1
|
Kim TW, Piao J, Bocchi VD, Koo SY, Choi SJ, Chaudhry F, Yang D, Cho HS, Hergenreder E, Perera LR, Joshi S, Mrad ZA, Claros N, Donohue SA, Frank AK, Walsh R, Mosharov EV, Betel D, Tabar V, Studer L. Enhanced yield and subtype identity of hPSC-derived midbrain dopamine neuron by modulation of WNT and FGF18 signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631400. [PMID: 39829874 PMCID: PMC11741396 DOI: 10.1101/2025.01.06.631400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
While clinical trials are ongoing using human pluripotent stem cell-derived midbrain dopamine (mDA) neuron precursor grafts in Parkinson's disease (PD), current protocols to derive mDA neurons remain suboptimal. In particular, the yield of TH+ mDA neurons after in vivo grafting and the expression of some mDA neuron and subtype-specific markers can be further improved. For example, characterization of mDA grafts by single cell transcriptomics has yielded only a small proportion of mDA neurons and a considerable fraction of contaminating cell populations. Here we present an optimized mDA neuron differentiation strategy that builds on our clinical grade ("Boost") protocol but includes the addition of FGF18 and IWP2 treatment ("Boost+") at the mDA neurogenesis stage. We demonstrate that Boost+ mDA neurons show higher expression of EN1, PITX3 and ALDH1A1. Improvements in both mDA neurons yield and transcriptional similarity to primary mDA neurons is observed both in vitro and in grafts. Furthermore, grafts are enriched in authentic A9 mDA neurons by single nucSeq. Functional studies in vitro demonstrate increased dopamine production and release and improved electrophysiological properties. In vivo analyses show increased percentages of TH+ mDA neurons resulting in efficient rescue of amphetamine induced rotation behavior in the 6-OHDA rat model and rescue of some motor deficits in non-drug induced assays, including the ladder rung assay that is not improved by Boost mDA neurons. The Boost+ conditions present an optimized protocol with advantages for disease modeling and mDA neuron grafting paradigms.
Collapse
|
2
|
Nolbrant S, Wallace JL, Ding J, Zhu T, Sevetson JL, Kajtez J, Baldacci IA, Corrigan EK, Hoglin K, McMullen R, Schmitz MT, Breevoort A, Swope D, Wu F, Pavlovic BJ, Salama SR, Kirkeby A, Huang H, Schaefer NK, Pollen AA. INTERSPECIES ORGANOIDS REVEAL HUMAN-SPECIFIC MOLECULAR FEATURES OF DOPAMINERGIC NEURON DEVELOPMENT AND VULNERABILITY. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.14.623592. [PMID: 39605599 PMCID: PMC11601475 DOI: 10.1101/2024.11.14.623592] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The disproportionate expansion of telencephalic structures during human evolution involved tradeoffs that imposed greater connectivity and metabolic demands on midbrain dopaminergic neurons. Despite the central role of dopaminergic neurons in human-enriched disorders, molecular specializations associated with human-specific features and vulnerabilities of the dopaminergic system remain unexplored. Here, we establish a phylogeny-in-a-dish approach to examine gene regulatory evolution by differentiating pools of human, chimpanzee, orangutan, and macaque pluripotent stem cells into ventral midbrain organoids capable of forming long-range projections, spontaneous activity, and dopamine release. We identify human-specific gene expression changes related to axonal transport of mitochondria and reactive oxygen species buffering and candidate cis- and trans-regulatory mechanisms underlying gene expression divergence. Our findings are consistent with a model of evolved neuroprotection in response to tradeoffs related to brain expansion and could contribute to the discovery of therapeutic targets and strategies for treating disorders involving the dopaminergic system.
Collapse
Affiliation(s)
- Sara Nolbrant
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- These authors contributed equally
| | - Jenelle L. Wallace
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- These authors contributed equally
| | - Jingwen Ding
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- These authors contributed equally
| | - Tianjia Zhu
- Department of Radiology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Jess L. Sevetson
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Cruz, CA, United States of America
- Genomics Institute, University of California Santa Cruz, CA, United States of America
| | - Janko Kajtez
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW)), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Isabella A. Baldacci
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Emily K. Corrigan
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Kaylynn Hoglin
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Reed McMullen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Matthew T. Schmitz
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Arnar Breevoort
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Dani Swope
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Fengxia Wu
- Department of Radiology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Anatomy and Neurobiology, Shandong University, Jinan, Shandong Province, China
| | - Bryan J. Pavlovic
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Sofie R. Salama
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Cruz, CA, United States of America
- Genomics Institute, University of California Santa Cruz, CA, United States of America
| | - Agnete Kirkeby
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW)), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Experimental Medical Sciences, Wallenberg Center for Molecular Medicine (WCMM) and Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Hao Huang
- Department of Radiology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Nathan K. Schaefer
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Alex A. Pollen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Lead contact
| |
Collapse
|
3
|
Xu P, He H, Gao Q, Zhou Y, Wu Z, Zhang X, Sun L, Hu G, Guan Q, You Z, Zhang X, Zheng W, Xiong M, Chen Y. Human midbrain dopaminergic neuronal differentiation markers predict cell therapy outcome in a Parkinson's disease model. J Clin Invest 2022; 132:156768. [PMID: 35700056 PMCID: PMC9282930 DOI: 10.1172/jci156768] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/07/2022] [Indexed: 11/17/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-based replacement therapy holds great promise in treating Parkinson's disease (PD). However, the heterogeneity of hPSC-derived donor cells and the low yield of midbrain dopaminergic (mDA) neurons after transplantation hinder its broad clinical application. Here, we depicted the single-cell molecular landscape during mDA neuron differentiation. We found that this process recapitulated the development of multiple but adjacent fetal brain regions including ventral midbrain, isthmus, and ventral hindbrain, resulting in heterogenous donor cell population. We reconstructed the differentiation trajectory of mDA lineage and identified CLSTN2 and PTPRO as specific surface markers of mDA progenitors, which were predictive of mDA neuron differentiation and could facilitate highly enriched mDA neurons (up to 80%) following progenitor sorting and transplantation. Marker sorted progenitors exhibited higher therapeutic potency in correcting motor deficits of PD mice. Different marker sorted grafts had a strikingly consistent cellular composition, in which mDA neurons were enriched, while off-target neuron types were mostly depleted, suggesting stable graft outcomes. Our study provides a better understanding of cellular heterogeneity during mDA neuron differentiation, and establishes a strategy to generate highly purified donor cells to achieve stable and predictable therapeutic outcomes, raising the prospect of hPSC-based PD cell replacement therapies.
