1
|
Doll HM, Risgaard RD, Thurston H, Chen RJ, Sousa AM. Evolutionary innovations in the primate dopaminergic system. Curr Opin Genet Dev 2024; 88:102236. [PMID: 39153332 PMCID: PMC11384322 DOI: 10.1016/j.gde.2024.102236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/12/2024] [Indexed: 08/19/2024]
Abstract
The human brain has evolved unique capabilities compared to other vertebrates. The mechanistic basis of these derived traits remains a fundamental question in biology due to its relevance to the origin of our cognitive abilities and behavioral repertoire, as well as to human-specific aspects of neuropsychiatric and neurodegenerative diseases. Comparisons of the human brain to those of nonhuman primates and other mammals have revealed that differences in the neuromodulatory systems, especially in the dopaminergic system, may govern some of these behavioral and cognitive alterations, including increased vulnerability to certain brain disorders. In this review, we highlight and discuss recent findings of human- and primate-specific alterations of the dopaminergic system, focusing on differences in anatomy, circuitry, and molecular properties.
Collapse
Affiliation(s)
- Hannah M Doll
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan D Risgaard
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Hailey Thurston
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Rachel J Chen
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - André Mm Sousa
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
2
|
Zhang X, Zhang Y, Wang B, Xie C, Wang J, Fang R, Dong H, Fan G, Wang M, He Y, Shen C, Duan Y, Zhao J, Liu Z, Li Q, Ma Y, Yu M, Wang J, Fei J, Xiao L, Huang F. Pyroptosis-mediator GSDMD promotes Parkinson's disease pathology via microglial activation and dopaminergic neuronal death. Brain Behav Immun 2024; 119:129-145. [PMID: 38552923 DOI: 10.1016/j.bbi.2024.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/02/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024] Open
Abstract
GSDMD-mediated pyroptosis occurs in the nigrostriatal pathway in Parkinson's disease animals, yet the role of GSDMD in neuroinflammation and death of dopaminergic neurons in Parkinson's disease remains elusive. Here, our in vivo and in vitro studies demonstrated that GSDMD, as a pyroptosis executor, contributed to glial reaction and death of dopaminergic neurons across different Parkinson's disease models. The ablation of the Gsdmd attenuated Parkinson's disease damage by reducing dopaminergic neuronal death, microglial activation, and detrimental transformation. Disulfiram, an inhibitor blocking GSDMD pore formation, efficiently curtailed pyroptosis, thereby lessening the pathology of Parkinson's disease. Additionally, a modification in GSDMD was identified in the blood of Parkinson's disease patients in contrast to healthy subjects. Therefore, the detected alteration in GSDMD within the blood of Parkinson's disease patients and the protective impact of disulfiram could be promising for the diagnostic and therapeutic approaches against Parkinson's disease.
Collapse
Affiliation(s)
- Xiaoshuang Zhang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yunhe Zhang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Boya Wang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Chuantong Xie
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jinghui Wang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Rong Fang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Hongtian Dong
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Guangchun Fan
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Mengze Wang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yongtao He
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Chenye Shen
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yufei Duan
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jiayin Zhao
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Zhaolin Liu
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Qing Li
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yuanyuan Ma
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Mei Yu
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jian Wang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jian Fei
- School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Engineering Research Center for Model Organisms, Shanghai Model Organisms Center, INC., Pudong, Shanghai 201203, China.
| | - Lei Xiao
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.
| | - Fang Huang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.
| |
Collapse
|
3
|
Seiler JL, Zhuang X, Nelson AB, Lerner TN. Dopamine across timescales and cell types: Relevance for phenotypes in Parkinson's disease progression. Exp Neurol 2024; 374:114693. [PMID: 38242300 DOI: 10.1016/j.expneurol.2024.114693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
Dopamine neurons in the substantia nigra pars compacta (SNc) synthesize and release dopamine, a critical neurotransmitter for movement and learning. SNc dopamine neurons degenerate in Parkinson's Disease (PD), causing a host of motor and non-motor symptoms. Here, we review recent conceptual advances in our basic understanding of the dopamine system - including our rapidly advancing knowledge of dopamine neuron heterogeneity - with special attention to their importance for understanding PD. In PD patients, dopamine neuron degeneration progresses from lateral SNc to medial SNc, suggesting clinically relevant heterogeneity in dopamine neurons. With technical advances in dopamine system interrogation, we can understand the relevance of this heterogeneity for PD progression and harness it to develop new treatments.
Collapse
Affiliation(s)
- Jillian L Seiler
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Xiaowen Zhuang
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Alexandra B Nelson
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Talia N Lerner
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern University Interdepartmental Neuroscience Program (NUIN), Evanston, IL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
4
|
Rocha GS, Freire MAM, Paiva KM, Oliveira RF, Morais PLAG, Santos JR, Cavalcanti JRLP. The neurobiological effects of senescence on dopaminergic system: A comprehensive review. J Chem Neuroanat 2024; 137:102415. [PMID: 38521203 DOI: 10.1016/j.jchemneu.2024.102415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/26/2024] [Accepted: 03/15/2024] [Indexed: 03/25/2024]
Abstract
Over time, the body undergoes a natural, multifactorial, and ongoing process named senescence, which induces changes at the molecular, cellular, and micro-anatomical levels in many body systems. The brain, being a highly complex organ, is particularly affected by this process, potentially impairing its numerous functions. The brain relies on chemical messengers known as neurotransmitters to function properly, with dopamine being one of the most crucial. This catecholamine is responsible for a broad range of critical roles in the central nervous system, including movement, learning, cognition, motivation, emotion, reward, hormonal release, memory consolidation, visual performance, sexual drive, modulation of circadian rhythms, and brain development. In the present review, we thoroughly examine the impact of senescence on the dopaminergic system, with a primary focus on the classic delimitations of the dopaminergic nuclei from A8 to A17. We provide in-depth information about their anatomy and function, particularly addressing how senescence affects each of these nuclei.
Collapse
Affiliation(s)
- Gabriel S Rocha
- Behavioral and Evolutionary Neurobiology Laboratory, Federal University of Sergipe (UFS), Itabaiana, Brazil
| | - Marco Aurelio M Freire
- Behavioral and Evolutionary Neurobiology Laboratory, Federal University of Sergipe (UFS), Itabaiana, Brazil
| | - Karina M Paiva
- Laboratory of Experimental Neurology, State University of Rio Grande do Norte (UERN), Mossoró, Brazil
| | - Rodrigo F Oliveira
- Laboratory of Experimental Neurology, State University of Rio Grande do Norte (UERN), Mossoró, Brazil
| | - Paulo Leonardo A G Morais
- Laboratory of Experimental Neurology, State University of Rio Grande do Norte (UERN), Mossoró, Brazil
| | - José Ronaldo Santos
- Behavioral and Evolutionary Neurobiology Laboratory, Federal University of Sergipe (UFS), Itabaiana, Brazil
| | | |
Collapse
|
5
|
Kelly EA, Love TM, Fudge JL. Corticotropin-releasing factor-dopamine interactions in male and female macaque: Beyond the classic VTA. Synapse 2024; 78:e22284. [PMID: 37996987 PMCID: PMC10842953 DOI: 10.1002/syn.22284] [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: 07/14/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 11/25/2023]
Abstract
Dopamine (DA) is involved in stress and stress-related illnesses, including many psychiatric disorders. Corticotropin-releasing factor (CRF) plays a role in stress responses and targets the ventral midbrain DA system, which is composed of DA and non-DA cells, and divided into specific subregions. Although CRF inputs to the midline A10 nuclei ("classic VTA") are known, in monkeys, CRF-containing terminals are also highly enriched in the expanded A10 parabrachial pigmented nucleus (PBP) and in the A8 retrorubral field subregions. We characterized CRF-labeled synaptic terminals on DA (tyrosine hydroxylase, TH+) and non-DA (TH-) cell types in the PBP and A8 regions using immunoreactive electron microscopy (EM) in male and female macaques. CRF labeling was present mostly in axon terminals, which mainly contacted TH-negative dendrites in both subregions. Most CRF-positive terminals had symmetric profiles. In both PBP and A8, CRF symmetric (putative inhibitory) synapses onto TH-negative dendrites were significantly greater than asymmetric (putative excitatory) profiles. This overall pattern was similar in males and females, despite shifts in the size of these effects between regions depending on sex. Because stress and gonadal hormone shifts can influence CRF expression, we also did hormonal assays over a 6-month time period and found little variability in basal cortisol across similarly housed animals at the same age. Together our findings suggest that at baseline, CRF-positive synaptic terminals in the primate PBP and A8 are poised to regulate DA indirectly through synaptic contacts onto non-DA neurons.
