja Gomez Ramos
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, Belvaux, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Jochen Ohnmacht
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, Belvaux, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Nikola de Lange
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Elena Valceschini
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Aurélien Ginolhac
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Marie Catillon
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Daniele Ferrante
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Aleksandar Rakovic
- Institute of Neurogenetics, University of Lübeck, 23538, Lübeck, Germany
| | - Rashi Halder
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - François Massart
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Giuseppe Arena
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Paul Antony
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Silvia Bolognin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, 23538, Lübeck, Germany
| | - Roland Krause
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Marcel H Schulz
- Institute for Cardiovascular Regeneration, Goethe University, 60590, Frankfurt, Germany
- German Centre for Cardiovascular Research, Partner site Rhein-Main, 60590, Frankfurt am Main, Germany
- Cardio-Pulmonary Institute, Goethe University, Frankfurt am Main, Germany
| | - Thomas Sauter
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Rejko Krüger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
- Centre Hospitalier de Luxembourg (CHL), L-1210, Luxembourg, Luxembourg
- Luxembourg Institute of Health (LIH), L-1445, Luxembourg, Luxembourg
| | - Lasse Sinkkonen
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, Belvaux, Luxembourg.
| |
Collapse
|
9
|
Ye P, Fang Q, Hu X, Zou W, Huang M, Ke M, Li Y, Liu M, Cai X, Zhang C, Hua N, Al-Sheikh U, Liu X, Yu P, Jiang P, Pan PY, Luo J, Jiang LH, Xu S, Fang EF, Su H, Kang L, Yang W. TRPM2 as a conserved gatekeeper determines the vulnerability of DA neurons by mediating ROS sensing and calcium dyshomeostasis. Prog Neurobiol 2023; 231:102530. [PMID: 37739206 DOI: 10.1016/j.pneurobio.2023.102530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/01/2023] [Accepted: 09/17/2023] [Indexed: 09/24/2023]
Abstract
Different dopaminergic (DA) neuronal subgroups exhibit distinct vulnerability to stress, while the underlying mechanisms are elusive. Here we report that the transient receptor potential melastatin 2 (TRPM2) channel is preferentially expressed in vulnerable DA neuronal subgroups, which correlates positively with aging in Parkinson's Disease (PD) patients. Overexpression of human TRPM2 in the DA neurons of C. elegans resulted in selective death of ADE but not CEP neurons in aged worms. Mechanistically, TRPM2 activation mediates FZO-1/CED-9-dependent mitochondrial hyperfusion and mitochondrial permeability transition (MPT), leading to ADE death. In mice, TRPM2 knockout reduced vulnerable substantia nigra pars compacta (SNc) DA neuronal death induced by stress. Moreover, the TRPM2-mediated vulnerable DA neuronal death pathway is conserved from C. elegans to toxin-treated mice model and PD patient iPSC-derived DA neurons. The vulnerable SNc DA neuronal loss is the major symptom and cause of PD, and therefore the TRPM2-mediated pathway serves as a promising therapeutic target against PD.
Collapse
Affiliation(s)
- Peiwu Ye
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qiuyuan Fang
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xupang Hu
- Second Clinical Medical College, Affiliated Secondary Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310011, China
| | - Wenjuan Zou
- Department of Neurobiology and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang 310053, China
| | - Miaodan Huang
- Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Minjing Ke
- Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yunhao Li
- Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Min Liu
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaobo Cai
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Congyi Zhang
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ning Hua
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Umar Al-Sheikh
- Department of Neurobiology and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang 310053, China
| | - Xingyu Liu
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Peilin Yu
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Peiran Jiang
- School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ping-Yue Pan
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane, Piscataway, NJ 08854, USA
| | - Jianhong Luo
- School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lin-Hua Jiang
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK; Sino-UK Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang 453000, China; University of Leeds, Leeds LS2 9JT, UK
| | - Suhong Xu
- Center for Stem Cell and Regenerative Medicine and Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Evandro F Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Huanxing Su
- Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| | - Lijun Kang
- Second Clinical Medical College, Affiliated Secondary Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310011, China; School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Wei Yang
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
10
|
Willmore L, Minerva AR, Engelhard B, Murugan M, McMannon B, Oak N, Thiberge SY, Peña CJ, Witten IB. Overlapping representations of food and social stimuli in mouse VTA dopamine neurons. Neuron 2023; 111:3541-3553.e8. [PMID: 37657441 DOI: 10.1016/j.neuron.2023.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 05/17/2023] [Accepted: 08/03/2023] [Indexed: 09/03/2023]
Abstract
Dopamine neurons of the ventral tegmental area (VTADA) respond to food and social stimuli and contribute to both forms of motivation. However, it is unclear whether the same or different VTADA neurons encode these different stimuli. To address this question, we performed two-photon calcium imaging in mice presented with food and conspecifics and found statistically significant overlap in the populations responsive to both stimuli. Both hunger and opposite-sex social experience further increased the proportion of neurons that respond to both stimuli, implying that increasing motivation for one stimulus increases overlap. In addition, single-nucleus RNA sequencing revealed significant co-expression of feeding- and social-hormone-related genes in individual VTADA neurons. Taken together, our functional and transcriptional data suggest overlapping VTADA populations underlie food and social motivation.
Collapse
Affiliation(s)
- Lindsay Willmore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Adelaide R Minerva
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Ben Engelhard
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA; Faculty of Medicine, Technion, Haifa 3525433, Israel.
| | - Malavika Murugan
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Brenna McMannon
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Nirja Oak
- Faculty of Medicine, Technion, Haifa 3525433, Israel
| | - Stephan Y Thiberge
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Catherine J Peña
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA.
| | - Ilana B Witten
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
11
|
Burkert N, Roy S, Häusler M, Wuttke D, Müller S, Wiemer J, Hollmann H, Oldrati M, Ramirez-Franco J, Benkert J, Fauler M, Duda J, Goaillard JM, Pötschke C, Münchmeyer M, Parlato R, Liss B. Deep learning-based image analysis identifies a DAT-negative subpopulation of dopaminergic neurons in the lateral Substantia nigra. Commun Biol 2023; 6:1146. [PMID: 37950046 PMCID: PMC10638391 DOI: 10.1038/s42003-023-05441-6] [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: 12/08/2022] [Accepted: 10/10/2023] [Indexed: 11/12/2023] Open
Abstract
Here we present a deep learning-based image analysis platform (DLAP), tailored to autonomously quantify cell numbers, and fluorescence signals within cellular compartments, derived from RNAscope or immunohistochemistry. We utilised DLAP to analyse subtypes of tyrosine hydroxylase (TH)-positive dopaminergic midbrain neurons in mouse and human brain-sections. These neurons modulate complex behaviour, and are differentially affected in Parkinson's and other diseases. DLAP allows the analysis of large cell numbers, and facilitates the identification of small cellular subpopulations. Using DLAP, we identified a small subpopulation of TH-positive neurons (~5%), mainly located in the very lateral Substantia nigra (SN), that was immunofluorescence-negative for the plasmalemmal dopamine transporter (DAT), with ~40% smaller cell bodies. These neurons were negative for aldehyde dehydrogenase 1A1, with a lower co-expression rate for dopamine-D2-autoreceptors, but a ~7-fold higher likelihood of calbindin-d28k co-expression (~70%). These results have important implications, as DAT is crucial for dopamine signalling, and is commonly used as a marker for dopaminergic SN neurons.
Collapse
Affiliation(s)
- Nicole Burkert
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Shoumik Roy
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany.
| | - Max Häusler
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | | | - Sonja Müller
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Johanna Wiemer
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Helene Hollmann
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Marvin Oldrati
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Jorge Ramirez-Franco
- UMR_S 1072, Aix Marseille Université, INSERM, Faculté de Médecine Secteur Nord, Marseille, France
- INT, Aix Marseille Université, CNRS, Campus Santé Timone, Marseille, France
| | - Julia Benkert
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Michael Fauler
- Institute of General Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Johanna Duda
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Jean-Marc Goaillard
- UMR_S 1072, Aix Marseille Université, INSERM, Faculté de Médecine Secteur Nord, Marseille, France
- INT, Aix Marseille Université, CNRS, Campus Santé Timone, Marseille, France
| | - Christina Pötschke
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
| | - Moritz Münchmeyer
- Wolution GmbH & Co. KG, 82152, Munich, Germany
- Department of Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Rosanna Parlato
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany
- Division of Neurodegenerative Disorders, Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, 68167, Mannheim, Germany
| | - Birgit Liss
- Institute of Applied Physiology, Medical Faculty, Ulm University, 89081, Ulm, Germany.
- Linacre College & New College, Oxford University, OX1 2JD, Oxford, UK.
| |
Collapse
|
12
|
Hernandez G, Kouwenhoven WM, Poirier E, Lebied K, Lévesque D, Rompré PP. Dorsal raphe stimulation relays a reward signal to the ventral tegmental area via GluN2C NMDA receptors. PLoS One 2023; 18:e0293564. [PMID: 37930965 PMCID: PMC10627466 DOI: 10.1371/journal.pone.0293564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/15/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Glutamate relays a reward signal from the dorsal raphe (DR) to the ventral tegmental area (VTA). However, the role of the different subtypes of N-methyl-D-aspartate (NMDA) receptors is complex and not clearly understood. Therefore, we measured NMDA receptors subunits expression in limbic brain areas. In addition, we studied the effects of VTA down-regulation of GluN2C NMDA receptor on the reward signal that arises from DR electrical stimulation. METHODS Using qPCR, we identified the relative composition of the different Grin2a-d subunits of the NMDA receptors in several brain areas. Then, we used fluorescent in situ hybridization (FISH) to evaluate the colocalization of Grin2c and tyrosine hydroxylase (TH) mRNA in VTA neurons. To assess the role of GluN2C in brain stimulation reward, we downregulated this receptor using small interfering RNA (siRNA) in rats self-stimulating for electrical pulses delivered to the DR. To delineate further the specific role of GluN2C in relaying the reward signal, we pharmacologically altered the function of VTA NMDA receptors by bilaterally microinjecting the NMDA receptor antagonist PPPA. RESULTS We identified GluN2C as the most abundant subunit of the NMDA receptor expressed in the VTA. FISH revealed that about 50% of TH-positive neurons colocalize with Grin2c transcript. siRNA manipulation produced a selective down-regulation of the GluN2C protein subunit and a significant reduction in brain stimulation reward. Interestingly, PPPA enhanced brain stimulation reward, but only in rats that received the nonactive RNA sequence. CONCLUSION The present results suggest that VTA glutamate neurotransmission relays a reward signal initiated by DR stimulation by acting on GluN2C NMDA receptors.