Collapse
Affiliation(s)
- Peibo Xu
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Hui He
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Qinqin Gao
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yingying Zhou
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Ziyan Wu
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xiao Zhang
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Linyu Sun
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Gang Hu
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Qian Guan
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zhiwen You
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xinyue Zhang
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Wenping Zheng
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Man Xiong
- Institute State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Yuejun Chen
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
4
|
Liu Y, Deng J, Liu Y, Li W, Nie X. FGF, Mechanism of Action, Role in Parkinson's Disease, and Therapeutics. Front Pharmacol 2021; 12:675725. [PMID: 34234672 PMCID: PMC8255968 DOI: 10.3389/fphar.2021.675725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease associated with severe disability and adverse effects on life quality. In PD, motor dysfunction can occur, such as quiescence, muscle stiffness, and postural instability. PD is also associated with autonomic nervous dysfunction, sleep disorders, psychiatric symptoms, and other non-motor symptoms. Degeneration of dopaminergic neurons in the substantia nigra compact (SNPC), Lewy body, and neuroinflammation are the main pathological features of PD. The death or dysfunction of dopaminergic neurons in the dense part of the substantia nigra leads to dopamine deficiency in the basal ganglia and motor dysfunction. The formation of the Lewy body is associated with the misfolding of α-synuclein, which becomes insoluble and abnormally aggregated. Astrocytes and microglia mainly cause neuroinflammation, and the activation of a variety of pro-inflammatory transcription factors and regulatory proteins leads to the degeneration of dopaminergic neurons. At present, PD is mainly treated with drugs that increase dopamine concentration or directly stimulate dopamine receptors. Fibroblast growth factor (FGF) is a family of cellular signaling proteins strongly associated with neurodegenerative diseases such as PD. FGF and its receptor (FGFR) play an essential role in the development and maintenance of the nervous system as well as in neuroinflammation and have been shown to improve the survival rate of dopaminergic neurons. This paper summarized the mechanism of FGF and its receptors in the pathological process of PD and related signaling pathways, involving the development and protection of dopaminergic neurons in SNPC, α-synuclein aggregation, mitochondrial dysfunction, and neuroinflammation. It provides a reference for developing drugs to slow down or prevent the potential of PD.
Collapse
Affiliation(s)
- Yiqiu Liu
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Junyu Deng
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Ye Liu
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Wei Li
- College of Pharmacy, Zunyi Medical University, Zunyi, China
- Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Xuqiang Nie
- College of Pharmacy, Zunyi Medical University, Zunyi, China
- Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, College of Pharmacy, Zunyi Medical University, Zunyi, China
- Key Lab of the Basic Pharmacology of the Ministry of Education, College of Pharmacy, Zunyi Medical University, Zunyi, China
| |
Collapse
|
5
|
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: 145] [Impact Index Per Article: 29.0] [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
|
6
|
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: 23] [Impact Index Per Article: 3.8] [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
|
7
|
Kim JY, Lee JS, Hwang HS, Lee DR, Park CY, Jung SJ, You YR, Kim DS, Kim DW. Wnt signal activation induces midbrain specification through direct binding of the beta-catenin/TCF4 complex to the EN1 promoter in human pluripotent stem cells. Exp Mol Med 2018; 50:1-13. [PMID: 29650976 PMCID: PMC5938028 DOI: 10.1038/s12276-018-0044-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/30/2017] [Accepted: 12/19/2017] [Indexed: 12/12/2022] Open
Abstract
The canonical Wnt signal pathway plays a pivotal role in anteroposterior patterning and midbrain specification during early neurogenesis. Activating Wnt signal has been a strategy for differentiating human pluripotent stem cells (PSCs) into midbrain dopaminergic (DA) neurons; however, the underlying molecular mechanism(s) of how the Wnt signal drives posterior fate remained unclear. In this study, we found that activating the canonical Wnt signal significantly upregulated the expression of EN1, a midbrain-specific marker, in a fibroblast growth factor signal-dependent manner in human PSC-derived neural precursor cells (NPCs). The EN1 promoter region contains a putative TCF4-binding site that directly interacts with the β-catenin/TCF complex upon Wnt signal activation. Once differentiated, NPCs treated with a Wnt signal agonist gave rise to functional midbrain neurons including glutamatergic, GABAergic, and DA neurons. Our results provide a potential molecular mechanism that underlies midbrain specification of human PSC-derived NPCs by Wnt activation, as well as a differentiation paradigm for generating human midbrain neurons that may serve as a cellular platform for studying the ontogenesis of midbrain neurons and neurological diseases relevant to the midbrain.
Collapse
Affiliation(s)
- Ji Young Kim
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Korea.,Brain Korea 21 PLUS Program for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Jae Souk Lee
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Korea.,Brain Korea 21 PLUS Program for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Hyun Sub Hwang
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Korea
| | - Dongjin R Lee
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Korea
| | - Chul-Yong Park
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Korea
| | - Sung Jun Jung
- Department of Physiology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seoul, 04763, Korea
| | - Young Rang You
- Department of Biotechnology, Brain Korea 21 PLUS program for Biotechnology, College of Life Science & Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Korea
| | - Dae-Sung Kim
- Department of Biotechnology, Brain Korea 21 PLUS program for Biotechnology, College of Life Science & Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Korea.
| | - Dong-Wook Kim
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Korea. .,Brain Korea 21 PLUS Program for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| |
Collapse
|
8
|
Jha MK, Kim JH, Song GJ, Lee WH, Lee IK, Lee HW, An SSA, Kim S, Suk K. Functional dissection of astrocyte-secreted proteins: Implications in brain health and diseases. Prog Neurobiol 2017; 162:37-69. [PMID: 29247683 DOI: 10.1016/j.pneurobio.2017.12.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 10/23/2017] [Accepted: 12/08/2017] [Indexed: 02/07/2023]
Abstract
Astrocytes, which are homeostatic cells of the central nervous system (CNS), display remarkable heterogeneity in their morphology and function. Besides their physical and metabolic support to neurons, astrocytes modulate the blood-brain barrier, regulate CNS synaptogenesis, guide axon pathfinding, maintain brain homeostasis, affect neuronal development and plasticity, and contribute to diverse neuropathologies via secreted proteins. The identification of astrocytic proteome and secretome profiles has provided new insights into the maintenance of neuronal health and survival, the pathogenesis of brain injury, and neurodegeneration. Recent advances in proteomics research have provided an excellent catalog of astrocyte-secreted proteins. This review categorizes astrocyte-secreted proteins and discusses evidence that astrocytes play a crucial role in neuronal activity and brain function. An in-depth understanding of astrocyte-secreted proteins and their pathways is pivotal for the development of novel strategies for restoring brain homeostasis, limiting brain injury/inflammation, counteracting neurodegeneration, and obtaining functional recovery.
Collapse
Affiliation(s)
- Mithilesh Kumar Jha
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jong-Heon Kim
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Gyun Jee Song
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Ho-Won Lee
- Department of Neurology, Brain Science and Engineering Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Seong Soo A An
- Department of BioNano Technology, Gachon University, Gyeonggi-do, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Gyeonggi-do, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
9
|
Smidt MP. Molecular Programming of Mesodiencephalic Dopaminergic Neuronal Subsets. Front Neuroanat 2017; 11:59. [PMID: 28769772 PMCID: PMC5515899 DOI: 10.3389/fnana.2017.00059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/05/2017] [Indexed: 11/26/2022] Open
Abstract
Dopamine neurons of the substantia nigra compacta (SNc) and ventral tegmental area (VTA) are critical components of the neuronal machinery to control emotion and movement in mammals. The slow and gradual death of these neurons as seen in Parkinson's disease has triggered a large investment in research toward unraveling the molecular determinants that are used to generate these neurons and to get an insight in their apparent selective vulnerability. Here, I set out to summarize the current view on the molecular distinctions that exist within this mesodiencephalic dopamine (mdDA) system and elaborate on the molecular programming that is responsible for creating such diversity.