Collapse
Affiliation(s)
- E A Kelly
- Departments of Neuroscience, Del Monte Institute of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - T M Love
- Department of Biostatistics, Del Monte Institute of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - J L Fudge
- Departments of Neuroscience, Del Monte Institute of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Psychiatry, Del Monte Institute of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
6
|
Krasowska D, Małek A, Kurzepa J, Kapka-Skrzypczak L, Krasowska D, Kurzepa J. Melanin-The Éminence Grise of Melanoma and Parkinson's Disease Development. Cancers (Basel) 2023; 15:5541. [PMID: 38067245 PMCID: PMC10705212 DOI: 10.3390/cancers15235541] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2025] Open
Abstract
A common feature of Parkinson's disease (PD) and melanoma is their starting points being based on cells capable of converting tyrosine into melanin. Melanocytes produce two types of melanin: eumelanin and pheomelanin. These dyes are designed to protect epidermal cells from the harmful effects of UV radiation. Neurones of the substantia nigra, which degenerate during PD, produce neuromelanin, the physiological role of which is not fully explained. This article discusses the potential role of melanins in the pathogenesis of both diseases. Melanins, due to their ability to accumulate toxic substances, may become their sources over time. The use of glutathione for the synthesis of pheomelanins and neuromelanins may reduce the antioxidant capacity of cells, leading to an excessive synthesis of free radicals. This study also tested the hypothesis that certain drugs used in the treatment of PD (L-DOPA, MAO-B and COMT inhibitors, and amantadine), aimed at increasing dopamine concentration, could potentially contribute to the development of melanoma. The role and properties of melanins should continue to be researched. Whether excessive melanin synthesis or its accumulation in the extracellular space may be factors initiating the development of diseases remains an open question.
Collapse
Affiliation(s)
- Danuta Krasowska
- Department of Medical Chemistry, Medical University of Lublin, 20-059 Lublin, Poland; (A.M.); (J.K.)
| | - Agata Małek
- Department of Medical Chemistry, Medical University of Lublin, 20-059 Lublin, Poland; (A.M.); (J.K.)
| | - Joanna Kurzepa
- 1st Department of Medical Radiology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Lucyna Kapka-Skrzypczak
- Department of Molecular Biology and Translational Research, Institute of Rural Health, 20-090 Lublin, Poland;
- World Institute for Family Health, Calisia University, 62-800 Kalisz, Poland
| | - Dorota Krasowska
- Department of Dermatology, Venereology and Pediatric Dermatology, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Jacek Kurzepa
- Department of Medical Chemistry, Medical University of Lublin, 20-059 Lublin, Poland; (A.M.); (J.K.)
| |
Collapse
|
7
|
Marshall P. Finding an Optimal Level of GDNF Overexpression: Insights from Dopamine Cycling. Cell Mol Neurobiol 2023; 43:3179-3189. [PMID: 37410316 PMCID: PMC10477250 DOI: 10.1007/s10571-023-01375-z] [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: 03/14/2023] [Accepted: 06/12/2023] [Indexed: 07/07/2023]
Abstract
The application of glial cell line-derive neurotrophic factor (GDNF) to cell cultures and animal models has demonstrated positive effects upon dopaminergic neuronal survival and development, function, restoration, and protection. On this basis, recombinant GDNF protein has been trialled in the treatment of late-stage human Parkinson's disease patients with only limited success that is likely due to a lack of viable receptor targets in an advanced state of neurodegeneration. The latest research points to more refined approaches of modulating GDNF signalling and an optimal quantity and spatial regulation of GDNF can be extrapolated using regulation of dopamine as a proxy measure. The basic research literature on dopaminergic effects of GDNF in animal models is reviewed, concluding that a twofold increase in natively expressing cells increases dopamine turnover and maximises neuroprotective and beneficial motor effects whilst minimising hyperdopaminergia and other side-effects. Methodological considerations for measurement of dopamine levels and neuroanatomical distinctions are made between populations of dopamine neurons and their respective effects upon movement and behaviour that will inform future research into this still-relevant growth factor.
Collapse
Affiliation(s)
- Pepin Marshall
- Neuroscience Center, University of Helsinki, 00014, Helsinki, Finland.
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
8
|
Azcorra M, Gaertner Z, Davidson C, He Q, Kim H, Nagappan S, Hayes CK, Ramakrishnan C, Fenno L, Kim YS, Deisseroth K, Longnecker R, Awatramani R, Dombeck DA. Unique functional responses differentially map onto genetic subtypes of dopamine neurons. Nat Neurosci 2023; 26:1762-1774. [PMID: 37537242 PMCID: PMC10545540 DOI: 10.1038/s41593-023-01401-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 07/05/2023] [Indexed: 08/05/2023]
Abstract
Dopamine neurons are characterized by their response to unexpected rewards, but they also fire during movement and aversive stimuli. Dopamine neuron diversity has been observed based on molecular expression profiles; however, whether different functions map onto such genetic subtypes remains unclear. In this study, we established that three genetic dopamine neuron subtypes within the substantia nigra pars compacta, characterized by the expression of Slc17a6 (Vglut2), Calb1 and Anxa1, each have a unique set of responses to rewards, aversive stimuli and accelerations and decelerations, and these signaling patterns are highly correlated between somas and axons within subtypes. Remarkably, reward responses were almost entirely absent in the Anxa1+ subtype, which instead displayed acceleration-correlated signaling. Our findings establish a connection between functional and genetic dopamine neuron subtypes and demonstrate that molecular expression patterns can serve as a common framework to dissect dopaminergic functions.
Collapse
Affiliation(s)
- Maite Azcorra
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Department of Neurology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Zachary Gaertner
- Department of Neurology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Connor Davidson
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Qianzi He
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Hailey Kim
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Shivathmihai Nagappan
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Cooper K Hayes
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Lief Fenno
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
- Departments of Neuroscience & Psychiatry, The University of Texas at Austin, Austin, TX, USA
| | - Yoon Seok Kim
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Richard Longnecker
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL, USA
| | - Rajeshwar Awatramani
- Department of Neurology, Northwestern University, Chicago, IL, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Daniel A Dombeck
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
9
|
Otero MG, Bell S, Laperle AH, Lawless G, Myers Z, Castro MA, Villalba JM, Svendsen CN. Organ-Chips Enhance the Maturation of Human iPSC-Derived Dopamine Neurons. Int J Mol Sci 2023; 24:14227. [PMID: 37762529 PMCID: PMC10531789 DOI: 10.3390/ijms241814227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
While cells in the human body function in an environment where the blood supply constantly delivers nutrients and removes waste, cells in conventional tissue culture well platforms are grown with a static pool of media above them and often lack maturity, limiting their utility to study cell biology in health and disease. In contrast, organ-chip microfluidic systems allow the growth of cells under constant flow, more akin to the in vivo situation. Here, we differentiated human induced pluripotent stem cells into dopamine neurons and assessed cellular properties in conventional multi-well cultures and organ-chips. We show that organ-chip cultures, compared to multi-well cultures, provide an overall greater proportion and homogeneity of dopaminergic neurons as well as increased levels of maturation markers. These organ-chips are an ideal platform to study mature dopamine neurons to better understand their biology in health and ultimately in neurological disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Clive N. Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (M.G.O.)