Collapse
Affiliation(s)
- Giovanni Hernandez
- Département de Neurosciences (Faculté de Médecine), Université de Montréal, Montréal, QC, Canada
| | - Willemieke M. Kouwenhoven
- Département de Pharmacologie et Physiologie (Faculté de Médecine), Université de Montréal, Montréal, QC, Canada
| | - Emmanuelle Poirier
- Département de Neurosciences (Faculté de Médecine), Université de Montréal, Montréal, QC, Canada
| | - Karim Lebied
- Département de Neurosciences (Faculté de Médecine), Université de Montréal, Montréal, QC, Canada
| | - Daniel Lévesque
- Département de Pharmacie (Faculté de Pharmacie), Université de Montréal, Montréal, QC, Canada
| | - Pierre-Paul Rompré
- Département de Neurosciences (Faculté de Médecine), Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
13
|
Caldwell M, Ayo-Jibunoh V, Mendoza JC, Brimblecombe KR, Reynolds LM, Zhu Jiang XY, Alarcon C, Fiore E, N Tomaio J, Phillips GR, Mingote S, Flores C, Casaccia P, Liu J, Cragg SJ, McCloskey DP, Yetnikoff L. Axo-glial interactions between midbrain dopamine neurons and oligodendrocyte lineage cells in the anterior corpus callosum. Brain Struct Funct 2023; 228:1993-2006. [PMID: 37668732 PMCID: PMC10516790 DOI: 10.1007/s00429-023-02695-y] [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: 06/14/2023] [Accepted: 08/09/2023] [Indexed: 09/06/2023]
Abstract
Oligodendrocyte progenitor cells (OPCs) receive synaptic innervation from glutamatergic and GABAergic axons and can be dynamically regulated by neural activity, resulting in activity-dependent changes in patterns of axon myelination. However, it remains unclear to what extent other types of neurons may innervate OPCs. Here, we provide evidence implicating midbrain dopamine neurons in the innervation of oligodendrocyte lineage cells in the anterior corpus callosum and nearby white matter tracts of male and female adult mice. Dopaminergic axon terminals were identified in the corpus callosum of DAT-Cre mice after injection of an eYFP reporter virus into the midbrain. Furthermore, fast-scan cyclic voltammetry revealed monoaminergic transients in the anterior corpus callosum, consistent with the anatomical findings. Using RNAscope, we further demonstrate that ~ 40% of Olig2 + /Pdfgra + cells and ~ 20% of Olig2 + /Pdgfra- cells in the anterior corpus callosum express Drd1 and Drd2 transcripts. These results suggest that oligodendrocyte lineage cells may respond to dopamine released from midbrain dopamine axons, which could affect myelination. Together, this work broadens our understanding of neuron-glia interactions with important implications for myelin plasticity by identifying midbrain dopamine axons as a potential regulator of corpus callosal oligodendrocyte lineage cells.
Collapse
Affiliation(s)
- Megan Caldwell
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Vanessa Ayo-Jibunoh
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Josue Criollo Mendoza
- Department of Biology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Katherine R Brimblecombe
- Centre for Integrative Neuroscience, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Lauren M Reynolds
- Plasticité du Cerveau, CNRS UMR8249, École Supérieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Paris, France
| | - Xin Yan Zhu Jiang
- Department of Biology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Colin Alarcon
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Elizabeth Fiore
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Jacquelyn N Tomaio
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, USA
| | - Greg R Phillips
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Department of Biology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
- Center for Developmental Neuroscience, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Susana Mingote
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, USA
| | - Cecilia Flores
- Department of Psychiatry and of Neurology and Neuroscience, McGill University, and Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Patrizia Casaccia
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, USA
- Department of Neuroscience and Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jia Liu
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, USA
| | - Stephanie J Cragg
- Centre for Integrative Neuroscience, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3PT, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Dan P McCloskey
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA
| | - Leora Yetnikoff
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, 365 5Th Ave, New York, NY, 10016, USA.
- Department of Psychology, College of Staten Island, City University of New York, 2800 Victory Boulevard, Staten Island, NY, 10314, USA.
| |
Collapse
|
14
|
Cobb-Lewis DE, Sansalone L, Khaliq ZM. Contributions of the Sodium Leak Channel NALCN to Pacemaking of Medial Ventral Tegmental Area and Substantia Nigra Dopaminergic Neurons. J Neurosci 2023; 43:6841-6853. [PMID: 37640554 PMCID: PMC10573758 DOI: 10.1523/jneurosci.0930-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/31/2023] Open
Abstract
We tested the role of the sodium leak channel, NALCN, in pacemaking of dopaminergic neuron (DAN) subpopulations from adult male and female mice. In situ hybridization revealed NALCN RNA in all DANs, with lower abundance in medial ventral tegmental area (VTA) relative to substantia nigra pars compacta (SNc). Despite lower relative abundance of NALCN, we found that acute pharmacological blockade of NALCN in medial VTA DANs slowed pacemaking by 49.08%. We also examined the electrophysiological properties of projection-defined VTA DAN subpopulations identified by retrograde labeling. Inhibition of NALCN reduced pacemaking in DANs projecting to medial nucleus accumbens (NAc) and others projecting to lateral NAc by 70.74% and 31.98%, respectively, suggesting that NALCN is a primary driver of pacemaking in VTA DANs. In SNc DANs, potentiating NALCN by lowering extracellular calcium concentration speeded pacemaking in wildtype but not NALCN conditional knockout mice, demonstrating functional presence of NALCN. In contrast to VTA DANs, however, pacemaking in SNc DANs was unaffected by inhibition of NALCN. Instead, we found that inhibition of NALCN increased the gain of frequency-current plots at firing frequencies slower than spontaneous firing. Similarly, inhibition of the hyperpolarization-activated cyclic nucleotide-gated (HCN) conductance increased gain but had little effect on pacemaking. Interestingly, simultaneous inhibition of NALCN and HCN resulted in significant reduction in pacemaker rate. Thus, we found NALCN makes substantial contributions to driving pacemaking in VTA DAN subpopulations. In SNc DANs, NALCN is not critical for pacemaking but inhibition of NALCN makes cells more sensitive to hyperpolarizing stimuli.SIGNIFICANCE STATEMENT Pacemaking in midbrain dopaminergic neurons (DAN) relies on multiple subthreshold conductances, including a sodium leak. Whether the sodium leak channel, NALCN, contributes to pacemaking in DANs located in the VTA and the SNc has not yet been determined. Using electrophysiology and pharmacology, we show that NALCN plays a prominent role in driving pacemaking in projection-defined VTA DAN subpopulations. By contrast, pacemaking in SNc neurons does not rely on NALCN. Instead, the presence of NALCN regulates the excitability of SNc DANs by reducing the gain of the neuron's response to inhibitory stimuli. Together, these findings will inform future efforts to obtain DAN subpopulation-specific treatments for use in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Dana E Cobb-Lewis
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
- Institute for Neuroscience, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| | - Lorenzo Sansalone
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
| | - Zayd M Khaliq
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
15
|
Jin R, Sun S, Hu Y, Zhang H, Sun X. Neuropeptides Modulate Feeding via the Dopamine Reward Pathway. Neurochem Res 2023:10.1007/s11064-023-03954-4. [PMID: 37233918 DOI: 10.1007/s11064-023-03954-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Dopamine (DA) is a catecholamine neurotransmitter widely distributed in the central nervous system. It participates in various physiological functions, such as feeding, anxiety, fear, sleeping and arousal. The regulation of feeding is exceptionally complex, involving energy homeostasis and reward motivation. The reward system comprises the ventral tegmental area (VTA), nucleus accumbens (NAc), hypothalamus, and limbic system. This paper illustrates the detailed mechanisms of eight typical orexigenic and anorexic neuropeptides that regulate food intake through the reward system. According to recent literature, neuropeptides released from the hypothalamus and other brain regions regulate reward feeding predominantly through dopaminergic neurons projecting from the VTA to the NAc. In addition, their effect on the dopaminergic system is mediated by the prefrontal cortex, paraventricular thalamus, laterodorsal tegmental area, amygdala, and complex neural circuits. Research on neuropeptides involved in reward feeding can help identify more targets to treat diseases with metabolic disorders, such as obesity.
Collapse
Affiliation(s)
- Ruijie Jin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Shanbin Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Yang Hu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Hongfei Zhang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xiangrong Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
16
|
Willmore L, Minerva AR, Engelhard B, Murugan M, McMannon B, Oak N, Thiberge SY, Peña CJ, Witten IB. Overlapping representations of food and social stimuli in VTA dopamine neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541104. [PMID: 37293057 PMCID: PMC10245666 DOI: 10.1101/2023.05.17.541104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Dopamine neurons of the ventral tegmental area (VTA DA ) respond to food and social stimuli and contribute to both forms of motivation. However, it is unclear if the same or different VTA DA neurons encode these different stimuli. To address this question, we performed 2-photon calcium imaging in mice presented with food and conspecifics, and found statistically significant overlap in the populations responsive to both stimuli. Both hunger and opposite-sex social experience further increased the proportion of neurons that respond to both stimuli, implying that modifying motivation for one stimulus affects responses to both stimuli. In addition, single-nucleus RNA sequencing revealed significant co-expression of feeding- and social-hormone related genes in individual VTA DA neurons. Taken together, our functional and transcriptional data suggest overlapping VTA DA populations underlie food and social motivation.
Collapse
Affiliation(s)
- Lindsay Willmore
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Adelaide R. Minerva
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Ben Engelhard
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
- Department of Medicine, Technion, Haifa, 3525433, Israel
| | - Malavika Murugan
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Brenna McMannon
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Nirja Oak
- Department of Medicine, Technion, Haifa, 3525433, Israel
| | - Stephan Y. Thiberge
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Catherine J. Peña
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Ilana B. Witten
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| |
Collapse
|
17
|
Kamath T, Macosko EZ. Insights into Neurodegeneration in Parkinson's Disease from Single-Cell and Spatial Genomics. Mov Disord 2023; 38:518-525. [PMID: 36881930 PMCID: PMC11056908 DOI: 10.1002/mds.29374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/12/2023] [Accepted: 02/16/2023] [Indexed: 03/09/2023] Open
Abstract
Parkinson's disease (PD) is pathologically defined by the death of dopaminergic (DA) neurons within the pars compacta of the substantia nigra. To date, the cause of this multifaceted disease remains largely unclear, which may contribute in part to a current lack of disease-modifying therapies. Recent advances in single-cell and spatial genomic profiling tools have provided powerful new ways to measure cellular state changes in brain diseases. Here, we describe how these tools have offered insight into these complex disorders and highlight a recently performed comprehensive study of DA neuron susceptibility in PD. The data generated by this recent work provide evidence for the role of specific pathways and common genetic variants resulting in the loss of a critical DA subtype in PD. We conclude by outlining a set of basic and translational opportunities that arise from those data and insights gathered from this work. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Tushar Kamath
- Stanley Center for Psychiatric Research, Broad Institute, 75 Ames Street Cambridge, MA 02139
- Harvard Medical School and Harvard/MIT MD-PhD Program, Harvard University, Cambridge, MA 02139 USA
| | - Evan Z. Macosko
- Stanley Center for Psychiatric Research, Broad Institute, 75 Ames Street Cambridge, MA 02139
- Massachusetts General Hospital, Department of Psychiatry, Boston, MA USA
| |
Collapse
|
18
|
Birtele M, Storm P, Sharma Y, Kajtez J, Wahlestedt JN, Sozzi E, Nilsson F, Stott S, He XL, Mattsson B, Ottosson DR, Barker RA, Fiorenzano A, Parmar M. Single-cell transcriptional and functional analysis of dopaminergic neurons in organoid-like cultures derived from human fetal midbrain. Development 2022; 149:285890. [PMID: 36305490 PMCID: PMC10114107 DOI: 10.1242/dev.200504] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 09/12/2022] [Indexed: 11/06/2022]
Abstract
Significant efforts are ongoing to develop refined differentiation protocols to generate midbrain dopamine (DA) neurons from pluripotent stem cells for application in disease modeling, diagnostics, drug screening and cell-based therapies for Parkinson's disease. An increased understanding of the timing and molecular mechanisms that promote the generation of distinct subtypes of human midbrain DA during development will be essential for guiding future efforts to generate molecularly defined and subtype-specific DA neurons from pluripotent stem cells. Here, we use droplet-based single-cell RNA sequencing to transcriptionally profile the developing human ventral midbrain (VM) when the DA neurons are generated (6-11 weeks post-conception) and their subsequent differentiation into functional mature DA neurons in primary fetal 3D organoid-like cultures. This approach reveals that 3D cultures are superior to monolayer conditions for their ability to generate and maintain mature DA neurons; hence, they have the potential to be used for studying human VM development. These results provide a unique transcriptional profile of the developing human fetal VM and functionally mature human DA neurons that can be used to guide stem cell-based therapies and disease modeling approaches in Parkinson's disease.