Collapse
Affiliation(s)
- Marten P Smidt
- Molecular NeuroScience, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
| |
Collapse
|
10
|
Shiraishi A, Muguruma K, Sasai Y. Generation of thalamic neurons from mouse embryonic stem cells. Development 2017; 144:1211-1220. [PMID: 28219951 DOI: 10.1242/dev.144071] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/03/2017] [Indexed: 01/08/2023]
Abstract
The thalamus is a diencephalic structure that plays crucial roles in relaying and modulating sensory and motor information to the neocortex. The thalamus develops in the dorsal part of the neural tube at the level of the caudal forebrain. However, the molecular mechanisms that are essential for thalamic differentiation are still unknown. Here, we have succeeded in generating thalamic neurons from mouse embryonic stem cells (mESCs) by modifying the default method that induces the most-anterior neural type in self-organizing culture. A low concentration of the caudalizing factor insulin and a MAPK/ERK kinase inhibitor enhanced the expression of the caudal forebrain markers Otx2 and Pax6. BMP7 promoted an increase in thalamic precursors such as Tcf7l2+/Gbx2+ and Tcf7l2+/Olig3+ cells. mESC thalamic precursors began to express the glutamate transporter vGlut2 and the axon-specific marker VGF, similar to mature projection neurons. The mESC thalamic neurons extended their axons to cortical layers in both organotypic culture and subcortical transplantation. Thus, we have identified the minimum elements sufficient for in vitro generation of thalamic neurons. These findings expand our knowledge of thalamic development.
Collapse
Affiliation(s)
- Atsushi Shiraishi
- Laboratory for Organogenesis and Neurogenesis, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan.,Laboratory of Growth Regulation, Institute for Virus Research, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.,Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Keiko Muguruma
- Laboratory for Organogenesis and Neurogenesis, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan .,Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Yoshiki Sasai
- Laboratory for Organogenesis and Neurogenesis, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| |
Collapse
|
11
|
Nouri N, Awatramani R. A novel floor plate boundary defined by adjacent En1 and Dbx1 microdomains distinguishes midbrain dopamine and hypothalamic neurons. Development 2017; 144:916-927. [PMID: 28174244 DOI: 10.1242/dev.144949] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/18/2017] [Indexed: 12/13/2022]
Abstract
The mesodiencephalic floor plate (mdFP) is the source of diverse neuron types. Yet, how this structure is compartmentalized has not been clearly elucidated. Here, we identify a novel boundary subdividing the mdFP into two microdomains, defined by engrailed 1 (En1) and developing brain homeobox 1 (Dbx1). Utilizing simultaneous dual and intersectional fate mapping, we demonstrate that this boundary is precisely formed with minimal overlap between En1 and Dbx1 microdomains, unlike many other boundaries. We show that the En1 microdomain gives rise to dopaminergic (DA) neurons, whereas the Dbx1 microdomain gives rise to subthalamic (STN), premammillary (PM) and posterior hypothalamic (PH) populations. To determine whether En1 is sufficient to induce DA neuron production beyond its normal limit, we generated a mouse strain that expresses En1 in the Dbx1 microdomain. In mutants, we observed ectopic production of DA neurons derived from the Dbx1 microdomain, at the expense of STN and PM populations. Our findings provide new insights into subdivisions in the mdFP, and will impact current strategies for the conversion of stem cells into DA neurons.
Collapse
Affiliation(s)
- Navid Nouri
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Rajeshwar Awatramani
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
12
|
Fernández-Santiago R, Carballo-Carbajal I, Castellano G, Torrent R, Richaud Y, Sánchez-Danés A, Vilarrasa-Blasi R, Sánchez-Pla A, Mosquera JL, Soriano J, López-Barneo J, Canals JM, Alberch J, Raya Á, Vila M, Consiglio A, Martín-Subero JI, Ezquerra M, Tolosa E. Aberrant epigenome in iPSC-derived dopaminergic neurons from Parkinson's disease patients. EMBO Mol Med 2015; 7:1529-46. [PMID: 26516212 PMCID: PMC4693505 DOI: 10.15252/emmm.201505439] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 09/24/2015] [Accepted: 09/28/2015] [Indexed: 12/13/2022] Open
Abstract
The epigenomic landscape of Parkinson's disease (PD) remains unknown. We performed a genomewide DNA methylation and a transcriptome studies in induced pluripotent stem cell (iPSC)-derived dopaminergic neurons (DAn) generated by cell reprogramming of somatic skin cells from patients with monogenic LRRK2-associated PD (L2PD) or sporadic PD (sPD), and healthy subjects. We observed extensive DNA methylation changes in PD DAn, and of RNA expression, which were common in L2PD and sPD. No significant methylation differences were present in parental skin cells, undifferentiated iPSCs nor iPSC-derived neural cultures not-enriched-in-DAn. These findings suggest the presence of molecular defects in PD somatic cells which manifest only upon differentiation into the DAn cells targeted in PD. The methylation profile from PD DAn, but not from controls, resembled that of neural cultures not-enriched-in-DAn indicating a failure to fully acquire the epigenetic identity own to healthy DAn in PD. The PD-associated hypermethylation was prominent in gene regulatory regions such as enhancers and was related to the RNA and/or protein downregulation of a network of transcription factors relevant to PD (FOXA1, NR3C1, HNF4A, and FOSL2). Using a patient-specific iPSC-based DAn model, our study provides the first evidence that epigenetic deregulation is associated with monogenic and sporadic PD.
Collapse
Affiliation(s)
- Rubén Fernández-Santiago
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Hospital Clínic of Barcelona Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) University of Barcelona (UB), Barcelona, Spain Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain Cell Therapy Program, Faculty of Medicine, University of Barcelona (UB), Barcelona, Spain
| | - Iria Carballo-Carbajal
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain Neurodegenerative Diseases Research Laboratory, Hospital Vall d'Hebron Vall d'Hebron Research Institute (VHIR) Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Giancarlo Castellano
- Department of Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona (UB) Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Roger Torrent
- Institute for Biomedicine (IBUB) University of Barcelona (UB), Barcelona, Spain
| | - Yvonne Richaud
- Control of Stem Cell Potency Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain Centre for Networked Biomedical Research on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| | | | - Roser Vilarrasa-Blasi
- Department of Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona (UB) Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Alex Sánchez-Pla
- Department of Statistics, University of Barcelona (UB), Barcelona, Spain Department of Statistics, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - José Luis Mosquera
- Department of Statistics, University of Barcelona (UB), Barcelona, Spain
| | - Jordi Soriano
- Departament d'Estructura i Constituents de la Matèria (ECM), Facultat de Física, University of Barcelona (UB), Barcelona, Spain
| | - José López-Barneo
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain Institute of Biomedicine of Seville (IBiS) Hospital Universitario Virgen del Rocío Consejo Superior de Investigaciones Científicas (CSIC) University of Seville, Seville, Spain
| | - Josep M Canals
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain Cell Therapy Program, Faculty of Medicine, University of Barcelona (UB), Barcelona, Spain Department of Cell Biology, Immunology and Neuroscience, Faculty of Medicine, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) University of Barcelona (UB), Barcelona, Spain
| | - Jordi Alberch
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain Cell Therapy Program, Faculty of Medicine, University of Barcelona (UB), Barcelona, Spain Department of Cell Biology, Immunology and Neuroscience, Faculty of Medicine, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) University of Barcelona (UB), Barcelona, Spain
| | - Ángel Raya
- Control of Stem Cell Potency Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain Centre for Networked Biomedical Research on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Miquel Vila
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain Neurodegenerative Diseases Research Laboratory, Hospital Vall d'Hebron Vall d'Hebron Research Institute (VHIR) Universitat Autònoma de Barcelona (UAB), Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Antonella Consiglio
- Institute for Biomedicine (IBUB) University of Barcelona (UB), Barcelona, Spain Department of Molecular and Translational Medicine, University of Brescia and National Institute of Neuroscience, Brescia, Italy
| | - José I Martín-Subero
- Department of Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona (UB) Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Mario Ezquerra