| |
Collapse
|
10
|
Lin CP, Knoop LEJ, Frigerio I, Bol JGJM, Rozemuller AJM, Berendse HW, Pouwels PJW, van de Berg WDJ, Jonkman LE. Nigral Pathology Contributes to Microstructural Integrity of Striatal and Frontal Tracts in Parkinson's Disease. Mov Disord 2023; 38:1655-1667. [PMID: 37347552 DOI: 10.1002/mds.29510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Motor and cognitive impairment in Parkinson's disease (PD) is associated with dopaminergic dysfunction that stems from substantia nigra (SN) degeneration and concomitant α-synuclein accumulation. Diffusion magnetic resonance imaging (MRI) can detect microstructural alterations of the SN and its tracts to (sub)cortical regions, but their pathological sensitivity is still poorly understood. OBJECTIVE To unravel the pathological substrate(s) underlying microstructural alterations of SN, and its tracts to the dorsal striatum and dorsolateral prefrontal cortex (DLPFC) in PD. METHODS Combining post-mortem in situ MRI and histopathology, T1-weighted and diffusion MRI, and neuropathological samples of nine PD, six PD with dementia (PDD), five dementia with Lewy bodies (DLB), and 10 control donors were collected. From diffusion MRI, mean diffusivity (MD) and fractional anisotropy (FA) were derived from the SN, and tracts between the SN and caudate nucleus, putamen, and DLPFC. Phosphorylated-Ser129-α-synuclein and tyrosine hydroxylase immunohistochemistry was included to quantify nigral Lewy pathology and dopaminergic degeneration, respectively. RESULTS Compared to controls, PD and PDD/DLB showed increased MD of the SN and SN-DLPFC tract, as well as increased FA of the SN-caudate nucleus tract. Both PD and PDD/DLB showed nigral Lewy pathology and dopaminergic loss compared to controls. Increased MD of the SN and FA of SN-caudate nucleus tract were associated with SN dopaminergic loss. Whereas increased MD of the SN-DLPFC tract was associated with increased SN Lewy neurite load. CONCLUSIONS In PD and PDD/DLB, diffusion MRI captures microstructural alterations of the SN and tracts to the dorsal striatum and DLPFC, which differentially associates with SN dopaminergic degeneration and Lewy neurite pathology. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Chen-Pei Lin
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Lydian E J Knoop
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Irene Frigerio
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - John G J M Bol
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Henk W Berendse
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Neurology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Petra J W Pouwels
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Wilma D J van de Berg
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Laura E Jonkman
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Kohe SE, Gowing EK, Seo S, Oorschot DE. A Novel Rat Model of ADHD-like Hyperactivity/Impulsivity after Delayed Reward Has Selective Loss of Dopaminergic Neurons in the Right Ventral Tegmental Area. Int J Mol Sci 2023; 24:11252. [PMID: 37511013 PMCID: PMC10379272 DOI: 10.3390/ijms241411252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
In attention deficit hyperactivity disorder (ADHD), hyperactivity and impulsivity occur in response to delayed reward. Herein we report a novel animal model in which male Sprague-Dawley rats exposed to repeated hypoxic brain injury during the equivalent of extreme prematurity were ADHD-like hyperactive/impulsive in response to delayed reward and attentive at 3 months of age. Thus, a unique animal model of one of the presentations/subtypes of ADHD was discovered. An additional finding is that the repeated hypoxia rats were not hyperactive in the widely used open field test, which is not ADHD specific. Hence, it is recommended that ADHD-like hyperactivity and ADHD-like impulsivity, specifically in response to delayed reward, be a primary component in the design of future experiments that characterize potential animal models of ADHD, replacing open field testing of hyperactivity. Unknown is whether death and/or activity of midbrain dopaminergic neurons contributed to the ADHD-like hyperactivity/impulsivity detected after delayed reward. Hence, we stereologically measured the absolute number of dopaminergic neurons in four midbrain subregions and the average somal/nuclear volume of those neurons. Repeated hypoxia rats had a significant specific loss of dopaminergic neurons in the right ventral tegmental area (VTA) at 2 weeks of age and 18 months of age, providing new evidence of a site of pathology. No dopaminergic neuronal loss occurred in three other midbrain regions. Fewer VTA dopaminergic neurons correlated with increased ADHD-like hyperactivity and impulsivity. Novel early intervention therapies to rescue VTA dopaminergic neurons and potentially prevent ADHD-like hyperactivity/impulsivity can now be investigated.
Collapse
Affiliation(s)
- Sarah E Kohe
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| | - Emma K Gowing
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| | - Steve Seo
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| | - Dorothy E Oorschot
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
12
|
Geng Y, Wang Z, Zhou J, Zhu M, Liu J, James TD. Recent progress in the development of fluorescent probes for imaging pathological oxidative stress. Chem Soc Rev 2023. [PMID: 37190785 DOI: 10.1039/d2cs00172a] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Oxidative stress is closely related to the physiopathology of numerous diseases. Reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive sulfur species (RSS) are direct participants and important biomarkers of oxidative stress. A comprehensive understanding of their changes can help us evaluate disease pathogenesis and progression and facilitate early diagnosis and drug development. In recent years, fluorescent probes have been developed for real-time monitoring of ROS, RNS and RSS levels in vitro and in vivo. In this review, conventional design strategies of fluorescent probes for ROS, RNS, and RSS detection are discussed from three aspects: fluorophores, linkers, and recognition groups. We introduce representative fluorescent probes for ROS, RNS, and RSS detection in cells, physiological/pathological processes (e.g., Inflammation, Drug Induced Organ Injury and Ischemia/Reperfusion Injury etc.), and specific diseases (e.g., neurodegenerative diseases, epilepsy, depression, diabetes and cancer, etc.). We then highlight the achievements, current challenges, and prospects for fluorescent probes in the pathophysiology of oxidative stress-related diseases.
Collapse
Affiliation(s)
- Yujie Geng
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Zhuo Wang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Jiaying Zhou
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Mingguang Zhu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Jiang Liu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Tony D James
- Department of Chemistry, University of Bath, Bath BA2 7AY, UK.
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| |
Collapse
|
13
|
Zakaria Z, Idris Z, Abdul Halim S, Ghani ARI, Abdullah JM. Subthalamic Nucleus (STN)-Deep Brain Stimulation Reduces the Power of Mu and Beta Rhythms and Enhances Synchrony at the Motor Cortices in Parkinson's Disease: A Report of Two Cases. Cureus 2023; 15:e35057. [PMID: 36942168 PMCID: PMC10024512 DOI: 10.7759/cureus.35057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2023] [Indexed: 02/18/2023] Open
Abstract
The motor circuit in Parkinson's disease (PD) involves the basal ganglia, thalamus, motor cortex, and cerebellum. Hence, subthalamic nucleus (STN) or globus pallidus internus deep brain stimulation is commonly used in treating refractory Parkinson's patients. During the procedure, the local field potential (LPF) is commonly made along the trajectory of the STN. Two cases were assessed, where an electroencephalographic recording at the sensorimotor cortices was also performed with and without stimulation at the optimal STN electrode site. The 'on' stimulation state associated with clinical improvement correlated with a marked reduction in the late theta (7.5 Hz), alpha (10.5 Hz) (Mu wave), and beta (20 Hz) wave power. Besides, more synchronized and coherent brainwaves were noted when the stimulation was 'on'.