Collapse
Affiliation(s)
- Marcella Birtele
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Petter Storm
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Yogita Sharma
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Janko Kajtez
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Jenny Nelander Wahlestedt
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Edoardo Sozzi
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Fredrik Nilsson
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Simon Stott
- Department of Clinical Neuroscience and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0PY, UK
| | - Xiaoling L He
- Department of Clinical Neuroscience and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0PY, UK
| | - Bengt Mattsson
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Daniella Rylander Ottosson
- Regenerative Neurophysiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Roger A Barker
- Department of Clinical Neuroscience and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0PY, UK
| | - Alessandro Fiorenzano
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| |
Collapse
|
19
|
Fujita T, Aoki N, Mori C, Serizawa S, Kihara-Negishi F, Homma KJ, Yamaguchi S. Dopaminergic nuclei in the chick midbrain express serotonin receptor subfamily genes. Front Physiol 2022; 13:1030621. [PMID: 36425295 PMCID: PMC9679639 DOI: 10.3389/fphys.2022.1030621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/28/2022] [Indexed: 08/05/2023] Open
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is a phylogenetically conserved modulator of numerous aspects of neural functions. Serotonergic neurons in the dorsal and median raphe nucleus provide ascending innervation to the entire forebrain and midbrain. Another important neural modulatory system exists in the midbrain, the dopaminergic system, which is associated to reward processing and motivation control. Dopaminergic neurons are distributed and clustered in the brain, classically designated as groups A8-A16. Among them, groups A8-A10 associated with reward processing and motivation control are located in the midbrain and projected to the forebrain. Recently, midbrain dopaminergic neurons were shown to be innervated by serotonergic neurons and modulated by 5-HT, with the crosstalk between serotonergic and dopaminergic systems attracting increased attention. In birds, previous studies revealed that midbrain dopaminergic neurons are located in the A8-A10 homologous clusters. However, the detailed distribution of dopaminergic neurons and the crosstalk between serotonergic and dopaminergic systems in the bird are poorly understood. To improve the understanding of the regulation of the dopaminergic by the serotonergic system, we performed in situ hybridization in the chick brainstem. We prepared RNA probes for chick orthologues of dopaminergic neuron-related genes; tyrosine hydroxylase (TH) and dopa decarboxylase (DDC), noradrenaline related genes; noradrenaline transporter (NAT) and dopamine beta-hydroxylase (DBH), and serotonin receptor genes; 5-HTR1A, 5-HTR1B, 5-HTR1D, 5-HTR1E, 5-HTR1F, 5-HTR2A, 5-HTR2B, 5-HTR2C, 5-HTR3A, 5-HTR4, 5-HTR5A, and 5-HTR7. We confirmed that the expression of tyrosine hydroxylase (TH) and NAT was well matched in all chick dopaminergic nuclei examined. This supported that the compensation of the function of dopamine transporter (DAT) by NAT is a general property of avian dopaminergic neurons. Furthermore, we showed that 5-HTR1A and 5-HTR1B were expressed in midbrain dopaminergic nuclei, suggesting the serotonergic regulation of the dopaminergic system via these receptors in chicks. Our findings will help us understand the interactions between the dopaminergic and serotonergic systems in birds at the molecular level.
Collapse
Affiliation(s)
- Toshiyuki Fujita
- Department of Biological Sciences, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo, Japan
| | - Naoya Aoki
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo, Japan
| | - Chihiro Mori
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo, Japan
| | - Shouta Serizawa
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo, Japan
| | - Fumiko Kihara-Negishi
- Department of Biological Sciences, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo, Japan
| | - Koichi J. Homma
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo, Japan
| | - Shinji Yamaguchi
- Department of Biological Sciences, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo, Japan
| |
Collapse
|
20
|
Kanarik M, Grimm O, Mota NR, Reif A, Harro J. ADHD co-morbidities: A review of implication of gene × environment effects with dopamine-related genes. Neurosci Biobehav Rev 2022; 139:104757. [PMID: 35777579 DOI: 10.1016/j.neubiorev.2022.104757] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 02/07/2023]
Abstract
ADHD is a major burden in adulthood, where co-morbid conditions such as depression, substance use disorder and obesity often dominate the clinical picture. ADHD has substantial shared heritability with other mental disorders, contributing to comorbidity. However, environmental risk factors exist but their interaction with genetic makeup, especially in relation to comorbid disorders, remains elusive. This review for the first time summarizes present knowledge on gene x environment (GxE) interactions regarding the dopamine system. Hitherto, mainly candidate (GxE) studies were performed, focusing on the genes DRD4, DAT1 and MAOA. Some evidence suggest that the variable number tandem repeats in DRD4 and MAOA may mediate GxE interactions in ADHD generally, and comorbid conditions specifically. Nevertheless, even for these genes, common variants are bound to suggest risk only in the context of gender and specific environments. For other polymorphisms, evidence is contradictory and less convincing. Particularly lacking are longitudinal studies testing the interaction of well-defined environmental with polygenic risk scores reflecting the dopamine system in its entirety.
Collapse
Affiliation(s)
- Margus Kanarik
- Chair of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Ravila 14A Chemicum, 50411 Tartu, Estonia
| | - Oliver Grimm
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Nina Roth Mota
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Jaanus Harro
- Chair of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Ravila 14A Chemicum, 50411 Tartu, Estonia; Psychiatry Clinic, North Estonia Medical Centre, Paldiski Road 52, 10614 Tallinn, Estonia.
| |
Collapse
|
21
|
Mitra S, Basu S, Singh O, Srivastava A, Singru PS. Calcium-binding proteins typify the dopaminergic neuronal subtypes in the ventral tegmental area of zebra finch, Taeniopygia guttata. J Comp Neurol 2022; 530:2562-2586. [PMID: 35715989 DOI: 10.1002/cne.25352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 11/11/2022]
Abstract
Calcium-binding proteins (CBPs) regulate neuronal function in midbrain dopamine (DA)-ergic neurons in mammals by buffering and sensing the intracellular Ca2+ , and vesicular release. In birds, the equivalent set of neurons are important in song learning, directed singing, courtship, and energy balance, yet the status of CBPs in these neurons is unknown. Herein, for the first time, we probe the nature of CBPs, namely, Calbindin-, Calretinin-, Parvalbumin-, and Secretagogin-expressing DA neurons in the ventral tegmental area (VTA) and substantia nigra (SN) in the midbrain of zebra finch, Taeniopygia guttata. qRT-PCR analysis of ventral midbrain tissue fragment revealed higher Calbindin- and Calretinin-mRNA levels compared to Parvalbumin and Secretagogin. Application of immunofluorescence showed CBP-immunoreactive (-i) neurons in VTA (anterior [VTAa], mid [VTAm], caudal [VTAc]), SN (compacta [SNc], and reticulata [SNr]). Compared to VTAa, higher Calbindin- and Parvalbumin-immunoreactivity (-ir), and lower Calretinin-ir were observed in VTAm and VTAc. Secretagogin-ir was highly localized to VTAa. In SN, Calbindin- and Calretinin-ir were higher in SNc, SNr was Parvalbumin enriched, and Secretagogin-ir was not detected. Weak, moderate, and intense tyrosine hydroxylase (TH)-i VTA neurons were demarcated as subtypes 1, 2, and 3, respectively. While subtype 1 TH-i neurons were neither Calbindin- nor Calretinin-i, ∼80 and ∼65% subtype 2 and ∼30 and ∼45% subtype 3 TH-i neurons co-expressed Calbindin and Calretinin, respectively. All TH-i neuronal subtypes co-expressed Parvalbumin with reciprocal relationship with TH-ir. We suggest that the CBPs may determine VTA DA neuronal heterogeneity and differentially regulate their activity in T. guttata.