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Hospital Clínic of Barcelona Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) University of Barcelona (UB), Barcelona, Spain Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain Cell Therapy Program, Faculty of Medicine, University of Barcelona (UB), Barcelona, Spain
| | - Eduardo Tolosa
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Hospital Clínic of Barcelona Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) University of Barcelona (UB), Barcelona, Spain Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain Cell Therapy Program, Faculty of Medicine, University of Barcelona (UB), Barcelona, Spain Movement Disorders Unit, Department of Neurology, Hospital Clínic of Barcelona Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) University of Barcelona (UB), Barcelona, Spain
| |
Collapse
|
13
|
Gazea M, Tasouri E, Tolve M, Bosch V, Kabanova A, Gojak C, Kurtulmus B, Novikov O, Spatz J, Pereira G, Hübner W, Brodski C, Tucker KL, Blaess S. Primary cilia are critical for Sonic hedgehog-mediated dopaminergic neurogenesis in the embryonic midbrain. Dev Biol 2015; 409:55-71. [PMID: 26542012 DOI: 10.1016/j.ydbio.2015.10.033] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 10/21/2015] [Accepted: 10/30/2015] [Indexed: 02/07/2023]
Abstract
Midbrain dopaminergic (mDA) neurons modulate various motor and cognitive functions, and their dysfunction or degeneration has been implicated in several psychiatric diseases. Both Sonic Hedgehog (Shh) and Wnt signaling pathways have been shown to be essential for normal development of mDA neurons. Primary cilia are critical for the development of a number of structures in the brain by serving as a hub for essential developmental signaling cascades, but their role in the generation of mDA neurons has not been examined. We analyzed mutant mouse lines deficient in the intraflagellar transport protein IFT88, which is critical for primary cilia function. Conditional inactivation of Ift88 in the midbrain after E9.0 results in progressive loss of primary cilia, a decreased size of the mDA progenitor domain, and a reduction in mDA neurons. We identified Shh signaling as the primary cause of these defects, since conditional inactivation of the Shh signaling pathway after E9.0, through genetic ablation of Gli2 and Gli3 in the midbrain, results in a phenotype basically identical to the one seen in Ift88 conditional mutants. Moreover, the expansion of the mDA progenitor domain observed when Shh signaling is constitutively activated does not occur in absence of Ift88. In contrast, clusters of Shh-responding progenitors are maintained in the ventral midbrain of the hypomorphic Ift88 mouse mutant, cobblestone. Despite the residual Shh signaling, the integrity of the mDA progenitor domain is severely disturbed, and consequently very few mDA neurons are generated in cobblestone mutants. Our results identify for the first time a crucial role of primary cilia in the induction of mDA progenitors, define a narrow time window in which Shh-mediated signaling is dependent upon normal primary cilia function for this purpose, and suggest that later Wnt signaling-dependent events act independently of primary cilia.
Collapse
Affiliation(s)
- Mary Gazea
- Institute of Reconstructive Neurobiology, University of Bonn, 53127 Bonn, Germany
| | - Evangelia Tasouri
- Interdisciplinary Center for Neurosciences, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Marianna Tolve
- Institute of Reconstructive Neurobiology, University of Bonn, 53127 Bonn, Germany
| | - Viktoria Bosch
- Institute of Reconstructive Neurobiology, University of Bonn, 53127 Bonn, Germany
| | - Anna Kabanova
- Institute of Reconstructive Neurobiology, University of Bonn, 53127 Bonn, Germany
| | - Christian Gojak
- Department of Biophysical Chemistry, University of Heidelberg, 69120 Heidelberg, Germany; Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, 70569 Stuttgart, Germany
| | - Bahtiyar Kurtulmus
- Molecular Biology of Centrosomes and Cilia, German Cancer Research Center, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Orna Novikov
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Joachim Spatz
- Department of Biophysical Chemistry, University of Heidelberg, 69120 Heidelberg, Germany; Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, 70569 Stuttgart, Germany
| | - Gislene Pereira
- Molecular Biology of Centrosomes and Cilia, German Cancer Research Center, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Wolfgang Hübner
- Molecular Biophotonics, University of Bielefeld, 33615 Bielefeld, Germany
| | - Claude Brodski
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Kerry L Tucker
- Interdisciplinary Center for Neurosciences, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany; University of New England, College of Osteopathic Medicine, Department of Biomedical Sciences, Center for Excellence in the Neurosciences, Biddeford, ME 04005, USA.
| | - Sandra Blaess
- University of New England, College of Osteopathic Medicine, Department of Biomedical Sciences, Center for Excellence in the Neurosciences, Biddeford, ME 04005, USA.
| |
Collapse
|
14
|
Bodea GO, Blaess S. Establishing diversity in the dopaminergic system. FEBS Lett 2015; 589:3773-85. [PMID: 26431946 DOI: 10.1016/j.febslet.2015.09.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 09/13/2015] [Accepted: 09/16/2015] [Indexed: 11/19/2022]
Abstract
Midbrain dopaminergic neurons (MbDNs) modulate cognitive processes, regulate voluntary movement, and encode reward prediction errors and aversive stimuli. While the degeneration of MbDNs underlies the motor defects in Parkinson's disease, imbalances in dopamine levels are associated with neuropsychiatric disorders such as depression, schizophrenia and substance abuse. In recent years, progress has been made in understanding how MbDNs, which constitute a relatively small neuronal population in the brain, can contribute to such diverse functions and dysfunctions. In particular, important insights have been gained regarding the distinct molecular, neurochemical and network properties of MbDNs. How this diversity of MbDNs is established during brain development is only starting to be unraveled. In this review, we summarize the current knowledge on the diversity in MbDN progenitors and differentiated MbDNs in the developing rodent brain. We discuss the signaling pathways, transcription factors and transmembrane receptors that contribute to setting up these diverse MbDN subpopulations. A better insight into the processes that establish diversity in MbDNs will ultimately improve the understanding of the architecture and function of the dopaminergic system in the adult brain.
Collapse
Affiliation(s)
- Gabriela O Bodea
- Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia; Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - Sandra Blaess
- Institute of Reconstructive Neurobiology, Life and Brain Center, University of Bonn, Bonn, Germany.
| |
Collapse
|
15
|
Nouri N, Patel MJ, Joksimovic M, Poulin JF, Anderegg A, Taketo MM, Ma YC, Awatramani R. Excessive Wnt/beta-catenin signaling promotes midbrain floor plate neurogenesis, but results in vacillating dopamine progenitors. Mol Cell Neurosci 2015; 68:131-42. [PMID: 26164566 PMCID: PMC4633300 DOI: 10.1016/j.mcn.2015.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 06/30/2015] [Accepted: 07/04/2015] [Indexed: 01/10/2023] Open
Abstract
The floor plate (FP), a ventral midline structure of the developing neural tube, has differential neurogenic capabilities along the anterior-posterior axis. The midbrain FP, unlike the hindbrain and spinal cord floor plate, is highly neurogenic and produces midbrain dopaminergic (mDA) neurons. Canonical Wnt/beta-catenin signaling, at least in part, is thought to account for the difference in neurogenic capability. Removal of beta-catenin results in mDA progenitor specification defects as well as a profound reduction of neurogenesis. To examine the effects of excessive Wnt/beta-catenin signaling on mDA specification and neurogenesis, we have analyzed a model wherein beta-catenin is conditionally stabilized in the Shh+domain. Here, we show that the Foxa2+/Lmx1a+ domain is extended rostrally in mutant embryos, suggesting that canonical Wnt/beta-catenin signaling can drive FP expansion along the rostrocaudal axis. Although excess canonical Wnt/beta-catenin signaling generally promotes neurogenesis at midbrain levels, less tyrosine hydroxylase (Th)+, mDA neurons are generated, particularly impacting the Substantia Nigra pars compacta. This is likely because of improper progenitor specification. Excess canonical Wnt/beta-catenin signaling causes downregulation of net Lmx1b, Shh and Foxa2 levels in mDA progenitors. Moreover, these progenitors assume a mixed identity to that of Lmx1a+/Lmx1b+/Nkx6-1+/Neurog1+ progenitors. We also show by lineage tracing analysis that normally, Neurog1+ progenitors predominantly give rise to Pou4f1+ neurons, but not Th+ neurons. Accordingly, in the mutant embryos, Neurog1+ progenitors at the midline generate ectopic Pou4f1+ neurons at the expense of Th+ mDA neurons. Our study suggests that an optimal dose of Wnt/beta-catenin signaling is critical for proper establishment of the mDA progenitor character. Our findings will impact embryonic stem cell protocols that utilize Wnt pathway reagents to derive mDA neuron models and therapeutics for Parkinson's disease.