Collapse
Affiliation(s)
- Zaitun Zakaria
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia (USM), Kota Bharu, MYS
| | - Zamzuri Idris
- Department of Neurosciences, School of Medical Sciences, Hospital Universiti Sains Malaysia (HUSM), Kota Bharu, MYS
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia (USM), Kota Bharu, MYS
| | - Sanihah Abdul Halim
- Department of Medicine, School of Medical Sciences, Universiti Sains Malaysia (USM) Kubang Kerian, Kota Bharu, MYS
| | - Abdul Rahman Izaini Ghani
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia (USM) Kubang Kerian, Kota Bharu, MYS
| | - Jafri M Abdullah
- Department of Neurosurgery, Universiti Sains Malaysia (USM) Health Campus, Kota Bharu, MYS
| |
Collapse
|
14
|
Prakash N. Developmental pathways linked to the vulnerability of adult midbrain dopaminergic neurons to neurodegeneration. Front Mol Neurosci 2022; 15:1071731. [PMID: 36618829 PMCID: PMC9815185 DOI: 10.3389/fnmol.2022.1071731] [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: 10/16/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
The degeneration of dopaminergic and other neurons in the aging brain is considered a process starting well beyond the infantile and juvenile period. In contrast to other dopamine-associated neuropsychiatric disorders, such as schizophrenia and drug addiction, typically diagnosed during adolescence or young adulthood and, thus, thought to be rooted in the developing brain, Parkinson's Disease (PD) is rarely viewed as such. However, evidences have accumulated suggesting that several factors might contribute to an increased vulnerability to death of the dopaminergic neurons at an already very early (developmental) phase in life. Despite the remarkable ability of the brain to compensate such dopamine deficits, the early loss or dysfunction of these neurons might predispose an individual to suffer from PD because the critical threshold of dopamine function will be reached much earlier in life, even if the time-course and strength of naturally occurring and age-dependent dopaminergic cell death is not markedly altered in this individual. Several signaling and transcriptional pathways required for the proper embryonic development of the midbrain dopaminergic neurons, which are the most affected in PD, either continue to be active in the adult mammalian midbrain or are reactivated at the transition to adulthood and under neurotoxic conditions. The persistent activity of these pathways often has neuroprotective functions in adult midbrain dopaminergic neurons, whereas the reactivation of silenced pathways under pathological conditions can promote the survival and even regeneration of these neurons in the lesioned or aging brain. This article summarizes our current knowledge about signaling and transcription factors involved in midbrain dopaminergic neuron development, whose reduced gene dosage or signaling activity are implicated in a lower survival rate of these neurons in the postnatal or aging brain. It also discusses the evidences supporting the neuroprotection of the midbrain dopaminergic system after the external supply or ectopic expression of some of these secreted and nuclear factors in the adult and aging brain. Altogether, the timely monitoring and/or correction of these signaling and transcriptional pathways might be a promising approach to a much earlier diagnosis and/or prevention of PD.
Collapse
|
15
|
Gaertner Z, Azcorra M, Dombeck DA, Awatramani R. Molecular heterogeneity in the substantia nigra: A roadmap for understanding PD motor pathophysiology. Neurobiol Dis 2022; 175:105925. [DOI: 10.1016/j.nbd.2022.105925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022] Open
|
16
|
Molecular and Cellular Interactions in Pathogenesis of Sporadic Parkinson Disease. Int J Mol Sci 2022; 23:ijms232113043. [PMID: 36361826 PMCID: PMC9657547 DOI: 10.3390/ijms232113043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/16/2022] [Accepted: 10/25/2022] [Indexed: 11/23/2022] Open
Abstract
An increasing number of the population all around the world suffer from age-associated neurodegenerative diseases including Parkinson’s disease (PD). This disorder presents different signs of genetic, epigenetic and environmental origin, and molecular, cellular and intracellular dysfunction. At the molecular level, α-synuclein (αSyn) was identified as the principal molecule constituting the Lewy bodies (LB). The gut microbiota participates in the pathogenesis of PD and may contribute to the loss of dopaminergic neurons through mitochondrial dysfunction. The most important pathogenetic link is an imbalance of Ca2+ ions, which is associated with redox imbalance in the cells and increased generation of reactive oxygen species (ROS). In this review, genetic, epigenetic and environmental factors that cause these disorders and their cause-and-effect relationships are considered. As a constituent of environmental factors, the example of organophosphates (OPs) is also reviewed. The role of endothelial damage in the pathogenesis of PD is discussed, and a ‘triple hit hypothesis’ is proposed as a modification of Braak’s dual hit one. In the absence of effective therapies for neurodegenerative diseases, more and more evidence is emerging about the positive impact of nutritional structure and healthy lifestyle on the state of blood vessels and the risk of developing these diseases.
Collapse
|
17
|
Houlihan KL, Keoseyan PP, Juba AN, Margaryan T, Voss ME, Babaoghli AM, Norris JM, Adrian GJ, Tovmasyan A, Buhlman LM. Folic Acid Improves Parkin-Null Drosophila Phenotypes and Transiently Reduces Vulnerable Dopaminergic Neuron Mitochondrial Hydrogen Peroxide Levels and Glutathione Redox Equilibrium. Antioxidants (Basel) 2022; 11:antiox11102068. [PMID: 36290790 PMCID: PMC9598960 DOI: 10.3390/antiox11102068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Loss-of-function parkin mutations cause oxidative stress and degeneration of dopaminergic neurons in the substantia nigra. Several consequences of parkin mutations have been described; to what degree they contribute to selective neurodegeneration remains unclear. Specific factors initiating excessive reactive oxygen species production, inefficient antioxidant capacity, or a combination are elusive. Identifying key oxidative stress contributors could inform targeted therapy. The absence of Drosophila parkin causes selective degeneration of a dopaminergic neuron cluster that is functionally homologous to the substantia nigra. By comparing observations in these to similar non-degenerating neurons, we may begin to understand mechanisms by which parkin loss of function causes selective degeneration. Using mitochondrially targeted redox-sensitive GFP2 fused with redox enzymes, we observed a sustained increased mitochondrial hydrogen peroxide levels in vulnerable dopaminergic neurons of parkin-null flies. Only transient increases in hydrogen peroxide were observed in similar but non-degenerating neurons. Glutathione redox equilibrium is preferentially dysregulated in vulnerable neuron mitochondria. To shed light on whether dysregulated glutathione redox equilibrium primarily contributes to oxidative stress, we supplemented food with folic acid, which can increase cysteine and glutathione levels. Folic acid improved survival, climbing, and transiently decreased hydrogen peroxide and glutathione redox equilibrium but did not mitigate whole-brain oxidative stress.
Collapse
Affiliation(s)
- Katherine L. Houlihan
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
| | - Petros P. Keoseyan
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Amber N. Juba
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
| | - Tigran Margaryan
- Department of Translational Neuroscience, Ivy Brain Tumor Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Max E. Voss
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | | | - Justin M. Norris
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Greg J. Adrian
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Artak Tovmasyan
- Department of Translational Neuroscience, Ivy Brain Tumor Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Lori M. Buhlman
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
- Correspondence: ; Tel.: +1-623-752-3668
| |
Collapse
|
18
|
Ukgansan Protects Dopaminergic Neurons against MPTP-Induced Neurotoxicity via the Nurr1 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7393557. [PMID: 36193151 PMCID: PMC9526663 DOI: 10.1155/2022/7393557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/11/2022] [Accepted: 08/08/2022] [Indexed: 11/18/2022]
Abstract
Nuclear receptor-related 1 protein (Nurr1) is a nuclear hormone receptor that protects dopaminergic neurons and is a promising therapeutic target for Parkinson’s disease (PD). Parkinson’s disease is a neurodegenerative disorder caused by the destruction of dopaminergic neurons in the substantia nigra pars compacta (SNpc), and the long-term use of conventional dopamine replacement therapies causes many side effects, highlighting the need for new treatments such as complementary and alternative medicine. Ukgansan has been used in East Asia to treat neurological disorders, including neurodegenerative diseases, and has been reported to have strong effects in treating patients with PD. In addition, recent studies have reported that Ukgansan has a neuroprotective potential. However, there are no detailed studies on the mechanism of action of Nurr1. Thus, unlike previous studies, we focused on the Nurr1 pathways. We confirmed neurotoxicity and apoptosis signaling in the differentiated PC12 cells. In addition, to confirm the protective effect of Ukgansan, we conducted behavioral tests (motor coordination and postural balance, and bradykinesia) and tyrosine hydroxylase immunohistochemistry in both the SNpc and striatum. Specifically, this study demonstrated the effect of Ukgansan in protecting dopaminergic neurons and increasing Nurr1 involved in maintaining dopamine levels by activating Nurr1 expression in MPTP-induced PC12 cells and a mouse model of PD. In this mechanism, the loss of dopaminergic neurons and dopamine depletion were suppressed, and motor impairment caused by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity was improved. These results provide evidence that Ukgansan ameliorates PD’s motor symptoms and progression.