Collapse
Affiliation(s)
- Saptarsi Mitra
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sumela Basu
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Omprakash Singh
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Abhinav Srivastava
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Praful S Singru
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
22
|
Chen M, Liu F, Wen L, Hu X. Nonlinear relationship between CAN current and C a 2 + influx underpins synergistic action of muscarinic and NMDA receptors on bursts induction in midbrain dopaminergic neurons. Cogn Neurodyn 2022; 16:719-731. [PMID: 35603052 PMCID: PMC9120320 DOI: 10.1007/s11571-021-09740-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 10/08/2021] [Accepted: 10/19/2021] [Indexed: 12/29/2022] Open
Abstract
Bursting of midbrain dopamine (DA) neurons is believed to represent an important reward signal that instructs and reinforces goal-directed behaviors. In DA neurons, many afferents, including cholinergic and glutamatergic inputs, induce bursting, and it is suggested that a synergy exists between these afferents in bursting induction. However, the underlying mechanisms of the role and the synergy of muscarinic receptors (mAChRs) and NMDA receptors (NMDARs) in bursting induction remain unclear. Present work bestowed analysis using a mathematical model of DA neurons to demonstrate the underlying mechanisms. Activation of mAChRs, leading to rapid translocation of TRPC channels to cell surface, recruited C a 2 + -activated nonspecific (CAN) current ( I CAN ), meanwhile NMDARs excitation triggered C a 2 + influx, which induced the positive feedback loop of C a 2 + and I CAN , respectively, yielded a robust ramping depolarization with a superimposed high-frequency spiking. In some DA cells, neither NMDARs nor mAChRs induced positive feedback loop unless they were activated simultaneously to induce bursting. Our experimental results verified those theoretical findings. These together unveil the underlying mechanisms of the role and synergy of mAChRs and NMDARs in bursting induction emerge from the nonlinear relationship between C a 2 + influx and I CAN . Given the diverse and complex nature of neural circuitry and the DA neuron heterogeneity, our work provides new insights to understand specific afferents, the synergy between those afferents, and the differences in intrinsic excitability to be integrated by the bursting to accurately characterize the dopamine signals in the valances of reward and reinforcement, and a broad spectrum of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Mengjiao Chen
- College of Life Sciences, Leshan Normal University, Leshan, 614000 China
- Key Laboratory of Sichuan Institute for Protecting Endangered Birds in the Southwest Mountains, Leshan Normal University, Leshan, 614000 China
- Key Laboratory of MOE for Modern Teaching Technology, Shaanxi Normal University, Xi’an, 710062 China
| | - Fangqing Liu
- College of Life Sciences, Leshan Normal University, Leshan, 614000 China
- Key Laboratory of Sichuan Institute for Protecting Endangered Birds in the Southwest Mountains, Leshan Normal University, Leshan, 614000 China
| | - Longying Wen
- College of Life Sciences, Leshan Normal University, Leshan, 614000 China
- Key Laboratory of Sichuan Institute for Protecting Endangered Birds in the Southwest Mountains, Leshan Normal University, Leshan, 614000 China
| | - Xia Hu
- College of Life Sciences, Leshan Normal University, Leshan, 614000 China
| |
Collapse
|
23
|
Tang F, Liu H, Zhang XJ, Zheng HH, Dai YM, Zheng LY, Yang WH, Du YY, Liu J. Evidence for Dopamine Abnormalities Following Acute Methamphetamine Exposure Assessed by Neuromelanin-Sensitive Magnetic Resonance Imaging. Front Aging Neurosci 2022; 14:865825. [PMID: 35707702 PMCID: PMC9190254 DOI: 10.3389/fnagi.2022.865825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/20/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundNeuromelanin-sensitive magnetic resonance imaging (NM-MRI) is a newly developed MRI technique that provides a non-invasive way to indirectly measure of dopamine (DA) function. This study aimed to determine NM concentrations in brain regions following acute methamphetamine (MA) administration using NM-MRI and to explore whether NM-MRI can be used as a biomarker of DA function in non-neurodegenerative diseases.MethodsBaseline NM-MRI, T1-weighted and T2-weighted images were acquired from 27 rats before drug/placebo injection. The control group (n = 11) received acute placebo (Normal saline), while the experimental group (n = 16) received acute MA. NM-MRI scans were performed 5, 30, 60 and 90 min after injection. Regions of interest (ROIs), including the caudate putamen (CP), nucleus accumbens (NAc), hippocampus (HIP), substantia nigra (SN) and crus cerebri (CC), were manually drawn by an experienced radiologist. NM-MRI signal intensity in five brain regions at different time points (baseline and 5, 30, 60, and 90 min) were analyzed.ResultsIn both the control and experimental groups, at each time point (baseline and 5, 30, 60, and 90 min), the SN exhibited significantly higher NM-MRI signal intensity than the other brain regions (P < 0.05). In addition, acute MA administration resulted in a continuous upward trend in NM-MRI signal intensity in each brain region over time. However, there was no such trend over time in the control group. The NM-MRI signal intensity of SN in the experimental group was significantly higher at the 60 and 90 min compared with that in the control group (P values were 0.042 and 0.042 respectively). Within experimental group, the NM-MRI signal intensity of SN was significantly higher at the 60 and 90 min compared with that before MA administration (P values were 0.023 and 0.011 respectively). Increased amplitudes and rates of NM-MRI signal intensity were higher in the SN than in other brain regions after MA administration.ConclusionOur results indicated that NM was mainly deposited in the SN, and the conversion of DA to NM was most significant in the SN after acute MA exposure. Increased DA release induced by acute MA exposure may lead to increased accumulation of NM in multiple brain regions that can be revealed by NM-MRI. NM-MRI may serve as a powerful imaging tool that could have diverse research and clinical applications for detecting pathological changes in drug addiction and related non-neurodegenerative diseases.
Collapse
Affiliation(s)
- Fei Tang
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Hui Liu
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiao Jie Zhang
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Hui Hui Zheng
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Yong Ming Dai
- MR Collaboration, Central Research Institute, United Imaging Healthcare, Shanghai, China
| | - Li Yun Zheng
- MR Collaboration, Central Research Institute, United Imaging Healthcare, Shanghai, China
| | - Wen Han Yang
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Yan Yao Du
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Jun Liu
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Jun Liu,
| |
Collapse
|
24
|
Novak G, Kyriakis D, Grzyb K, Bernini M, Rodius S, Dittmar G, Finkbeiner S, Skupin A. Single-cell transcriptomics of human iPSC differentiation dynamics reveal a core molecular network of Parkinson's disease. Commun Biol 2022; 5:49. [PMID: 35027645 PMCID: PMC8758783 DOI: 10.1038/s42003-021-02973-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/14/2021] [Indexed: 01/02/2023] Open
Abstract
Parkinson's disease (PD) is the second-most prevalent neurodegenerative disorder, characterized by the loss of dopaminergic neurons (mDA) in the midbrain. The underlying mechanisms are only partly understood and there is no treatment to reverse PD progression. Here, we investigated the disease mechanism using mDA neurons differentiated from human induced pluripotent stem cells (hiPSCs) carrying the ILE368ASN mutation within the PINK1 gene, which is strongly associated with PD. Single-cell RNA sequencing (RNAseq) and gene expression analysis of a PINK1-ILE368ASN and a control cell line identified genes differentially expressed during mDA neuron differentiation. Network analysis revealed that these genes form a core network, members of which interact with all known 19 protein-coding Parkinson's disease-associated genes. This core network encompasses key PD-associated pathways, including ubiquitination, mitochondrial function, protein processing, RNA metabolism, and vesicular transport. Proteomics analysis showed a consistent alteration in proteins of dopamine metabolism, indicating a defect of dopaminergic metabolism in PINK1-ILE368ASN neurons. Our findings suggest the existence of a network onto which pathways associated with PD pathology converge, and offers an inclusive interpretation of the phenotypic heterogeneity of PD.
Collapse
Affiliation(s)
- Gabriela Novak
- The Integrative Cell Signalling Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.
- Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.
- Center for Systems and Therapeutics, the Gladstone Institutes and Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, 94158, USA.
| | - Dimitrios Kyriakis
- The Integrative Cell Signalling Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Kamil Grzyb
- The Integrative Cell Signalling Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Michela Bernini
- The Integrative Cell Signalling Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sophie Rodius
- Department of Infection and Immunity, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Gunnar Dittmar
- Department of Infection and Immunity, Luxembourg Institute of Health, Strassen, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Steven Finkbeiner
- Center for Systems and Therapeutics, the Gladstone Institutes and Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Alexander Skupin
- The Integrative Cell Signalling Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.
- University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
25
|
Tu H, Zhang ZW, Qiu L, Lin Y, Jiang M, Chia SY, Wei Y, Ng ASL, Reynolds R, Tan EK, Zeng L. Increased expression of pathological markers in Parkinson's disease dementia post-mortem brains compared to dementia with Lewy bodies. BMC Neurosci 2022; 23:3. [PMID: 34983390 PMCID: PMC8725407 DOI: 10.1186/s12868-021-00687-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 12/22/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are common age-related neurodegenerative diseases comprising Lewy body spectrum disorders associated with cortical and subcortical Lewy body pathology. Over 30% of PD patients develop PD dementia (PDD), which describes dementia arising in the context of established idiopathic PD. Furthermore, Lewy bodies frequently accompany the amyloid plaque and neurofibrillary tangle pathology of Alzheimer's disease (AD), where they are observed in the amygdala of approximately 60% of sporadic and familial AD. While PDD and DLB share similar pathological substrates, they differ in the temporal onset of motor and cognitive symptoms; however, protein markers to distinguish them are still lacking. METHODS Here, we systematically studied a series of AD and PD pathogenesis markers, as well as mitochondria, mitophagy, and neuroinflammation-related indicators, in the substantia nigra (SN), temporal cortex (TC), and caudate and putamen (CP) regions of human post-mortem brain samples from individuals with PDD and DLB and condition-matched controls. RESULTS We found that p-APPT668 (TC), α-synuclein (CP), and LC3II (CP) are all increased while the tyrosine hydroxylase (TH) (CP) is decreased in both PDD and DLB compared to control. Also, the levels of Aβ42 and DD2R, IBA1, and p-LRRK2S935 are all elevated in PDD compared to control. Interestingly, protein levels of p-TauS199/202 in CP and DD2R, DRP1, and VPS35 in TC are all increased in PDD compared to DLB. CONCLUSIONS Together, our comprehensive and systematic study identified a set of signature proteins that will help to understand the pathology and etiology of PDD and DLB at the molecular level.
Collapse
Affiliation(s)
- Haitao Tu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Zhi Wei Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Lifeng Qiu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Yuning Lin
- Guangxi University of Chinese Medicine, 179 Mingxiu Dong Rd., Nanning, 530001, Guangxi, China
| | - Mei Jiang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-Sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
- Department of Human Anatomy, Institute of Stem Cell and Regenerative Medicine, Dongguan Campus, Guangdong Medical University, Dongguan, China
| | - Sook-Yoong Chia
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Yanfei Wei
- Guangxi University of Chinese Medicine, 179 Mingxiu Dong Rd., Nanning, 530001, Guangxi, China
| | - Adeline S L Ng
- Department of Neurology, National Neuroscience Institute, Singapore, 308433, Singapore
- DUKE-NUS Graduate Medical School, Neuroscience & Behavioral Disorders Program, Singapore, 169857, Singapore
| | - Richard Reynolds
- Division of Neuroscience, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Novena Campus, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore, 308433, Singapore
- DUKE-NUS Graduate Medical School, Neuroscience & Behavioral Disorders Program, Singapore, 169857, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore.
- DUKE-NUS Graduate Medical School, Neuroscience & Behavioral Disorders Program, Singapore, 169857, Singapore.