Collapse
Affiliation(s)
- Navid Nouri
- Northwestern University, Feinberg Medical School, Department of Neurology and Center for Genetic Medicine, 7-113 Lurie Bldg., 303 E Superior Street, Chicago, IL 60611, USA.
| | - Meera J Patel
- Northwestern University, Feinberg Medical School, Department of Neurology and Center for Genetic Medicine, 7-113 Lurie Bldg., 303 E Superior Street, Chicago, IL 60611, USA; Committee on Neurobiology, University of Chicago, 924 E 57th St. R222, Chicago, IL 60637, USA.
| | - Milan Joksimovic
- Northwestern University, Feinberg Medical School, Department of Neurology and Center for Genetic Medicine, 7-113 Lurie Bldg., 303 E Superior Street, Chicago, IL 60611, USA.
| | - Jean-Francois Poulin
- Northwestern University, Feinberg Medical School, Department of Neurology and Center for Genetic Medicine, 7-113 Lurie Bldg., 303 E Superior Street, Chicago, IL 60611, USA.
| | - Angela Anderegg
- Northwestern University, Feinberg Medical School, Department of Neurology and Center for Genetic Medicine, 7-113 Lurie Bldg., 303 E Superior Street, Chicago, IL 60611, USA.
| | - M Mark Taketo
- Graduate School of Medicine, Kyoto University, Yoshida-Konoé-cho, Sakyo, Kyoto 606-8501, Japan.
| | - Yong-Chao Ma
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Children's Hospital of Chicago Research Center, 2430 North Halsted Street, Room C321, Chicago, IL 60614, USA.
| | - Rajeshwar Awatramani
- Northwestern University, Feinberg Medical School, Department of Neurology and Center for Genetic Medicine, 7-113 Lurie Bldg., 303 E Superior Street, Chicago, IL 60611, USA.
| |
Collapse
|
16
|
Blaess S, Ang SL. Genetic control of midbrain dopaminergic neuron development. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:113-34. [PMID: 25565353 DOI: 10.1002/wdev.169] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 10/31/2014] [Accepted: 11/16/2014] [Indexed: 12/31/2022]
Abstract
UNLABELLED Midbrain dopaminergic neurons are involved in regulating motor control, reward behavior, and cognition. Degeneration or dysfunction of midbrain dopaminergic neurons is implicated in several neuropsychiatric disorders such as Parkinson's disease, substance use disorders, depression, and schizophrenia. Understanding the developmental processes that generate midbrain dopaminergic neurons will facilitate the generation of dopaminergic neurons from stem cells for cell replacement therapies to substitute degenerating cells in Parkinson's disease patients and will forward our understanding on how functional diversity of dopaminergic neurons in the adult brain is established. Midbrain dopaminergic neurons develop in a multistep process. Following the induction of the ventral midbrain, a distinct dopaminergic progenitor domain is specified and dopaminergic progenitors undergo proliferation, neurogenesis, and differentiation. Subsequently, midbrain dopaminergic neurons acquire a mature dopaminergic phenotype, migrate to their final position and establish projections and connections to their forebrain targets. This review will discuss insights gained on the signaling network of secreted molecules, cell surface receptors, and transcription factors that regulate specification and differentiation of midbrain dopaminergic progenitors and neurons, from the induction of the ventral midbrain to the migration of dopaminergic neurons. For further resources related to this article, please visit the WIREs website. CONFLICT OF INTEREST The authors have declared no conflicts of interest for this article.
Collapse
Affiliation(s)
- Sandra Blaess
- Institute of Reconstructive Neurobiology, Life and Brain Center, University of Bonn, Bonn, Germany
| | | |
Collapse
|
17
|
Lahti L, Haugas M, Tikker L, Airavaara M, Voutilainen MH, Anttila J, Kumar S, Inkinen C, Salminen M, Partanen J. Differentiation and molecular heterogeneity of inhibitory and excitatory neurons associated with midbrain dopaminergic nuclei. Development 2015; 143:516-29. [DOI: 10.1242/dev.129957] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/18/2015] [Indexed: 12/24/2022]
Abstract
Local inhibitory GABAergic and excitatory glutamatergic neurons are important for midbrain dopaminergic and hindbrain serotonergic pathways controlling motivation, mood, and voluntary movements. Such neurons reside both within the dopaminergic nuclei, and in adjacent brain structures, including the rostromedial and laterodorsal tegmental nuclei. Compared to the monoaminergic neurons, the development, heterogeneity, and molecular characteristics of these regulatory neurons are poorly understood. We show here that different GABAergic and glutamatergic subgroups associated with the monoaminergic nuclei express specific transcription factors. These neurons share common origins in the ventrolateral rhombomere 1, where postmitotic selector genes Tal1, Gata2, and Gata3 control the balance between the generation of inhibitory and excitatory neurons. In the absence of Tal1, or both Gata2 and Gata3, the GABAergic precursors adopt glutamatergic fates and populate the glutamatergic nuclei in excessive numbers. Together, our results uncover developmental regulatory mechanisms, molecular characteristics, and heterogeneity of central regulators of monoaminergic circuits.
Collapse
Affiliation(s)
- Laura Lahti
- Department of Biosciences, P.O. Box 56, Viikinkaari 9, FIN-00014 University of Helsinki, Helsinki, Finland
| | - Maarja Haugas
- Department of Biosciences, P.O. Box 56, Viikinkaari 9, FIN-00014 University of Helsinki, Helsinki, Finland
| | - Laura Tikker
- Department of Biosciences, P.O. Box 56, Viikinkaari 9, FIN-00014 University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Institute of Biotechnology, P.O. Box 56, Viikinkaari 9, FIN-00014 University of Helsinki, Helsinki, Finland
| | - Merja H. Voutilainen
- Institute of Biotechnology, P.O. Box 56, Viikinkaari 9, FIN-00014 University of Helsinki, Helsinki, Finland
| | - Jenni Anttila
- Institute of Biotechnology, P.O. Box 56, Viikinkaari 9, FIN-00014 University of Helsinki, Helsinki, Finland
| | - Suman Kumar
- Department of Biosciences, P.O. Box 56, Viikinkaari 9, FIN-00014 University of Helsinki, Helsinki, Finland
| | - Caisa Inkinen
- Department of Biosciences, P.O. Box 56, Viikinkaari 9, FIN-00014 University of Helsinki, Helsinki, Finland
| | - Marjo Salminen
- Department of Veterinary Biosciences, Agnes Sjöbergin katu 2, FIN-00014 University of Helsinki, Helsinki, Finland
| | - Juha Partanen
- Department of Biosciences, P.O. Box 56, Viikinkaari 9, FIN-00014 University of Helsinki, Helsinki, Finland
| |
Collapse
|
18
|
Yang F, Liu Y, Tu J, Wan J, Zhang J, Wu B, Chen S, Zhou J, Mu Y, Wang L. Activated astrocytes enhance the dopaminergic differentiation of stem cells and promote brain repair through bFGF. Nat Commun 2014; 5:5627. [PMID: 25517983 PMCID: PMC4284631 DOI: 10.1038/ncomms6627] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/21/2014] [Indexed: 02/06/2023] Open
Abstract
Astrocytes provide neuroprotective effects against degeneration of dopaminergic (DA) neurons and play a fundamental role in DA differentiation of neural stem cells. Here we show that light illumination of astrocytes expressing engineered channelrhodopsin variant (ChETA) can remarkably enhance the release of basic fibroblast growth factor (bFGF) and significantly promote the DA differentiation of human embryonic stem cells (hESCs) in vitro. Light activation of transplanted astrocytes in the substantia nigra (SN) also upregulates bFGF levels in vivo and promotes the regenerative effects of co-transplanted stem cells. Importantly, upregulation of bFGF levels, by specific light activation of endogenous astrocytes in the SN, enhances the DA differentiation of transplanted stem cells and promotes brain repair in a mouse model of Parkinson's disease (PD). Our study indicates that astrocyte-derived bFGF is required for regulation of DA differentiation of the stem cells and may provide a strategy targeting astrocytes for treatment of PD.