Collapse
|
19
|
Zareba MR, Furman W, Binder M. Influence of age and cognitive performance on resting-state functional connectivity of dopaminergic and noradrenergic centers. Brain Res 2022; 1796:148082. [PMID: 36115586 DOI: 10.1016/j.brainres.2022.148082] [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: 03/26/2022] [Revised: 08/24/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022]
Abstract
Aging is associated with structural and functional changes in the brain, with a decline in cognitive functions observed as its inevitable concomitant. The body of literature suggests dopamine and noradrenaline as prominent candidate neuromodulators to mediate these effects; however, knowledge regarding the underlying mechanisms is scarce. To fill this gap, we compared resting-state functional connectivity (FC) patterns of ventral tegmental area (VTA), substantia nigra pars compacta (SNc) and locus coeruleus (LC) in healthy young (20-35 years; N = 37) and older adults (55-80 years; N = 27). Additionally, we sought FC patterns of these structures associated with performance in tasks probing executive, attentional and reward functioning, and we compared the functional coupling of the bilateral SNc. The results showed that individual SNc had stronger coupling with ipsilateral cortical and subcortical areas along with the contralateral cerebellum in the whole sample, and that the strength of connections of this structure with angular gyrus and lateral orbitofrontal cortex predicted visuomotor search abilities. In turn, older age was associated with greater local synchronization within VTA, its lower FC with caudate, mediodorsal thalamus, and SNc, as well as higher FC of both midbrain dopaminergic seeds with red nuclei. LC functional coupling showed no differences between the groups and was not associated with any of the behavioral functions. To the best of our knowledge, this work is the first to report the age-related effects on VTA local synchronization and its connectivity with key recipients of dopaminergic innervation, such as striatum and mediodorsal thalamus.
Collapse
Affiliation(s)
- Michal Rafal Zareba
- Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland; Institute of Psychology, Faculty of Philosophy, Jagiellonian University, Krakow, Poland
| | - Wiktoria Furman
- Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland; Institute of Psychology, Faculty of Philosophy, Jagiellonian University, Krakow, Poland.
| | - Marek Binder
- Institute of Psychology, Faculty of Philosophy, Jagiellonian University, Krakow, Poland
| |
Collapse
|
20
|
Kelly EA, Contreras J, Duan A, Vassell R, Fudge JL. Unbiased Stereological Estimates of Dopaminergic and GABAergic Neurons in the A10, A9, and A8 Subregions in the Young Male Macaque. Neuroscience 2022; 496:152-164. [PMID: 35738547 PMCID: PMC9329254 DOI: 10.1016/j.neuroscience.2022.06.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/25/2022] [Accepted: 06/10/2022] [Indexed: 11/28/2022]
Abstract
The ventral midbrain is the primary source of dopamine- (DA) expressing neurons in most species. GABA-ergic and glutamatergic cell populations are intermixed among DA-expressing cells and purported to regulate both local and long-range dopamine neuron activity. Most work has been conducted in rodent models, however due to evolutionary expansion of the ventral midbrain in primates, the increased size and complexity of DA subpopulations warrants further investigation. Here, we quantified the number of DA neurons, and their GABA-ergic complement in classic DA cell groups A10 (midline ventral tegmental area nuclei [VTA] and parabrachial pigmented nucleus [PBP]), A9 (substantia nigra, pars compacta [SNc]) and A8 (retrorubral field [RRF]) in the macaque. Because the PBP is a disproportionately expanded feature of the A10 group, and has unique connectional features in monkeys, we analyzed A10 data by dividing it into 'classic' midline nuclei and the PBP. Unbiased stereology revealed total putative DA neuron counts to be 210,238 ± 17,127 (A10 = 110,319 ± 9649, A9 = 87,399 ± 7751 and A8 = 12,520 ± 827). Putative GABAergic neurons were fewer overall, and evenly dispersed across the DA subpopulations (GAD67 = 71,215 ± 5663; A10 = 16,836 ± 2743; A9 = 24,855 ± 3144 and A8 = 12,633 ± 3557). Calculating the GAD67/TH ratio for each subregion revealed differential balances of these two cell types across the DA subregions. The A8 subregion had the highest complement of GAD67-positive neurons compared to TH-positive neurons (1:1), suggesting a potentially high capacity for GABAergic inhibition of DA output in this region.
Collapse
Affiliation(s)
- Emily A Kelly
- Department of Neuroscience, University of Rochester Medical Center, United States
| | - Jancy Contreras
- Department of Neuroscience, The City University of New York, United States
| | - Annie Duan
- Department of Neuroscience, University of Rochester Medical Center, United States
| | - Rochelle Vassell
- Department of Neuroscience, University of Rochester Medical Center, United States
| | - Julie L Fudge
- Department of Neuroscience, University of Rochester Medical Center, United States; Department of Psychiatry, University of Rochester Medical Center, United States.
| |
Collapse
|
21
|
Radulovic J, Ivkovic S, Adzic M. From chronic stress and anxiety to neurodegeneration: Focus on neuromodulation of the axon initial segment. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:481-495. [PMID: 35034756 DOI: 10.1016/b978-0-12-819410-2.00025-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
To adapt to the sustained demands of chronic stress, discrete brain circuits undergo structural and functional changes often resulting in anxiety disorders. In some individuals, anxiety disorders precede the development of motor symptoms of Parkinson's disease (PD) caused by degeneration of neurons in the substantia nigra (SN). Here, we present a circuit framework for probing a causal link between chronic stress, anxiety, and PD, which postulates a central role of abnormal neuromodulation of the SN's axon initial segment by brainstem inputs. It is grounded in findings demonstrating that the earliest PD pathologies occur in the stress-responsive, emotion regulation network of the brainstem, which provides the SN with dense aminergic and cholinergic innervation. SN's axon initial segment (AIS) has unique features that support the sustained and bidirectional propagation of activity in response to synaptic inputs. It is therefore, especially sensitive to circuit-mediated stress-induced imbalance of neuromodulation, and thus a plausible initiating site of neurodegeneration. This could explain why, although secondary to pathophysiologies in other brainstem nuclei, SN degeneration is the most extensive. Consequently, the cardinal symptom of PD, severe motor deficits, arise from degeneration of the nigrostriatal pathway rather than other brainstem nuclei. Understanding when and how circuit dysfunctions underlying anxiety can progress to neurodegeneration, raises the prospect of timed interventions for reversing, or at least impeding, the early pathophysiologies that lead to PD and possibly other neurodegenerative disorders.