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Novena Campus, 11 Mandalay Road, Singapore, 308232, Singapore.
| |
Collapse
|
26
|
Oosterveen T, Garção P, Moles-Garcia E, Soleilhavoup C, Travaglio M, Sheraz S, Peltrini R, Patrick K, Labas V, Combes-Soia L, Marklund U, Hohenstein P, Panman L. Pluripotent stem cell derived dopaminergic subpopulations model the selective neuron degeneration in Parkinson's disease. Stem Cell Reports 2021; 16:2718-2735. [PMID: 34678205 PMCID: PMC8581055 DOI: 10.1016/j.stemcr.2021.09.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 01/06/2023] Open
Abstract
In Parkinson’s disease (PD), substantia nigra (SN) dopaminergic (DA) neurons degenerate, while related ventral tegmental area (VTA) DA neurons remain relatively unaffected. Here, we present a methodology that directs the differentiation of mouse and human pluripotent stem cells toward either SN- or VTA-like DA lineage and models their distinct vulnerabilities. We show that the level of WNT activity is critical for the induction of the SN- and VTA-lineage transcription factors Sox6 and Otx2, respectively. Both WNT signaling modulation and forced expression of these transcription factors can drive DA neurons toward the SN- or VTA-like fate. Importantly, the SN-like lineage enriched DA cultures recapitulate the selective sensitivity to mitochondrial toxins as observed in PD, while VTA-like neuron-enriched cultures are more resistant. Furthermore, a proteomics approach led to the identification of compounds that alter SN neuronal survival, demonstrating the utility of our strategy for disease modeling and drug discovery. Derivation of distinct dopaminergic subpopulations from pluripotent stem cells Wnt signaling inhibitors promote SN dopaminergic neuron specification Modeling selective vulnerability of SN dopaminergic neurons in vitro Proteomics reveals pathways that promote SN dopaminergic neuron survival
Collapse
Affiliation(s)
- Tony Oosterveen
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Pedro Garção
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Emma Moles-Garcia
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Clement Soleilhavoup
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Marco Travaglio
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Shahida Sheraz
- Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Rosa Peltrini
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Kieran Patrick
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Valerie Labas
- PRC, INRA, CNRS, University of Tours, IFCE, Nouzilly, France
| | | | - Ulrika Marklund
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | | | - Lia Panman
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK.
| |
Collapse
|
27
|
Carmichael K, Sullivan B, Lopez E, Sun L, Cai H. Diverse midbrain dopaminergic neuron subtypes and implications for complex clinical symptoms of Parkinson's disease. AGEING AND NEURODEGENERATIVE DISEASES 2021; 1. [PMID: 34532720 PMCID: PMC8442626 DOI: 10.20517/and.2021.07] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Parkinson’s disease (PD), the most common degenerative movement disorder, is clinically manifested with various motor and non-motor symptoms. Degeneration of midbrain substantia nigra pas compacta (SNc) dopaminergic neurons (DANs) is generally attributed to the motor syndrome. The underlying neuronal mechanisms of non-motor syndrome are largely unexplored. Besides SNc, midbrain ventral tegmental area (VTA) DANs also produce and release dopamine and modulate movement, reward, motivation, and memory. Degeneration of VTA DANs also occurs in postmortem brains of PD patients, implying an involvement of VTA DANs in PD-associated non-motor symptoms. However, it remains to be established that there is a distinct segregation of different SNc and VTA DAN subtypes in regulating different motor and non-motor functions, and that different DAN subpopulations are differentially affected by normal ageing or PD. Traditionally, the distinction among different DAN subtypes was mainly based on the location of cell bodies and axon terminals. With the recent advance of single cell RNA sequencing technology, DANs can be readily classified based on unique gene expression profiles. A combination of specific anatomic and molecular markers shows great promise to facilitate the identification of DAN subpopulations corresponding to different behavior modules under normal and disease conditions. In this review, we first summarize the recent progress in characterizing genetically, anatomically, and functionally diverse midbrain DAN subtypes. Then, we provide perspectives on how the preclinical research on the connectivity and functionality of DAN subpopulations improves our current understanding of cell-type and circuit specific mechanisms of the disease, which could be critically informative for designing new mechanistic treatments.
Collapse
Affiliation(s)
- Kathleen Carmichael
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.,The Graduate Partnership Program of NIH and Brown University, National Institutes of Health, Bethesda, MD 20892, USA
| | - Breanna Sullivan
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elena Lopez
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lixin Sun
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
28
|
Fiorenzano A, Sozzi E, Parmar M, Storm P. Dopamine Neuron Diversity: Recent Advances and Current Challenges in Human Stem Cell Models and Single Cell Sequencing. Cells 2021; 10:cells10061366. [PMID: 34206038 PMCID: PMC8226961 DOI: 10.3390/cells10061366] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 12/12/2022] Open
Abstract
Human midbrain dopamine (DA) neurons are a heterogeneous group of cells that share a common neurotransmitter phenotype and are in close anatomical proximity but display different functions, sensitivity to degeneration, and axonal innervation targets. The A9 DA neuron subtype controls motor function and is primarily degenerated in Parkinson’s disease (PD), whereas A10 neurons are largely unaffected by the condition, and their dysfunction is associated with neuropsychiatric disorders. Currently, DA neurons can only be reliably classified on the basis of topographical features, including anatomical location in the midbrain and projection targets in the forebrain. No systematic molecular classification at the genome-wide level has been proposed to date. Although many years of scientific efforts in embryonic and adult mouse brain have positioned us to better understand the complexity of DA neuron biology, many biological phenomena specific to humans are not amenable to being reproduced in animal models. The establishment of human cell-based systems combined with advanced computational single-cell transcriptomics holds great promise for decoding the mechanisms underlying maturation and diversification of human DA neurons, and linking their molecular heterogeneity to functions in the midbrain. Human pluripotent stem cells have emerged as a useful tool to recapitulate key molecular features of mature DA neuron subtypes. Here, we review some of the most recent advances and discuss the current challenges in using stem cells, to model human DA biology. We also describe how single cell RNA sequencing may provide key insights into the molecular programs driving DA progenitor specification into mature DA neuron subtypes. Exploiting the state-of-the-art approaches will lead to a better understanding of stem cell-derived DA neurons and their use in disease modeling and regenerative medicine.
Collapse
|
29
|
Current State-of-the-Art and Unresolved Problems in Using Human Induced Pluripotent Stem Cell-Derived Dopamine Neurons for Parkinson's Disease Drug Development. Int J Mol Sci 2021; 22:ijms22073381. [PMID: 33806103 PMCID: PMC8037675 DOI: 10.3390/ijms22073381] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/15/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Human induced pluripotent stem (iPS) cells have the potential to give rise to a new era in Parkinson's disease (PD) research. As a unique source of midbrain dopaminergic (DA) neurons, iPS cells provide unparalleled capabilities for investigating the pathogenesis of PD, the development of novel anti-parkinsonian drugs, and personalized therapy design. Significant progress in developmental biology of midbrain DA neurons laid the foundation for their efficient derivation from iPS cells. The introduction of 3D culture methods to mimic the brain microenvironment further expanded the vast opportunities of iPS cell-based research of the neurodegenerative diseases. However, while the benefits for basic and applied studies provided by iPS cells receive widespread coverage in the current literature, the drawbacks of this model in its current state, and in particular, the aspects of differentiation protocols requiring further refinement are commonly overlooked. This review summarizes the recent data on general and subtype-specific features of midbrain DA neurons and their development. Here, we review the current protocols for derivation of DA neurons from human iPS cells and outline their general weak spots. The associated gaps in the contemporary knowledge are considered and the possible directions for future research that may assist in improving the differentiation conditions and increase the efficiency of using iPS cell-derived neurons for PD drug development are discussed.
Collapse
|
30
|
Neurothreads: Development of supportive carriers for mature dopaminergic neuron differentiation and implantation. Biomaterials 2021; 270:120707. [PMID: 33601130 DOI: 10.1016/j.biomaterials.2021.120707] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 01/28/2021] [Accepted: 01/31/2021] [Indexed: 12/16/2022]
Abstract
In this study we present the use of elastic macroporous cryogels for differentiation and transplantation of mature neurons. We develop a coating suitable for long-term neuronal culture, including stem cell differentiation, by covalent immobilization of neural adhesion proteins. In the context of cell therapy for Parkinson's disease, we show compatibility with established dopaminergic differentiation of both immortalized mesencephalic progenitors - LUHMES - and human embryonic stem cells (hESCs). We adjust structural properties of the biomaterial to create carriers - Neurothreads - favourable for cell viability during transplantation. Finally, we show feasibility of preservation of mature neurons, supported by Neurothreads, one month after in-vivo transplantation. Preliminary data suggests that the Neurothread approach could provide more mature and less proliferative cells in vivo.
Collapse
|
31
|
Corti S, Bonjean R, Legier T, Rattier D, Melon C, Salin P, Toso EA, Kyba M, Kerkerian-Le Goff L, Maina F, Dono R. Enhanced differentiation of human induced pluripotent stem cells toward the midbrain dopaminergic neuron lineage through GLYPICAN-4 downregulation. Stem Cells Transl Med 2021; 10:725-742. [PMID: 33528918 PMCID: PMC8046045 DOI: 10.1002/sctm.20-0177] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 11/25/2020] [Accepted: 12/16/2020] [Indexed: 12/24/2022] Open
Abstract
Enhancing the differentiation potential of human induced pluripotent stem cells (hiPSC) into disease‐relevant cell types is instrumental for their widespread application in medicine. Here, we show that hiPSCs downregulated for the signaling modulator GLYPICAN‐4 (GPC4) acquire a new biological state characterized by increased hiPSC differentiation capabilities toward ventral midbrain dopaminergic (VMDA) neuron progenitors. This biological trait emerges both in vitro, upon exposing cells to VMDA neuronal differentiation signals, and in vivo, even when transplanting hiPSCs at the extreme conditions of floor‐plate stage in rat brains. Moreover, it is compatible with the overall neuronal maturation process toward acquisition of substantia nigra neuron identity. HiPSCs with downregulated GPC4 also retain self‐renewal and pluripotency in stemness conditions, in vitro, while losing tumorigenesis in vivo as assessed by flank xenografts. In conclusion, our results highlight GPC4 downregulation as a powerful approach to enhance generation of VMDA neurons. Outcomes may contribute to establish hiPSC lines suitable for translational applications.
Collapse
Affiliation(s)
- Serena Corti
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, Parc Scientifique de Luminy, NeuroMarseille, Marseille, France
| | - Remi Bonjean
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, Parc Scientifique de Luminy, NeuroMarseille, Marseille, France
| | - Thomas Legier
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, Parc Scientifique de Luminy, NeuroMarseille, Marseille, France
| | - Diane Rattier
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, Parc Scientifique de Luminy, NeuroMarseille, Marseille, France
| | - Christophe Melon
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, Parc Scientifique de Luminy, NeuroMarseille, Marseille, France
| | - Pascal Salin
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, Parc Scientifique de Luminy, NeuroMarseille, Marseille, France
| | - Erik A Toso
- Lillehei Heart Institute and Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael Kyba
- Lillehei Heart Institute and Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lydia Kerkerian-Le Goff
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, Parc Scientifique de Luminy, NeuroMarseille, Marseille, France
| | - Flavio Maina
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, Parc Scientifique de Luminy, NeuroMarseille, Marseille, France
| | - Rosanna Dono
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, Parc Scientifique de Luminy, NeuroMarseille, Marseille, France
| |
Collapse
|
32
|
Muddapu VR, Chakravarthy VS. Influence of energy deficiency on the subcellular processes of Substantia Nigra Pars Compacta cell for understanding Parkinsonian neurodegeneration. Sci Rep 2021; 11:1754. [PMID: 33462293 PMCID: PMC7814067 DOI: 10.1038/s41598-021-81185-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 12/23/2020] [Indexed: 01/29/2023] Open
Abstract
Parkinson's disease (PD) is the second most prominent neurodegenerative disease around the world. Although it is known that PD is caused by the loss of dopaminergic cells in substantia nigra pars compacta (SNc), the decisive cause of this inexorable cell loss is not clearly elucidated. We hypothesize that "Energy deficiency at a sub-cellular/cellular/systems level can be a common underlying cause for SNc cell loss in PD." Here, we propose a comprehensive computational model of SNc cell, which helps us to understand the pathophysiology of neurodegeneration at the subcellular level in PD. The aim of the study is to see how deficits in the supply of energy substrates (glucose and oxygen) lead to a deficit in adenosine triphosphate (ATP). The study also aims to show that deficits in ATP are the common factor underlying the molecular-level pathological changes, including alpha-synuclein aggregation, reactive oxygen species formation, calcium elevation, and dopamine dysfunction. The model suggests that hypoglycemia plays a more crucial role in leading to ATP deficits than hypoxia. We believe that the proposed model provides an integrated modeling framework to understand the neurodegenerative processes underlying PD.