Collapse
Affiliation(s)
- Fan Yang
- Shenzhen Key Lab of Neuropsychiatric Modulation, CAS Center for Excellence in Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yunhui Liu
- Shenzhen Key Lab of Neuropsychiatric Modulation, CAS Center for Excellence in Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jie Tu
- Shenzhen Key Lab of Neuropsychiatric Modulation, CAS Center for Excellence in Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jun Wan
- Shenzhen Key Lab of Neuropsychiatric Modulation, CAS Center for Excellence in Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jie Zhang
- Shenzhen Key Lab of Neuropsychiatric Modulation, CAS Center for Excellence in Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Bifeng Wu
- Shenzhen Key Lab of Neuropsychiatric Modulation, CAS Center for Excellence in Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Shanping Chen
- Shenzhen Key Lab of Neuropsychiatric Modulation, CAS Center for Excellence in Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jiawei Zhou
- State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yangling Mu
- Shenzhen Key Lab of Neuropsychiatric Modulation, CAS Center for Excellence in Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liping Wang
- Shenzhen Key Lab of Neuropsychiatric Modulation, CAS Center for Excellence in Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
19
|
Veenvliet JV, Smidt MP. Molecular mechanisms of dopaminergic subset specification: fundamental aspects and clinical perspectives. Cell Mol Life Sci 2014; 71:4703-27. [PMID: 25064061 PMCID: PMC11113784 DOI: 10.1007/s00018-014-1681-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 07/04/2014] [Accepted: 07/10/2014] [Indexed: 12/22/2022]
Abstract
Dopaminergic (DA) neurons in the ventral mesodiencephalon control locomotion and emotion and are affected in psychiatric and neurodegenerative diseases, such as Parkinson's disease (PD). A clinical hallmark of PD is the specific degeneration of DA neurons located within the substantia nigra (SNc), whereas neurons in the ventral tegmental area remain unaffected. Recent advances have highlighted that the selective vulnerability of the SNc may originate in subset-specific molecular programming during DA neuron development, and significantly increased our understanding of the molecular code that drives specific SNc development. We here present an up-to-date overview of molecular mechanisms that direct DA subset specification, integrating our current knowledge about subset-specific roles of transcription factors, signaling pathways and morphogenes. We discuss strategies to further unravel subset-specific gene-regulatory networks, and the clinical promise of fundamental knowledge about subset specification of DA neurons, with regards to cell replacement therapy and cell-type-specific vulnerability in PD.
Collapse
Affiliation(s)
- Jesse V. Veenvliet
- Department of Molecular Neuroscience, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Marten P. Smidt
- Department of Molecular Neuroscience, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| |
Collapse
|
20
|
Allodi I, Hedlund E. Directed midbrain and spinal cord neurogenesis from pluripotent stem cells to model development and disease in a dish. Front Neurosci 2014; 8:109. [PMID: 24904255 PMCID: PMC4033221 DOI: 10.3389/fnins.2014.00109] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 04/28/2014] [Indexed: 12/29/2022] Open
Abstract
Induction of specific neuronal fates is restricted in time and space in the developing CNS through integration of extrinsic morphogen signals and intrinsic determinants. Morphogens impose regional characteristics on neural progenitors and establish distinct progenitor domains. Such domains are defined by unique expression patterns of fate determining transcription factors. These processes of neuronal fate specification can be recapitulated in vitro using pluripotent stem cells. In this review, we focus on the generation of dopamine neurons and motor neurons, which are induced at ventral positions of the neural tube through Sonic hedgehog (Shh) signaling, and defined at anteroposterior positions by fibroblast growth factor (Fgf) 8, Wnt1, and retinoic acid (RA). In vitro utilization of these morphogenic signals typically results in the generation of multiple neuronal cell types, which are defined at the intersection of these signals. If the purpose of in vitro neurogenesis is to generate one cell type only, further lineage restriction can be accomplished by forced expression of specific transcription factors in a permissive environment. Alternatively, cell-sorting strategies allow for selection of neuronal progenitors or mature neurons. However, modeling development, disease and prospective therapies in a dish could benefit from structured heterogeneity, where desired neurons are appropriately synaptically connected and thus better reflect the three-dimensional structure of that region. By modulating the extrinsic environment to direct sequential generation of neural progenitors within a domain, followed by self-organization and synaptic establishment, a reductionist model of that brain region could be created. Here we review recent advances in neuronal fate induction in vitro, with a focus on the interplay between cell intrinsic and extrinsic factors, and discuss the implications for studying development and disease in a dish.
Collapse
Affiliation(s)
- Ilary Allodi
- Department of Neuroscience, Karolinska Institutet Stockholm, Sweden
| | - Eva Hedlund
- Department of Neuroscience, Karolinska Institutet Stockholm, Sweden
| |
Collapse
|
21
|
Recurrent somatic mutations in ACVR1 in pediatric midline high-grade astrocytoma. Nat Genet 2014; 46:462-6. [PMID: 24705250 DOI: 10.1038/ng.2950] [Citation(s) in RCA: 341] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 03/14/2014] [Indexed: 12/24/2022]
Abstract
Pediatric midline high-grade astrocytomas (mHGAs) are incurable with few treatment targets identified. Most tumors harbor mutations encoding p.Lys27Met in histone H3 variants. In 40 treatment-naive mHGAs, 39 analyzed by whole-exome sequencing, we find additional somatic mutations specific to tumor location. Gain-of-function mutations in ACVR1 occur in tumors of the pons in conjunction with histone H3.1 p.Lys27Met substitution, whereas FGFR1 mutations or fusions occur in thalamic tumors associated with histone H3.3 p.Lys27Met substitution. Hyperactivation of the bone morphogenetic protein (BMP)-ACVR1 developmental pathway in mHGAs harboring ACVR1 mutations led to increased levels of phosphorylated SMAD1, SMAD5 and SMAD8 and upregulation of BMP downstream early-response genes in tumor cells. Global DNA methylation profiles were significantly associated with the p.Lys27Met alteration, regardless of the mutant histone H3 variant and irrespective of tumor location, supporting the role of this substitution in driving the epigenetic phenotype. This work considerably expands the number of potential treatment targets and further justifies pretreatment biopsy in pediatric mHGA as a means to orient therapeutic efforts in this disease.