Collapse
Affiliation(s)
- Jelena Radulovic
- Department of Neuroscience, Albert Einstein Medical College, Bronx, NY, United States; Department of Psychiatry and Behavioral Sciences, Albert Einstein Medical College, Bronx, NY, United States.
| | - Sanja Ivkovic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Miroslav Adzic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
22
|
Haynes JM, Sibuea SM, Aguiar AA, Li F, Ho JK, Pouton CW. Inhibition of β-catenin dependent WNT signalling upregulates the transcriptional repressor NR0B1 and downregulates markers of an A9 phenotype in human embryonic stem cell-derived dopaminergic neurons: Implications for Parkinson's disease. PLoS One 2021; 16:e0261730. [PMID: 34941945 PMCID: PMC8700011 DOI: 10.1371/journal.pone.0261730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/07/2021] [Indexed: 11/28/2022] Open
Abstract
In this study we investigate how β-catenin-dependent WNT signalling impacts midbrain dopaminergic neuron (mDA) specification. mDA cultures at day 65 of differentiation responded to 25 days of the tankyrase inhibitor XAV969 (XAV, 100nM) with reduced expression of markers of an A9 mDA phenotype (KCNJ6, ALDH1A1 and TH) but increased expression of the transcriptional repressors NR0B1 and NR0B2. Overexpression of NR0B1 and or NR0B2 promoted a loss of A9 dopaminergic neuron phenotype markers (KCNJ6, ALDH1A1 and TH). Overexpression of NR0B1, but not NR0B2 promoted a reduction in expression of the β-catenin-dependent WNT signalling pathway activator RSPO2. Analysis of Parkinson’s disease (PD) transcriptomic databases shows a profound PD-associated elevation of NR0B1 as well as reduced transcript for RSPO2. We conclude that reduced β-catenin-dependent WNT signalling impacts dopaminergic neuron identity, in vitro, through increased expression of the transcriptional repressor, NR0B1. We also speculate that dopaminergic neuron regulatory mechanisms may be perturbed in PD and that this may have an impact upon both existing nigral neurons and also neural progenitors transplanted as PD therapy.
Collapse
Affiliation(s)
- John M. Haynes
- Stem Cell Biology Group, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- * E-mail:
| | - Shanti M. Sibuea
- Stem Cell Biology Group, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Badan Pengawas Obat dan Makanan, Jakarta, Indonesia
| | - Alita A. Aguiar
- Stem Cell Biology Group, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Fangwei Li
- Stem Cell Biology Group, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Joan K. Ho
- Stem Cell Biology Group, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Colin W. Pouton
- Stem Cell Biology Group, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
23
|
Vidyadhara DJ, Yarreiphang H, Raju TR, Alladi PA. Differences in Neuronal Numbers, Morphology, and Developmental Apoptosis in Mice Nigra Provide Experimental Evidence of Ontogenic Origin of Vulnerability to Parkinson's Disease. Neurotox Res 2021; 39:1892-1907. [PMID: 34762290 DOI: 10.1007/s12640-021-00439-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 10/19/2022]
Abstract
Parkinson disease (PD) prevalence varies by ethnicity. In an earlier study, we replicated the reduced vulnerability to PD in an admixed population, using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-susceptible C57BL/6 J, MPTP-resistant CD-1 and their F1 crossbreds. In the present study, we investigated if the differences have a developmental origin. Substantia nigra was evaluated at postnatal days 2 (P2), P6, P10, P14, P18, and P22. C57BL/6 J mice had smaller nigra and fewer dopaminergic neurons than the CD-1 and crossbreds at P2, which persisted through development. A significant increase in numbers and nigral volume was observed across strains until P14. A drastic decline thereafter was specific to C57BL/6 J. CD-1 and crossbreds retained their numbers from P14 to stabilize with supernumerary neurons at adulthood. The neuronal size increased gradually to attain adult morphology at P10 in the resistant strains, vis-à-vis at P22 in C57BL/6 J. Accordingly, in comparison to C57BL/6 J, the nigra of CD-1 and reciprocal crossbreds possessed cytomorphological features of resilience, since birth. The considerably lesser dopaminergic neuronal loss in the CD-1 and crossbreds was seen at P2 and P14 and thereafter was complemented by attenuated developmental cell death. The differences in programmed cell death were confirmed by reduced TUNEL labelling, AIF, and caspase-3 expression. GDNF expression aligned with the cell death pattern at P2 and P14 in both nigra and striatum. Earlier maturity of nigra and its neurons appears to be better features that reflect as MPTP resistance at adulthood. Thus, variable MPTP vulnerability in mice and also differential susceptibility to PD in humans may arise early during nigral development.
Collapse
Affiliation(s)
- D J Vidyadhara
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
- Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Haorei Yarreiphang
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Trichur R Raju
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Phalguni Anand Alladi
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India.
- Formerly at Department of Neurophysiology, National Institute of Mental Health and Neuro-Sciences, Hosur Road, Bangalore, India.
| |
Collapse
|
24
|
Shim JH, Baek HM. Diffusion Measure Changes of Substantia Nigra Subregions and the Ventral Tegmental Area in Newly Diagnosed Parkinson's Disease. Exp Neurobiol 2021; 30:365-373. [PMID: 34737241 PMCID: PMC8572662 DOI: 10.5607/en21025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/13/2021] [Accepted: 09/22/2021] [Indexed: 12/22/2022] Open
Abstract
Historically, studies have extensively examined the basal ganglia in Parkinson’s disease for specific characteristics that can be observed with medical imaging. One particular methodology used for detecting changes that occur in Parkinson’s disease brains is diffusion tensor imaging, which yields diffusion indices such as fractional anisotropy and radial diffusivity that have been shown to correlate with axonal damage. In this study, we compare the diffusion measures of basal ganglia structures (with substantia nigra divided into subregions, pars compacta, and pars reticula), as well as the diffusion measures of the diffusion tracts that pass through each pair of basal ganglia structures to see if significant differences in diffusion measures can be observed in structures or tracts in newly diagnosed Parkinson’s disease patients. Additionally, we include the ventral tegmental area, a structure connected to various basal ganglia structures affected by dopaminergic neuronal loss and have historically shown significant alterations in Parkinson’s disease, in our analysis. We found significant fractional anisotropy differences in the putamen, and in the diffusion tracts that pass through pairs of both substantia nigra subregions, subthalamic nucleus, parabrachial pigmental nucleus, ventral tegmental area. Additionally, we found significant radial diffusivity differences in diffusion tracts that pass through the parabrachial nucleus, putamen, both substantia nigra subregions, and globus pallidus externa. We were able to find significant diffusion measure differences in structures and diffusion tracts, potentially due to compensatory mechanisms in response to dopaminergic neuronal loss that occurs in newly diagnosed Parkinson’s disease patients.
Collapse
Affiliation(s)
- Jae-Hyuk Shim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Hyeon-Man Baek
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| |
Collapse
|
25
|
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: 1.0] [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
|
26
|
Mannal N, Kleiner K, Fauler M, Dougalis A, Poetschke C, Liss B. Multi-Electrode Array Analysis Identifies Complex Dopamine Responses and Glucose Sensing Properties of Substantia Nigra Neurons in Mouse Brain Slices. Front Synaptic Neurosci 2021; 13:635050. [PMID: 33716704 PMCID: PMC7952765 DOI: 10.3389/fnsyn.2021.635050] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/08/2021] [Indexed: 12/16/2022] Open
Abstract
Dopaminergic (DA) midbrain neurons within the substantia nigra (SN) display an autonomous pacemaker activity that is crucial for dopamine release and voluntary movement control. Their progressive degeneration is a hallmark of Parkinson's disease. Their metabolically demanding activity-mode affects Ca2+ homeostasis, elevates metabolic stress, and renders SN DA neurons particularly vulnerable to degenerative stressors. Accordingly, their activity is regulated by complex mechanisms, notably by dopamine itself, via inhibitory D2-autoreceptors and the neuroprotective neuronal Ca2+ sensor NCS-1. Analyzing regulation of SN DA neuron activity-pattern is complicated by their high vulnerability. We studied this activity and its control by dopamine, NCS-1, and glucose with extracellular multi-electrode array (MEA) recordings from midbrain slices of juvenile and adult mice. Our tailored MEA- and spike sorting-protocols allowed high throughput and long recording times. According to individual dopamine-responses, we identified two distinct SN cell-types, in similar frequency: dopamine-inhibited and dopamine-excited neurons. Dopamine-excited neurons were either silent in the absence of dopamine, or they displayed pacemaker-activities, similar to that of dopamine-inhibited neurons. Inhibition of pacemaker-activity by dopamine is typical for SN DA neurons, and it can undergo prominent desensitization. We show for adult mice, that the number of SN DA neurons with desensitized dopamine-inhibition was increased (~60–100%) by a knockout of NCS-1, or by prevention of NCS-1 binding to D2-autoreceptors, while time-course and degrees of desensitization were not altered. The number of neurons with desensitized D2-responses was also higher (~65%) at high glucose-levels (25 mM), compared to lower glucose (2.5 mM), while again desensitization-kinetics were unaltered. However, spontaneous firing-rates were significantly higher at high glucose-levels (~20%). Moreover, transient glucose-deprivation (1 mM) induced a fast and fully-reversible pacemaker frequency reduction. To directly address and quantify glucose-sensing properties of SN DA neurons, we continuously monitored their electrical activity, while altering extracellular glucose concentrations stepwise from 0.5 mM up to 25 mM. SN DA neurons were excited by glucose, with EC50 values ranging from 0.35 to 2.3 mM. In conclusion, we identified a novel, common subtype of dopamine-excited SN neurons, and a complex, joint regulation of dopamine-inhibited neurons by dopamine and glucose, within the range of physiological brain glucose-levels.