Collapse
Affiliation(s)
- Vignayanandam Ravindernath Muddapu
- grid.417969.40000 0001 2315 1926Computational Neuroscience Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Sardar Patel Road, Chennai, 600036 Tamil Nadu India
| | - V. Srinivasa Chakravarthy
- grid.417969.40000 0001 2315 1926Computational Neuroscience Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Sardar Patel Road, Chennai, 600036 Tamil Nadu India
| |
Collapse
|
33
|
Single-Cell Technologies in Parkinson׳s Disease. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11613-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
34
|
A new mouse tool for studying dopaminergic neurons. J Neurosci Methods 2020; 347:108968. [PMID: 33039413 DOI: 10.1016/j.jneumeth.2020.108968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 10/06/2020] [Accepted: 10/06/2020] [Indexed: 11/21/2022]
Abstract
Dopaminergic neurons play important roles in brain function and studying dopaminergic system has been a major field of neuroscience research. Genetically modified mice have greatly advanced research of dopaminergic neurons. However, it has been shown that dopaminergic neurons contain several subtypes expressing distinct genetic markers, and it is challenging to study genetically different subtypes of dopaminergic neurons using current mouse lines. Here, we report a newly developed Dat-ires-flp line, in which flippase gene is knocked in after the stop codon of Slc6a3 (dopamine transporter, DAT) gene. We validated this line to show that flp is specifically expressed in dopaminergic neurons, and by breeding with Cre-expressing line (i.e. Vglut2-cre) we can study a specific subpopulation of dopaminergic neurons. We expect this line will be widely used to study different subtypes of dopaminergic neurons in diverse contexts.
Collapse
|
35
|
Yang S, Boudier-Revéret M, Choo YJ, Chang MC. Association between Chronic Pain and Alterations in the Mesolimbic Dopaminergic System. Brain Sci 2020; 10:brainsci10100701. [PMID: 33023226 PMCID: PMC7600461 DOI: 10.3390/brainsci10100701] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Chronic pain (pain lasting for >3 months) decreases patient quality of life and even occupational abilities. It can be controlled by treatment, but often persists even after management. To properly control pain, its underlying mechanisms must be determined. This review outlines the role of the mesolimbic dopaminergic system in chronic pain. The mesolimbic system, a neural circuit, delivers dopamine from the ventral tegmental area to neural structures such as the nucleus accumbens, prefrontal cortex, anterior cingulate cortex, and amygdala. It controls executive, affective, and motivational functions. Chronic pain patients suffer from low dopamine production and delivery in this system. The volumes of structures constituting the mesolimbic system are known to be decreased in such patients. Studies on administration of dopaminergic drugs to control chronic pain, with a focus on increasing low dopamine levels in the mesolimbic system, show that it is effective in patients with Parkinson’s disease, restless legs syndrome, fibromyalgia, dry mouth syndrome, lumbar radicular pain, and chronic back pain. However, very few studies have confirmed these effects, and dopaminergic drugs are not commonly used to treat the various diseases causing chronic pain. Thus, further studies are required to determine the effectiveness of such treatment for chronic pain.
Collapse
Affiliation(s)
- Seoyon Yang
- Department of Rehabilitation Medicine, Ewha Woman’s University Seoul Hospital, Ewha Woman’s University School of Medicine, Seoul 07804, Korea;
| | - Mathieu Boudier-Revéret
- Department of Physical Medicine and Rehabilitation, Centre Hospitalier de l’Université de Montréal, Montreal, QC H2W 1T8, Canada;
| | - Yoo Jin Choo
- Production R&D Division Advanced Interdisciplinary Team, Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Deagu 41061, Korea;
| | - Min Cheol Chang
- Department of Rehabilitation Medicine, College of Medicine, Yeungnam University, Daegu 42415, Korea
- Correspondence:
| |
Collapse
|
36
|
Eura N, Matsui TK, Luginbühl J, Matsubayashi M, Nanaura H, Shiota T, Kinugawa K, Iguchi N, Kiriyama T, Zheng C, Kouno T, Lan YJ, Kongpracha P, Wiriyasermkul P, Sakaguchi YM, Nagata R, Komeda T, Morikawa N, Kitayoshi F, Jong M, Kobashigawa S, Nakanishi M, Hasegawa M, Saito Y, Shiromizu T, Nishimura Y, Kasai T, Takeda M, Kobayashi H, Inagaki Y, Tanaka Y, Makinodan M, Kishimoto T, Kuniyasu H, Nagamori S, Muotri AR, Shin JW, Sugie K, Mori E. Brainstem Organoids From Human Pluripotent Stem Cells. Front Neurosci 2020; 14:538. [PMID: 32670003 PMCID: PMC7332712 DOI: 10.3389/fnins.2020.00538] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/01/2020] [Indexed: 01/10/2023] Open
Abstract
The brainstem is a posterior region of the brain, composed of three parts, midbrain, pons, and medulla oblongata. It is critical in controlling heartbeat, blood pressure, and respiration, all of which are life-sustaining functions, and therefore, damages to or disorders of the brainstem can be lethal. Brain organoids derived from human pluripotent stem cells (hPSCs) recapitulate the course of human brain development and are expected to be useful for medical research on central nervous system disorders. However, existing organoid models are limited in the extent hPSCs recapitulate human brain development and hence are not able to fully elucidate the diseases affecting various components of the brain such as brainstem. Here, we developed a method to generate human brainstem organoids (hBSOs), containing midbrain/hindbrain progenitors, noradrenergic and cholinergic neurons, dopaminergic neurons, and neural crest lineage cells. Single-cell RNA sequence (scRNA-seq) analysis, together with evidence from proteomics and electrophysiology, revealed that the cellular population in these organoids was similar to that of the human brainstem, which raises the possibility of making use of hBSOs in investigating central nervous system disorders affecting brainstem and in efficient drug screenings.
Collapse
Affiliation(s)
- Nobuyuki Eura
- Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Takeshi K. Matsui
- Department of Neurology, Nara Medical University, Kashihara, Japan
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Joachim Luginbühl
- Laboratory for Advanced Genomics Circuit, RIKEN Center for Integrative Medical Sciences, Yokohama,Japan
| | - Masaya Matsubayashi
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Hitoki Nanaura
- Department of Neurology, Nara Medical University, Kashihara, Japan
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Tomo Shiota
- Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Kaoru Kinugawa
- Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Naohiko Iguchi
- Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Takao Kiriyama
- Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Canbin Zheng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Tsukasa Kouno
- Laboratory for Advanced Genomics Circuit, RIKEN Center for Integrative Medical Sciences, Yokohama,Japan
| | - Yan Jun Lan
- Laboratory for Advanced Genomics Circuit, RIKEN Center for Integrative Medical Sciences, Yokohama,Japan
| | - Pornparn Kongpracha
- Laboratory of Biomolecular Dynamics, Department of Collaborative Research, Nara Medical University, Kashihara, Japan
| | - Pattama Wiriyasermkul
- Laboratory of Biomolecular Dynamics, Department of Collaborative Research, Nara Medical University, Kashihara, Japan
| | | | - Riko Nagata
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Tomoya Komeda
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Naritaka Morikawa
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Fumika Kitayoshi
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Miyong Jong
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Shinko Kobashigawa
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Mari Nakanishi
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Masatoshi Hasegawa
- Department of Radiation Oncology, Nara Medical University, Kashihara, Japan
| | - Yasuhiko Saito
- Department of Neurophysiology, Nara Medical University, Kashihara, Japan
| | - Takashi Shiromizu
- Department of Integrative Pharmacology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Yuhei Nishimura
- Department of Integrative Pharmacology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Takahiko Kasai
- Department of Laboratory Medicine and Pathology, National Hospital Organization Kinki-Chuo Chest Medical Center, Sakai, Japan
| | - Maiko Takeda
- Department of Laboratory Medicine and Pathology, National Hospital Organization Kinki-Chuo Chest Medical Center, Sakai, Japan
| | - Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Yusuke Inagaki
- Department of Orthopaedic Surgery, Nara Medical University, Kashihara, Japan
| | - Yasuhito Tanaka
- Department of Orthopaedic Surgery, Nara Medical University, Kashihara, Japan
| | - Manabu Makinodan
- Department of Psychiatry, Nara Medical University, Kashihara, Japan
| | | | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Shushi Nagamori
- Laboratory of Biomolecular Dynamics, Department of Collaborative Research, Nara Medical University, Kashihara, Japan
| | - Alysson R. Muotri
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, United States
| | - Jay W. Shin
- Laboratory for Advanced Genomics Circuit, RIKEN Center for Integrative Medical Sciences, Yokohama,Japan
| | - Kazuma Sugie
- Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Eiichiro Mori
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| |
Collapse
|
37
|
Brignani S, Raj DDA, Schmidt ERE, Düdükcü Ö, Adolfs Y, De Ruiter AA, Rybiczka-Tesulov M, Verhagen MG, van der Meer C, Broekhoven MH, Moreno-Bravo JA, Grossouw LM, Dumontier E, Cloutier JF, Chédotal A, Pasterkamp RJ. Remotely Produced and Axon-Derived Netrin-1 Instructs GABAergic Neuron Migration and Dopaminergic Substantia Nigra Development. Neuron 2020; 107:684-702.e9. [PMID: 32562661 DOI: 10.1016/j.neuron.2020.05.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/17/2020] [Accepted: 05/26/2020] [Indexed: 12/18/2022]
Abstract
The midbrain dopamine (mDA) system is composed of molecularly and functionally distinct neuron subtypes that mediate specific behaviors and show select disease vulnerability, including in Parkinson's disease. Despite progress in identifying mDA neuron subtypes, how these neuronal subsets develop and organize into functional brain structures remains poorly understood. Here we generate and use an intersectional genetic platform, Pitx3-ITC, to dissect the mechanisms of substantia nigra (SN) development and implicate the guidance molecule Netrin-1 in the migration and positioning of mDA neuron subtypes in the SN. Unexpectedly, we show that Netrin-1, produced in the forebrain and provided to the midbrain through axon projections, instructs the migration of GABAergic neurons into the ventral SN. This migration is required to confine mDA neurons to the dorsal SN. These data demonstrate that neuron migration can be controlled by remotely produced and axon-derived secreted guidance cues, a principle that is likely to apply more generally.