Collapse
|
22
|
Saritas-Yildirim B, Silva EM. The role of targeted protein degradation in early neural development. Genesis 2014; 52:287-99. [PMID: 24623518 DOI: 10.1002/dvg.22771] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/05/2014] [Accepted: 03/07/2014] [Indexed: 11/08/2022]
Abstract
As neural stem cells differentiate into neurons during neurogenesis, the proteome of the cells is restructured by de novo expression and selective removal of regulatory proteins. The control of neurogenesis at the level of gene regulation is well documented and the regulation of protein abundance through protein degradation via the Ubiquitin/26S proteasome pathway is a rapidly developing field. This review describes our current understanding of the role of the proteasome pathway in neurogenesis. Collectively, the studies show that targeted protein degradation is an important regulatory mechanism in the generation of new neurons.
Collapse
|
23
|
Mäkelä J, Tselykh TV, Maiorana F, Eriksson O, Do HT, Mudò G, Korhonen LT, Belluardo N, Lindholm D. Fibroblast growth factor-21 enhances mitochondrial functions and increases the activity of PGC-1α in human dopaminergic neurons via Sirtuin-1. SPRINGERPLUS 2014; 3:2. [PMID: 25932355 PMCID: PMC4409609 DOI: 10.1186/2193-1801-3-2] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 12/30/2013] [Indexed: 12/18/2022]
Abstract
Mitochondrial dysfunctions accompany several neurodegenerative disorders and contribute to disease pathogenesis among others in Parkinson’s disease (PD). Peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) is a major regulator of mitochondrial functions and biogenesis, and was suggested as a therapeutic target in PD. PGC-1α is regulated by both transcriptional and posttranslational events involving also the action of growth factors. Fibroblast growth factor-21 (FGF21) is a regulator of glucose and fatty acid metabolism in the body but little is known about its action in the brain. We show here that FGF21 increased the levels and activity of PGC-1α and elevated mitochondrial antioxidants in human dopaminergic cells in culture. The activation of PGC-1α by FGF21 occurred via the NAD+-dependent deacetylase Sirtuin-1 (SIRT1) subsequent to an increase in the enzyme, nicotinamide phosphoribosyltransferase (Nampt). FGF21 also enhanced mitochondrial respiratory capacity in human dopaminergic neurons as shown in real-time analyses of living cells. FGF21 is present in the brain including midbrain and is expressed by glial cells in culture. These results show that FGF21 activates PGC-1α and increases mitochondrial efficacy in human dopaminergic neurons suggesting that FGF21 could potentially play a role in dopaminergic neuron viability and in PD.
Collapse
Affiliation(s)
- Johanna Mäkelä
- Institute of Biomedicine/Biochemistry and Developmental Biology, University of Helsinki, Haartmaninkatu 8, FIN-00290 Helsinki, Finland ; Minerva Foundation Institute for Medical Research, Biomedicum-2, FIN-00290 Helsinki, Finland
| | - Timofey V Tselykh
- Institute of Biomedicine/Biochemistry and Developmental Biology, University of Helsinki, Haartmaninkatu 8, FIN-00290 Helsinki, Finland ; Minerva Foundation Institute for Medical Research, Biomedicum-2, FIN-00290 Helsinki, Finland
| | - Francesca Maiorana
- Institute of Biomedicine/Biochemistry and Developmental Biology, University of Helsinki, Haartmaninkatu 8, FIN-00290 Helsinki, Finland ; Department of Experimental Biomedicine and Clinical Neuroscience, Division of Human Physiology, University of Palermo, Corso Tukory 129, I-90134 Palermo, Italy
| | - Ove Eriksson
- Institute of Biomedicine/Biochemistry and Developmental Biology, University of Helsinki, Haartmaninkatu 8, FIN-00290 Helsinki, Finland
| | - Hai Thi Do
- Institute of Biomedicine/Biochemistry and Developmental Biology, University of Helsinki, Haartmaninkatu 8, FIN-00290 Helsinki, Finland ; Minerva Foundation Institute for Medical Research, Biomedicum-2, FIN-00290 Helsinki, Finland
| | - Giuseppa Mudò
- Department of Experimental Biomedicine and Clinical Neuroscience, Division of Human Physiology, University of Palermo, Corso Tukory 129, I-90134 Palermo, Italy
| | - Laura T Korhonen
- Institute of Biomedicine/Biochemistry and Developmental Biology, University of Helsinki, Haartmaninkatu 8, FIN-00290 Helsinki, Finland ; Minerva Foundation Institute for Medical Research, Biomedicum-2, FIN-00290 Helsinki, Finland
| | - Natale Belluardo
- Department of Experimental Biomedicine and Clinical Neuroscience, Division of Human Physiology, University of Palermo, Corso Tukory 129, I-90134 Palermo, Italy
| | - Dan Lindholm
- Institute of Biomedicine/Biochemistry and Developmental Biology, University of Helsinki, Haartmaninkatu 8, FIN-00290 Helsinki, Finland ; Minerva Foundation Institute for Medical Research, Biomedicum-2, FIN-00290 Helsinki, Finland
| |
Collapse
|
24
|
Smits SM, von Oerthel L, Hoekstra EJ, Burbach JPH, Smidt MP. Molecular marker differences relate to developmental position and subsets of mesodiencephalic dopaminergic neurons. PLoS One 2013; 8:e76037. [PMID: 24116087 PMCID: PMC3792114 DOI: 10.1371/journal.pone.0076037] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 08/23/2013] [Indexed: 12/02/2022] Open
Abstract
The development of mesodiencephalic dopaminergic (mdDA) neurons located in the substantia nigra compacta (SNc) and ventral tegmental area (VTA) follow a number of stages marked by distinct events. After preparation of the region by signals that provide induction and patterning, several transcription factors have been identified, which are involved in specifying the neuronal fate of these cells. The specific vulnerability of SNc neurons is thought to root in these specific developmental programs. The present study examines the positions of young postmitotic mdDA neurons to relate developmental position to mdDA subset specific markers. MdDA neurons were mapped relative to the neuromeric domains (prosomeres 1-3 (P1-3), midbrain, and hindbrain) as well as the longitudinal subdivisions (floor plate, basal plate, alar plate), as proposed by the prosomeric model. We found that postmitotic mdDA neurons are located mainly in the floorplate domain and very few in slightly more lateral domains. Moreover, mdDA neurons are present along a large proportion of the anterior/posterior axis extending from the midbrain to P3 in the diencephalon. The specific positions relate to some extent to the presence of specific subset markers as Ahd2. In the adult stage more of such subsets specific expressed genes are present and may represent a molecular map defining molecularly distinct groups of mdDA neurons.