Collapse
Affiliation(s)
- Nadja Mannal
- Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | | | - Michael Fauler
- Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | | | | | - Birgit Liss
- Institute of Applied Physiology, University of Ulm, Ulm, Germany.,Linacre and New College, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
27
|
A probabilistic atlas of the human ventral tegmental area (VTA) based on 7 Tesla MRI data. Brain Struct Funct 2021; 226:1155-1167. [PMID: 33580320 PMCID: PMC8036186 DOI: 10.1007/s00429-021-02231-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/26/2021] [Indexed: 12/12/2022]
Abstract
Functional magnetic resonance imaging (fMRI) BOLD signal is commonly localized by using neuroanatomical atlases, which can also serve for region of interest analyses. Yet, the available MRI atlases have serious limitations when it comes to imaging subcortical structures: only 7% of the 455 subcortical nuclei are captured by current atlases. This highlights the general difficulty in mapping smaller nuclei deep in the brain, which can be addressed using ultra-high field 7 Tesla (T) MRI. The ventral tegmental area (VTA) is a subcortical structure that plays a pivotal role in reward processing, learning and memory. Despite the significant interest in this nucleus in cognitive neuroscience, there are currently no available, anatomically precise VTA atlases derived from 7 T MRI data that cover the full region of the VTA. Here, we first provide a protocol for multimodal VTA imaging and delineation. We then provide a data description of a probabilistic VTA atlas based on in vivo 7 T MRI data.
Collapse
|
28
|
Fujishiro H, Torii Y, Iritani S, Ozaki N. Dopaminergic circuitry in late-life depression and Lewy body disease. Psychiatry Clin Neurosci 2021; 75:69-70. [PMID: 33247877 DOI: 10.1111/pcn.13181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/19/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Hiroshige Fujishiro
- Department of Psychiatry, Kawasaki Memorial Hospital, Kawasaki, Japan.,Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Youta Torii
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shuji Iritani
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
29
|
Nouri P, Götz S, Rauser B, Irmler M, Peng C, Trümbach D, Kempny C, Lechermeier CG, Bryniok A, Dlugos A, Euchner E, Beckers J, Brodski C, Klümper C, Wurst W, Prakash N. Dose-Dependent and Subset-Specific Regulation of Midbrain Dopaminergic Neuron Differentiation by LEF1-Mediated WNT1/b-Catenin Signaling. Front Cell Dev Biol 2020; 8:587778. [PMID: 33195246 PMCID: PMC7649324 DOI: 10.3389/fcell.2020.587778] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/01/2020] [Indexed: 01/07/2023] Open
Abstract
The mesodiencephalic dopaminergic (mdDA) neurons, including the nigrostriatal subset that preferentially degenerates in Parkinson’s Disease (PD), strongly depend on an accurately balanced Wingless-type MMTV integration site family member 1 (WNT1)/beta-catenin signaling pathway during their development. Loss of this pathway abolishes the generation of these neurons, whereas excessive WNT1/b-catenin signaling prevents their correct differentiation. The identity of the cells responding to this pathway in the developing mammalian ventral midbrain (VM) as well as the precise progression of WNT/b-catenin action in these cells are still unknown. We show that strong WNT/b-catenin signaling inhibits the differentiation of WNT/b-catenin-responding mdDA progenitors into PITX3+ and TH+ mdDA neurons by repressing the Pitx3 gene in mice. This effect is mediated by RSPO2, a WNT/b-catenin agonist, and lymphoid enhancer binding factor 1 (LEF1), an essential nuclear effector of the WNT/b-catenin pathway, via conserved LEF1/T-cell factor binding sites in the Pitx3 promoter. LEF1 expression is restricted to a caudolateral mdDA progenitor subset that preferentially responds to WNT/b-catenin signaling and gives rise to a fraction of all mdDA neurons. Our data indicate that an attenuation of WNT/b-catenin signaling in mdDA progenitors is essential for their correct differentiation into specific mdDA neuron subsets. This is an important consideration for stem cell-based regenerative therapies and in vitro models of neuropsychiatric diseases.
Collapse
Affiliation(s)
- Parivash Nouri
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| | - Sebastian Götz
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Benedict Rauser
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Changgeng Peng
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Advanced Institute of Translational Medicine, The First Rehabilitation Hospital of Shanghai, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Christian Kempny
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| | - Carina G Lechermeier
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Agnes Bryniok
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| | - Andrea Dlugos
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| | - Ellen Euchner
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Chair of Experimental Genetics, Technical University of Munich, Munich, Germany.,German Center for Diabetes Research, Neuherberg, Germany
| | - Claude Brodski
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Claudia Klümper
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Chair of Developmental Genetics, Helmholtz Zentrum München, Technical University of Munich/Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nilima Prakash
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| |
Collapse
|
30
|
Richter A, Reinhard F, Kraemer B, Gruber O. A high-resolution fMRI approach to characterize functionally distinct neural pathways within dopaminergic midbrain and nucleus accumbens during reward and salience processing. Eur Neuropsychopharmacol 2020; 36:137-150. [PMID: 32546416 DOI: 10.1016/j.euroneuro.2020.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/15/2020] [Accepted: 05/24/2020] [Indexed: 01/12/2023]
Abstract
Processing of reward and salience without reward association are known to critically rely on the dopamine system. A growing body of evidence from animal studies suggests that both functions may be subserved by distinct subregions in midbrain and ventral striatum, specifically nucleus accumbens (NAcc). Yet in vivo investigation of these brain structures in humans has been rare. Here we examined blood oxygen level dependent signals in response to frequently presented rewarding events and infrequently presented neutral events in 20 healthy subjects using high-resolution functional magnetic resonance imaging (fMRI) for imaging the human midbrain and NAcc. The present findings revealed distinct activation patterns in brain regions of interest, namely increased activation in substantia nigra pars compacta (SNc) and dorsolateral NAcc in response to neutral events, while the VTA and both the ventromedial and dorsolateral NAcc were significantly activated due to rewarding events. Moreover, psychophysiological interaction analyses demonstrated regionally specialized processing pathways, such as a dorsolateral pathway when processing salience per se, i.e. increased functional interactions between SNc, dorsolateral NAcc and dorsolateral and medial prefrontal cortex (PFC); and a ventromedial pathway during reward processing, i.e. increased functional coupling between VTA and ventromedial NAcc. Thus, these findings may not only accelerate the integration of animal models of brain function with human neuroscience but may also improve diagnosis and treatment in patients with neuropsychiatric disorders such as schizophrenia and depression in which dopaminergic dysfunction and aberrant attribution of salience have been implicated.