Collapse
Affiliation(s)
- Sara Brignani
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Divya D A Raj
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Ewoud R E Schmidt
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Özge Düdükcü
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Anna A De Ruiter
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Mateja Rybiczka-Tesulov
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Marieke G Verhagen
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Christiaan van der Meer
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Mark H Broekhoven
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Juan A Moreno-Bravo
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 17 Rue Moreau, 75012 Paris, France
| | - Laurens M Grossouw
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Emilie Dumontier
- Montreal Neurological Institute, 3801 University, Montréal, QC H3A 2B4, Canada
| | | | - Alain Chédotal
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 17 Rue Moreau, 75012 Paris, France
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands.
| |
Collapse
|
38
|
Cardoso T, Lévesque M. Toward Generating Subtype-Specific Mesencephalic Dopaminergic Neurons in vitro. Front Cell Dev Biol 2020; 8:443. [PMID: 32626706 PMCID: PMC7311634 DOI: 10.3389/fcell.2020.00443] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022] Open
Abstract
Mesencephalic dopaminergic (mDA) neurons derived from pluripotent stem cells (PSCs) have proven to be pivotal for disease modeling studies and as a source of transplantable tissue for regenerative therapies in Parkinson's disease (PD). Current differentiation protocols can generate standardized and reproducible cell products of dopaminergic neurons that elicit the characteristic transcriptional profile of ventral midbrain. Nonetheless, dopamine neurons residing in the mesencephalon comprise distinct groups of cells within diffusely defined anatomical boundaries and with distinct functional, electrophysiological, and molecular properties. Here we review recent single cell sequencing studies that are shedding light on the neuronal heterogeneity within the mesencephalon and discuss how resolving the complex molecular profile of distinct sub-populations within this region could help refine patterning and quality control assessment of PSC-derived mDA neurons to subtype-specificity in vitro. In turn, such advances would have important impact in improving cell replacement therapy, disease mechanistic studies and drug screening in PD.
Collapse
Affiliation(s)
- Tiago Cardoso
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.,CERVO Brain Research Center, Université Laval, Québec, QC, Canada
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.,CERVO Brain Research Center, Université Laval, Québec, QC, Canada
| |
Collapse
|
39
|
Molecular Regulation in Dopaminergic Neuron Development. Cues to Unveil Molecular Pathogenesis and Pharmacological Targets of Neurodegeneration. Int J Mol Sci 2020; 21:ijms21113995. [PMID: 32503161 PMCID: PMC7312927 DOI: 10.3390/ijms21113995] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022] Open
Abstract
The relatively few dopaminergic neurons in the mammalian brain are mostly located in the midbrain and regulate many important neural functions, including motor integration, cognition, emotive behaviors and reward. Therefore, alteration of their function or degeneration leads to severe neurological and neuropsychiatric diseases. Unraveling the mechanisms of midbrain dopaminergic (mDA) phenotype induction and maturation and elucidating the role of the gene network involved in the development and maintenance of these neurons is of pivotal importance to rescue or substitute these cells in order to restore dopaminergic functions. Recently, in addition to morphogens and transcription factors, microRNAs have been identified as critical players to confer mDA identity. The elucidation of the gene network involved in mDA neuron development and function will be crucial to identify early changes of mDA neurons that occur in pre-symptomatic pathological conditions, such as Parkinson’s disease. In addition, it can help to identify targets for new therapies and for cell reprogramming into mDA neurons. In this essay, we review the cascade of transcriptional and posttranscriptional regulation that confers mDA identity and regulates their functions. Additionally, we highlight certain mechanisms that offer important clues to unveil molecular pathogenesis of mDA neuron dysfunction and potential pharmacological targets for the treatment of mDA neuron dysfunction.
Collapse
|
40
|
Perez-Bonilla P, Santiago-Colon K, Leinninger GM. Lateral hypothalamic area neuropeptides modulate ventral tegmental area dopamine neurons and feeding. Physiol Behav 2020; 223:112986. [PMID: 32492498 DOI: 10.1016/j.physbeh.2020.112986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 05/19/2020] [Accepted: 05/26/2020] [Indexed: 01/26/2023]
Abstract
Understanding how the brain coordinates energy status with the motivation to eat is crucial to identify strategies to improve disordered body weight. The ventral tegmental area (VTA), known as the core of the mesolimbic system, is of particular interest in this regard because it controls the motivation to consume palatable, calorie-dense foods and to engage in volitional activity. The VTA is largely composed of dopamine (DA) neurons, but modulating these DA neurons has been alternately linked with promoting and suppressing feeding, suggesting heterogeneity in their function. Subsets of VTA DA neurons have recently been described based on their anatomical distribution, electrophysiological features, connectivity and molecular expression, but to date there are no signatures to categorize how DA neurons control feeding. Assessing the neuropeptide receptors expressed by VTA DA neurons may be useful in this regard, as many neuropeptides mediate anorexic or orexigenic responses. In particular, the lateral hypothalamic area (LHA) releases a wide variety of feeding-modulating neuropeptides to the VTA. Since VTA neurons intercept LHA neuropeptides known to either evoke or suppress feeding, expression of the cognate neuropeptide receptors within the VTA may point to VTA DA neuronal mechanisms to promote or suppress feeding, respectively. Here we review the role of the VTA in energy balance and the LHA neuropeptide signaling systems that act in the VTA, whose receptors might be used to classify how VTA DA neurons contribute to energy balance.
Collapse
Affiliation(s)
- Patricia Perez-Bonilla
- Neuroscience Graduate Program, USA; Pharmacology and Toxicology Graduate Program, USA; Michigan State University, East Lansing, MI 48114, USA
| | - Krystal Santiago-Colon
- Department of Biology, University of Puerto Rico - Cayey, USA; Bridge to the PhD in Neuroscience Program, USA
| | - Gina M Leinninger
- Department of Physiology, USA; Michigan State University, East Lansing, MI 48114, USA.
| |
Collapse
|
41
|
Poulin JF, Gaertner Z, Moreno-Ramos OA, Awatramani R. Classification of Midbrain Dopamine Neurons Using Single-Cell Gene Expression Profiling Approaches. Trends Neurosci 2020; 43:155-169. [PMID: 32101709 PMCID: PMC7285906 DOI: 10.1016/j.tins.2020.01.004] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/13/2019] [Accepted: 01/11/2020] [Indexed: 01/31/2023]
Abstract
Dysfunctional dopamine (DA) signaling has been associated with a broad spectrum of neuropsychiatric disorders, prompting investigations into how midbrain DA neuron heterogeneity may underpin this variety of behavioral symptoms. Emerging literature indeed points to functional heterogeneity even within anatomically defined DA clusters. Recognizing the need for a systematic classification scheme, several groups have used single-cell profiling to catalog DA neurons based on their gene expression profiles. We aim here not only to synthesize points of congruence but also to highlight key differences between the molecular classification schemes derived from these studies. In doing so, we hope to provide a common framework that will facilitate investigations into the functions of DA neuron subtypes in the healthy and diseased brain.
Collapse
Affiliation(s)
- Jean-Francois Poulin
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Zachary Gaertner
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Rajeshwar Awatramani
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
42
|
Brodski C, Blaess S, Partanen J, Prakash N. Crosstalk of Intercellular Signaling Pathways in the Generation of Midbrain Dopaminergic Neurons In Vivo and from Stem Cells. J Dev Biol 2019; 7:jdb7010003. [PMID: 30650592 PMCID: PMC6473842 DOI: 10.3390/jdb7010003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 12/25/2022] Open
Abstract
Dopamine-synthesizing neurons located in the mammalian ventral midbrain are at the center stage of biomedical research due to their involvement in severe human neuropsychiatric and neurodegenerative disorders, most prominently Parkinson’s Disease (PD). The induction of midbrain dopaminergic (mDA) neurons depends on two important signaling centers of the mammalian embryo: the ventral midline or floor plate (FP) of the neural tube, and the isthmic organizer (IsO) at the mid-/hindbrain boundary (MHB). Cells located within and close to the FP secrete sonic hedgehog (SHH), and members of the wingless-type MMTV integration site family (WNT1/5A), as well as bone morphogenetic protein (BMP) family. The IsO cells secrete WNT1 and the fibroblast growth factor 8 (FGF8). Accordingly, the FGF8, SHH, WNT, and BMP signaling pathways play crucial roles during the development of the mDA neurons in the mammalian embryo. Moreover, these morphogens are essential for the generation of stem cell-derived mDA neurons, which are critical for the modeling, drug screening, and cell replacement therapy of PD. This review summarizes our current knowledge about the functions and crosstalk of these signaling pathways in mammalian mDA neuron development in vivo and their applications in stem cell-based paradigms for the efficient derivation of these neurons in vitro.
Collapse
Affiliation(s)
- Claude Brodski
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel.
| | - Sandra Blaess
- Institute of Reconstructive Neurobiology, University of Bonn Medical Center, 53127 Bonn, Germany.
| | - Juha Partanen
- Faculty of Biological and Environmental Sciences, FIN00014-University of Helsinki, P.O. Box 56, Viikinkaari 9, FIN-00014 Helsinki, Finland.
| | - Nilima Prakash
- Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, 59063 Hamm, Germany.
| |
Collapse
|
43
|
Kang DS, Yang YR, Lee C, Park B, Park KI, Seo JK, Seo YK, Cho H, Lucio C, Suh PG. Netrin-1/DCC-mediated PLCγ1 activation is required for axon guidance and brain structure development. EMBO Rep 2018; 19:embr.201846250. [PMID: 30224412 DOI: 10.15252/embr.201846250] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 08/11/2018] [Accepted: 08/23/2018] [Indexed: 11/09/2022] Open
Abstract
Coordinated expression of guidance molecules and their signal transduction are critical for correct brain wiring. Previous studies have shown that phospholipase C gamma1 (PLCγ1), a signal transducer of receptor tyrosine kinases, plays a specific role in the regulation of neuronal cell morphology and motility in vitro However, several questions remain regarding the extracellular stimulus that triggers PLCγ1 signaling and the exact role PLCγ1 plays in nervous system development. Here, we demonstrate that PLCγ1 mediates axonal guidance through a netrin-1/deleted in colorectal cancer (DCC) complex. Netrin-1/DCC activates PLCγ1 through Src kinase to induce actin cytoskeleton rearrangement. Neuronal progenitor-specific knockout of Plcg1 in mice causes axon guidance defects in the dorsal part of the mesencephalon during embryogenesis. Adult Plcg1-deficient mice exhibit structural alterations in the corpus callosum, substantia innominata, and olfactory tubercle. These results suggest that PLCγ1 plays an important role in the correct development of white matter structure by mediating netrin-1/DCC signaling.
Collapse
Affiliation(s)
- Du-Seock Kang
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea.,College of Life Science & Bioengineering, Korea Advanced Institute of Science & Technology (KAIST), Daejeon, Korea
| | - Yong Ryoul Yang
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Cheol Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - BumWoo Park
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Kwang Il Park
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Jeong Kon Seo
- UNIST Central Research Facility, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Young Kyo Seo
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - HyungJoon Cho
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Cocco Lucio
- Cellular Signaling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Pann-Ghill Suh
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| |
Collapse
|
44
|
Tapia M, Baudot P, Formisano-Tréziny C, Dufour MA, Temporal S, Lasserre M, Marquèze-Pouey B, Gabert J, Kobayashi K, Goaillard JM. Neurotransmitter identity and electrophysiological phenotype are genetically coupled in midbrain dopaminergic neurons. Sci Rep 2018; 8:13637. [PMID: 30206240 PMCID: PMC6134142 DOI: 10.1038/s41598-018-31765-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/22/2018] [Indexed: 01/04/2023] Open
Abstract
Most neuronal types have a well-identified electrical phenotype. It is now admitted that a same phenotype can be produced using multiple biophysical solutions defined by ion channel expression levels. This argues that systems-level approaches are necessary to understand electrical phenotype genesis and stability. Midbrain dopaminergic (DA) neurons, although quite heterogeneous, exhibit a characteristic electrical phenotype. However, the quantitative genetic principles underlying this conserved phenotype remain unknown. Here we investigated the quantitative relationships between ion channels’ gene expression levels in midbrain DA neurons using single-cell microfluidic qPCR. Using multivariate mutual information analysis to decipher high-dimensional statistical dependences, we unravel co-varying gene modules that link neurotransmitter identity and electrical phenotype. We also identify new segregating gene modules underlying the diversity of this neuronal population. We propose that the newly identified genetic coupling between neurotransmitter identity and ion channels may play a homeostatic role in maintaining the electrophysiological phenotype of midbrain DA neurons.