Collapse
Affiliation(s)
- Simone M. Smits
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lars von Oerthel
- Molecular Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Elisa J. Hoekstra
- Molecular Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - J. Peter H Burbach
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marten P. Smidt
- Molecular Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
25
|
Tang M, Luo SX, Tang V, Huang EJ. Temporal and spatial requirements of Smoothened in ventral midbrain neuronal development. Neural Dev 2013; 8:8. [PMID: 23618354 PMCID: PMC3680293 DOI: 10.1186/1749-8104-8-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 04/05/2013] [Indexed: 11/11/2022] Open
Abstract
Background Several studies have indicated that Sonic hedgehog (Shh) regulates the expansion of dopaminergic (DA) progenitors and the subsequent generation of mature DA neurons. This prevailing view has been based primarily on in vitro culture results, and the exact in vivo function of Shh signaling in the patterning and neurogenesis of the ventral midbrain (vMB) remains unclear. Methods We characterized the transcriptional codes for the vMB progenitor domains, and correlated them with the expression patterns of Shh signaling effectors, including Shh, Smoothened, Patched, Gli1, Gli2 and Gli3. Results While Shh and its downstream effectors showed robust expression in the neurogenic niche for DA progenitors at embryonic day (E)8 to E8.5, their expression shifted to the lateral domains from E9.5 to E12.5. Consistent with this dynamic change, conditional mutants with region-specific removal of the Shh receptor Smoothened in the vMB progenitors (Shh-Cre;Smofl/fl) showed a transient reduction in DA progenitors and DA neurons at E10.5, but had more profound defects in neurons derived from the more lateral domains, including those in the red nucleus, oculomotor nucleus, and raphe nuclei. Conversely, constitutive activation of Smoothened signaling in vMB (Shh-Cre;SmoM2) showed transient expansion of the same progenitor population. To further characterize the nature of Shh-Smoothened signaling in vMB, we examined the BAT-GAL reporter and the expression of Wnt1 in vMB, and found that the antagonistic effects of Shh and Wnt signaling critically regulate the development of DA progenitors and DA neurons. Conclusion These results highlight previously unrecognized effects of Shh-Smoothened signaling in the region-specific neurogenesis within the vMB.
Collapse
Affiliation(s)
- Mianzhi Tang
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | | | | | | |
Collapse
|
26
|
Midbrain dopaminergic neurons: a review of the molecular circuitry that regulates their development. Dev Biol 2013; 379:123-38. [PMID: 23603197 DOI: 10.1016/j.ydbio.2013.04.014] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 03/27/2013] [Accepted: 04/12/2013] [Indexed: 12/21/2022]
Abstract
Dopaminergic (DA) neurons of the ventral midbrain (VM) play vital roles in the regulation of voluntary movement, emotion and reward. They are divided into the A8, A9 and A10 subgroups. The development of the A9 group of DA neurons is an area of intense investigation to aid the generation of these neurons from stem cell sources for cell transplantation approaches to Parkinson's disease (PD). This review discusses the molecular processes that are involved in the identity, specification, maturation, target innervation and survival of VM DA neurons during development. The complex molecular interactions of a number of genetic pathways are outlined, as well as recent advances in the mechanisms that regulate subset identity within the VM DA neuronal pool. A thorough understanding of the cellular and molecular mechanisms involved in the development of VM DA neurons will greatly facilitate the use of cell replacement therapy for the treatment of PD.
Collapse
|
27
|
Lahti L, Achim K, Partanen J. Molecular regulation of GABAergic neuron differentiation and diversity in the developing midbrain. Acta Physiol (Oxf) 2013; 207:616-27. [PMID: 23297792 DOI: 10.1111/apha.12062] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/10/2012] [Accepted: 12/26/2012] [Indexed: 12/23/2022]
Abstract
The midbrain GABAergic neurones control several aspects of behaviour, play important roles in psychiatric disease and are targets of medical treatments as well as drugs of abuse. However, their molecular diversity and regulation of development are only beginning to be understood. In this review, we briefly introduce distinct subpopulations of the midbrain GABAergic neurones and discuss knowledge on their development, including the developmental origins of midbrain GABAergic neurones as well as transcriptional regulatory mechanisms guiding their differentiation and identity. Important GABAergic neuron subpopulations are found within the dopaminergic (DA) nuclei in the ventral midbrain. GABAergic substantia nigra pars reticulata is the main output pathway of the basal ganglia system regulating voluntary movements. Recent studies have also highlighted importance of the GABAergic neurones associated with the ventral tegmental area for the control of DA neuron activity and motivated behaviours. Interestingly, the development of the GABAergic neurones associated with the DA nuclei is very different from the rest of the midbrain. Knowledge on developmental regulation can lead to insights into the molecular, structural and functional diversity of the midbrain GABAergic neurones and their subpopulations, cell groups of great physiological and medical interest.
Collapse
Affiliation(s)
- L. Lahti
- Department of Biosciences; Viikki Biocenter; University of Helsinki; Helsinki; Finland
| | - K. Achim
- European Molecular Biology Laboratory; Heidelberg; Germany
| | - J. Partanen
- Department of Biosciences; Viikki Biocenter; University of Helsinki; Helsinki; Finland
| |
Collapse
|
28
|
Xi J, Liu Y, Liu H, Chen H, Emborg ME, Zhang SC. Specification of midbrain dopamine neurons from primate pluripotent stem cells. Stem Cells 2013; 30:1655-63. [PMID: 22696177 DOI: 10.1002/stem.1152] [Citation(s) in RCA: 166] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
By sequentially applying sonic hedgehog (C25II) and CHIR99021 (GSK3β inhibitor) to induce the midbrain floor plate (FP) progenitors and fibroblast growth factor 8 (FGF8) to promote dopaminergic differentiation in a chemically defined medium, we have established a robust system for the generation of midbrain dopamine (DA) neurons from human and rhesus monkey embryonic stem cells and induced pluripotent stem cells (PSCs). We found that CHIR99021 specifies diencephalon to hind brain fates in a concentration-dependent manner and only a narrow concentration range of CHIR99021 at a particular window is necessary to induce the midbrain FP progenitors, expressing Corin, En1, FoxA2, and Lmx1a. FGF8 enhances the dopaminergic fate of the progenitors, thus generating DA neurons with midbrain characteristics, including expression of tyrosine hydroxylase, Lmx1a/b, FoxA2, FoxP1, Nurr1, and En1 as well as typical electrophysiological properties. More than half of these DA neurons expressed A9 DA neuron markers Girk2 and ALDH1a1. The new strategy will allow generation of enriched populations of functional midbrain DA neurons from both human and monkey PSCs for disease modeling, drug testing, and potential cell therapy.
Collapse
Affiliation(s)
- Jiajie Xi
- Institute of Stem Cell and Regenerative Medicine, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
29
|
Achim K, Peltopuro P, Lahti L, Li J, Salminen M, Partanen J. Distinct developmental origins and regulatory mechanisms for GABAergic neurons associated with dopaminergic nuclei in the ventral mesodiencephalic region. Development 2012; 139:2360-70. [PMID: 22627282 DOI: 10.1242/dev.076380] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
GABAergic neurons in the ventral mesodiencephalic region are highly important for the function of dopaminergic pathways that regulate multiple aspects of behavior. However, development of these neurons is poorly understood. We recently showed that molecular regulation of differentiation of the GABAergic neurons associated with the dopaminergic nuclei in the ventral midbrain (VTA and SNpr) is distinct from the rest of midbrain, but the reason for this difference remained elusive. Here, we have analyzed the developmental origin of the VTA and SNpr GABAergic neurons by genetic fate mapping. We demonstrate that the majority of these GABAergic neurons originate outside the midbrain, from rhombomere 1, and move into the ventral midbrain only as postmitotic neuronal precursors. We further show that Gata2, Gata3 and Tal1 define a subpopulation of GABAergic precursors in ventral rhombomere 1. A failure in GABAergic neuron differentiation in this region correlates with loss of VTA and SNpr GABAergic neurons in Tal1 mutant mice. In contrast to midbrain, GABAergic neurons of the anterior SNpr in the diencephalon are not derived from the rhombomere 1. These results suggest unique migratory pathways for the precursors of important GABAergic neuron subpopulations, and provide the basis for understanding diversity within midbrain GABAergic neurons.
Collapse
Affiliation(s)
- Kaia Achim
- Department of Biosciences, PO Box 56, Viikinkaari 5, FIN00014-University of Helsinki, Helsinki, Finland
| | | | | | | | | | | |
Collapse
|