Collapse
Affiliation(s)
- Mónica Tapia
- Unité de Neurobiologie des Canaux Ioniques et de la Synapse, INSERM UMR 1072, Aix Marseille Université, 13015, Marseille, France
| | - Pierre Baudot
- Unité de Neurobiologie des Canaux Ioniques et de la Synapse, INSERM UMR 1072, Aix Marseille Université, 13015, Marseille, France
| | - Christine Formisano-Tréziny
- Unité de Neurobiologie des Canaux Ioniques et de la Synapse, INSERM UMR 1072, Aix Marseille Université, 13015, Marseille, France
| | - Martial A Dufour
- Unité de Neurobiologie des Canaux Ioniques et de la Synapse, INSERM UMR 1072, Aix Marseille Université, 13015, Marseille, France
| | - Simone Temporal
- Unité de Neurobiologie des Canaux Ioniques et de la Synapse, INSERM UMR 1072, Aix Marseille Université, 13015, Marseille, France
| | - Manon Lasserre
- Unité de Neurobiologie des Canaux Ioniques et de la Synapse, INSERM UMR 1072, Aix Marseille Université, 13015, Marseille, France
| | - Béatrice Marquèze-Pouey
- Unité de Neurobiologie des Canaux Ioniques et de la Synapse, INSERM UMR 1072, Aix Marseille Université, 13015, Marseille, France
| | - Jean Gabert
- Unité de Neurobiologie des Canaux Ioniques et de la Synapse, INSERM UMR 1072, Aix Marseille Université, 13015, Marseille, France.,Département de Biochimie et Biologie Moléculaire, Hôpital Nord, Marseille, France
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Jean-Marc Goaillard
- Unité de Neurobiologie des Canaux Ioniques et de la Synapse, INSERM UMR 1072, Aix Marseille Université, 13015, Marseille, France.
| |
Collapse
|
45
|
Kelly EA, Fudge JL. The neuroanatomic complexity of the CRF and DA systems and their interface: What we still don't know. Neurosci Biobehav Rev 2018; 90:247-259. [PMID: 29704516 PMCID: PMC5993645 DOI: 10.1016/j.neubiorev.2018.04.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 04/14/2018] [Accepted: 04/15/2018] [Indexed: 12/28/2022]
Abstract
Corticotropin-releasing factor (CRF) is a neuropeptide that mediates the stress response. Long known to contribute to regulation of the adrenal stress response initiated in the hypothalamic-pituitary axis (HPA), a complex pattern of extrahypothalamic CRF expression is also described in rodents and primates. Cross-talk between the CRF and midbrain dopamine (DA) systems links the stress response to DA regulation. Classically CRF + cells in the extended amygdala and paraventricular nucleus (PVN) are considered the main source of this input, principally targeting the ventral tegmental area (VTA). However, the anatomic complexity of both the DA and CRF system has been increasingly elaborated in the last decade. The DA neurons are now recognized as having diverse molecular, connectional and physiologic properties, predicted by their anatomic location. At the same time, the broad distribution of CRF cells in the brain has been increasingly delineated using different species and techniques. Here, we review updated information on both CRF localization and newer conceptualizations of the DA system to reconsider the CRF-DA interface.
Collapse
Affiliation(s)
- E A Kelly
- University of Rochester, School of Medicine and Dentistry, The Ernest J Del Monte Institute for Neuroscience, Department of Neuroscience, Rochester, NY, United States
| | - J L Fudge
- University of Rochester, School of Medicine and Dentistry, The Ernest J Del Monte Institute for Neuroscience, Department of Neuroscience, Rochester, NY, United States; University of Rochester, School of Medicine and Dentistry, The Ernest J Del Monte Institute for Neuroscience, Department of Psychiatry, Rochester, NY, United States.
| |
Collapse
|
46
|
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: 1.3] [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
|
47
|
Cell-Specific RNA Quantification in Human SN DA Neurons from Heterogeneous Post-mortem Midbrain Samples by UV-Laser Microdissection and RT-qPCR. Methods Mol Biol 2018; 1723:335-360. [PMID: 29344870 DOI: 10.1007/978-1-4939-7558-7_19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cell specificity of gene expression analysis is from particular relevance when the abundance of target cells is not homogeneous in the compared tissue samples, like it is the case, e.g., when comparing brain tissues from controls and in neurodegenerative disease states. While single-cell gene expression profiling is already a methodological challenge per se, it becomes even more prone to artifacts when analyzing individual cells from human post-mortem samples. Not only because human samples can never be matched as precisely as those from animal models, but also, because the RNA-quality that can be obtained from human samples usually displays a high range of variability. Here, we detail our most actual method for combining contact-free UV-laser microdissection (UV-LMD) with reverse transcription and quantitative PCR (RT-qPCR) that addresses all these issues. We specifically optimized our protocols to quantify and compare mRNA as well as miRNA levels in human neurons from post-mortem brain tissue. As human post-mortem tissue samples are never perfectly matched (e.g., in respect to distinct donor ages and RNA integrity numbers RIN), we refined data analysis by applying a linear mixed effects model to RT-qPCR data, which allows dissecting and subtracting linear contributions of distinct confounders on detected gene expression levels (i.e., RIN, age). All these issues were considered for comparative gene expression analysis in dopamine (DA) midbrain neurons of the Substantia nigra (SN) from controls and Parkinson's disease (PD) specimens, as the preferential degeneration of SN DA neurons in the pathological hallmark of PD. By utilizing the here-described protocol we identified that a variety of genes-encoding for ion channels, dopamine metabolism proteins, and PARK gene products-display a transcriptional dysregulation in remaining human SN DA neurons from PD brains compared to those of controls. We show that the linear mixed effects model allows further stratification of RT-qPCR data, as it indicated that differential gene expression of some genes was rather correlated with different ages of the analyzed human brain samples than with the disease state.
Collapse
|
48
|
Rizzi G, Tan KR. Dopamine and Acetylcholine, a Circuit Point of View in Parkinson's Disease. Front Neural Circuits 2017; 11:110. [PMID: 29311846 PMCID: PMC5744635 DOI: 10.3389/fncir.2017.00110] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 12/14/2017] [Indexed: 12/30/2022] Open
Abstract
Data from the World Health Organization (National Institute on Aging, 2011) and the National Institutes of Health (He et al., 2016) predicts that while today the worldwide population over 65 years of age is estimated around 8.5%, this number will reach an astounding 17% by 2050. In this framework, solving current neurodegenerative diseases primarily associated with aging becomes more pressing than ever. In 2017, we celebrate a grim 200th anniversary since the very first description of Parkinson’s disease (PD) and its related symptomatology. Two centuries after this debilitating disease was first identified, finding a cure remains a hopeful goal rather than an attainable objective on the horizon. Tireless work has provided insight into the characterization and progression of the disease down to a molecular level. We now know that the main motor deficits associated with PD arise from the almost total loss of dopaminergic cells in the substantia nigra pars compacta. A concomitant loss of cholinergic cells entails a cognitive decline in these patients, and current therapies are only partially effective, often inducing side-effects after a prolonged treatment. This review covers some of the recent developments in the field of Basal Ganglia (BG) function in physiology and pathology, with a particular focus on the two main neuromodulatory systems known to be severely affected in PD, highlighting some of the remaining open question from three main stand points: - Heterogeneity of midbrain dopamine neurons. - Pairing of dopamine (DA) sub-circuits. - Dopamine-Acetylcholine (ACh) interaction. A vast amount of knowledge has been accumulated over the years from experimental conditions, but very little of it is reflected or used at a translational or clinical level. An initiative to implement the knowledge that is emerging from circuit-based approaches to tackle neurodegenerative disorders like PD will certainly be tremendously beneficial.
Collapse
Affiliation(s)
| | - Kelly R Tan
- Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
49
|
Parkinson's Disease Is Not Simply a Prion Disorder. J Neurosci 2017; 37:9799-9807. [PMID: 29021297 DOI: 10.1523/jneurosci.1787-16.2017] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 06/09/2017] [Accepted: 06/17/2017] [Indexed: 12/31/2022] Open
Abstract
The notion that prion-like spreading of misfolded α-synuclein (α-SYN) causes Parkinson's disease (PD) has received a great deal of attention. Although attractive in its simplicity, the hypothesis is difficult to reconcile with postmortem analysis of human brains and connectome-mapping studies. An alternative hypothesis is that PD pathology is governed by regional or cell-autonomous factors. Although these factors provide an explanation for the pattern of neuronal loss in PD, they do not readily explain the apparently staged distribution of Lewy pathology in many PD brains, the feature of the disease that initially motivated the spreading hypothesis by Braak and colleagues. While each hypothesis alone has its shortcomings, a synthesis of the two can explain much of what we know about the etiopathology of PD.Dual Perspectives Companion Paper: Prying into the Prion Hypothesis for Parkinson's Disease, by Patrik Brundin and Ronald Melki.
Collapse
|
50
|
Thomas TS, Baimel C, Borgland SL. Opioid and hypocretin neuromodulation of ventral tegmental area neuronal subpopulations. Br J Pharmacol 2017; 175:2825-2833. [PMID: 28849596 DOI: 10.1111/bph.13993] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/14/2017] [Accepted: 08/17/2017] [Indexed: 11/29/2022] Open
Abstract
The current view of the midbrain dopaminergic system is evolving towards a complex system of subpopulations of neurons with distinct afferent and efferent connections and, importantly, functionally different intrinsic characteristics. Recent literature on the phenotypic diversity of dopaminergic neurons has outlined that in the ventral tegmental area dopaminergic neurons are not as anatomically or electrophysiologically homogeneous as they were once thought to be. Instead, the midbrain dopaminergic system is now understood to be composed of anatomically and functionally heterogeneous dopaminergic subpopulations receiving specific afferent inputs and with different axonal projections. An additional layer of complexity is the neuromodulation of each of these dopaminergic circuits. This review will examine the distinguishing electrophysiological and neuromodulatory characteristics of the afferent and efferent connections of midbrain dopaminergic neurons. LINKED ARTICLES This article is part of a themed section on Emerging Areas of Opioid Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.14/issuetoc.
Collapse
Affiliation(s)
- Taylor S Thomas
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Corey Baimel
- Center for Neural Science, New York University, New York, NY, USA
| | | |
Collapse
|