Collapse
Affiliation(s)
- Anja Richter
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, Heidelberg University, Heidelberg, Germany; Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| | - Fabian Reinhard
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, Heidelberg University, Heidelberg, Germany
| | - Bernd Kraemer
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, Heidelberg University, Heidelberg, Germany
| | - Oliver Gruber
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
31
|
TGF-β Signaling Regulates SLC8A3 Expression and Prevents Oxidative Stress in Developing Midbrain Dopaminergic and Dorsal Raphe Serotonergic Neurons. Int J Mol Sci 2020; 21:ijms21082735. [PMID: 32326436 PMCID: PMC7216069 DOI: 10.3390/ijms21082735] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/08/2020] [Accepted: 04/12/2020] [Indexed: 01/15/2023] Open
Abstract
Calcium homeostasis is a cellular process required for proper cell function and survival, maintained by the coordinated action of several transporters, among them members of the Na+/Ca2+-exchanger family, such as SLC8A3. Transforming growth factor beta (TGF-β) signaling defines neuronal development and survival and may regulate the expression of channels and transporters. We investigated the regulation of SLC8A3 by TGF-β in a conditional knockout mouse with deletion of TGF-β signaling from Engrailed 1-expressing cells, i.e., in cells from the midbrain and rhombomere 1, and elucidated the underlying molecular mechanisms. The results show that SLC8A3 is significantly downregulated in developing dopaminergic and dorsal raphe serotonergic neurons in mutants and that low SLC8A3 abundance prevents the expression of the anti-apoptotic protein Bcl-xL. TGF-β signaling affects SLC8A3 via the canonical and p38 signaling pathway and may increase the binding of Smad4 to the Slc8a3 promoter. Expression of the lipid peroxidation marker malondialdehyde (MDA) was increased following knockdown of Slc8a3 expression in vitro. In neurons lacking TGF-β signaling, the number of MDA- and 4-hydroxynonenal (4-HNE)-positive cells was significantly increased, accompanied with increased cellular 4-HNE abundance. These results suggest that TGF-β contributes to the regulation of SLC8A3 expression in developing dopaminergic and dorsal raphe serotonergic neurons, thereby preventing oxidative stress.
Collapse
|
32
|
Farassat N, Costa KM, Stojanovic S, Albert S, Kovacheva L, Shin J, Egger R, Somayaji M, Duvarci S, Schneider G, Roeper J. In vivo functional diversity of midbrain dopamine neurons within identified axonal projections. eLife 2019; 8:48408. [PMID: 31580257 PMCID: PMC6791716 DOI: 10.7554/elife.48408] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/02/2019] [Indexed: 12/03/2022] Open
Abstract
Functional diversity of midbrain dopamine (DA) neurons ranges across multiple scales, from differences in intrinsic properties and connectivity to selective task engagement in behaving animals. Distinct in vitro biophysical features of DA neurons have been associated with different axonal projection targets. However, it is unknown how this translates to different firing patterns of projection-defined DA subpopulations in the intact brain. We combined retrograde tracing with single-unit recording and labelling in mouse brain to create an in vivo functional topography of the midbrain DA system. We identified differences in burst firing among DA neurons projecting to dorsolateral striatum. Bursting also differentiated DA neurons in the medial substantia nigra (SN) projecting either to dorsal or ventral striatum. We found differences in mean firing rates and pause durations among ventral tegmental area (VTA) DA neurons projecting to lateral or medial shell of nucleus accumbens. Our data establishes a high-resolution functional in vivo landscape of midbrain DA neurons.
Collapse
Affiliation(s)
- Navid Farassat
- Institute for Neurophysiology, Goethe University, Frankfurt, Germany
| | | | | | - Stefan Albert
- Institute for Mathematics, Goethe University, Frankfurt, Germany
| | - Lora Kovacheva
- Institute for Neurophysiology, Goethe University, Frankfurt, Germany
| | - Josef Shin
- Institute for Neurophysiology, Goethe University, Frankfurt, Germany
| | - Richard Egger
- Institute for Neurophysiology, Goethe University, Frankfurt, Germany
| | | | - Sevil Duvarci
- Institute for Neurophysiology, Goethe University, Frankfurt, Germany
| | - Gaby Schneider
- Institute for Mathematics, Goethe University, Frankfurt, Germany
| | - Jochen Roeper
- Institute for Neurophysiology, Goethe University, Frankfurt, Germany
| |
Collapse
|
33
|
Dolgacheva LP, Berezhnov AV, Fedotova EI, Zinchenko VP, Abramov AY. Role of DJ-1 in the mechanism of pathogenesis of Parkinson's disease. J Bioenerg Biomembr 2019; 51:175-188. [PMID: 31054074 PMCID: PMC6531411 DOI: 10.1007/s10863-019-09798-4] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/24/2019] [Indexed: 12/13/2022]
Abstract
DJ-1 protein has multiple specific mechanisms to protect dopaminergic neurons against neurodegeneration in Parkinson's disease. Wild type DJ-1 can acts as oxidative stress sensor and as an antioxidant. DJ-1 exhibits the properties of molecular chaperone, protease, glyoxalase, transcriptional regulator that protects mitochondria from oxidative stress. DJ-1 increases the expression of two mitochondrial uncoupling proteins (UCP 4 and UCP5), that decrease mitochondrial membrane potential and leads to the suppression of ROS production, optimizes of a number of mitochondrial functions, and is regarded as protection for the neuronal cell survival. We discuss also the stabilizing interaction of DJ-1 with the mitochondrial Bcl-xL protein, which regulates the activity of (Inositol trisphosphate receptor) IP3R, prevents the cytochrome c release from mitochondria and inhibits the apoptosis activation. Upon oxidative stress DJ-1 is able to regulate various transcription factors including nuclear factor Nrf2, PI3K/PKB, and p53 signal pathways. Stress-activated transcription factor Nrf2 regulates the pathways to protect cells against oxidative stress and metabolic pathways initiating the NADPH and ATP production. DJ-1 induces the Nrf2 dissociation from its inhibitor Keap1 (Kelch-like ECH-associated protein 1), promoting Nrf2 nuclear translocation and binding to antioxidant response elements. DJ-1 is shown to be a co-activator of the transcription factor NF-kB. Under nitrosative stress, DJ-1 may regulate PI3K/PKB signaling through PTEN transnitrosylation, which leads to inhibition of phosphatase activity. DJ-1 has a complex modulating effect on the p53 pathway: one side DJ-1 directly binds to p53 to restore its transcriptional activity and on the other hand DJ-1 can stimulate deacylation and suppress p53 transcriptional activity. The ability of the DJ-1 to induce activation of different transcriptional factors and change redox balance protect neurons against aggregation of α-synuclein and oligomer-induced neurodegeneration.
Collapse
Affiliation(s)
- Ludmila P Dolgacheva
- Institute of Cell Biophysics Russian Academy of Sciences, Pushchino, 142290, Russia.
| | - Alexey V Berezhnov
- Institute of Cell Biophysics Russian Academy of Sciences, Pushchino, 142290, Russia
| | - Evgeniya I Fedotova
- Institute of Cell Biophysics Russian Academy of Sciences, Pushchino, 142290, Russia
| | - Valery P Zinchenko
- Institute of Cell Biophysics Russian Academy of Sciences, Pushchino, 142290, Russia
| | - Andrey Y Abramov
- Department of Clinical and Movement Neurosciences, UCL Institute of Neurology, London, WC1N 3BG, UK.
| |
Collapse
|
34
|
Functional neuroanatomical review of the ventral tegmental area. Neuroimage 2019; 191:258-268. [PMID: 30710678 DOI: 10.1016/j.neuroimage.2019.01.062] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 12/19/2022] Open
Abstract
The ventral tegmental area (VTA) and substantia nigra pars compacta (SNc) are assumed to play a key role in dopamine-related functions such as reward-related behaviour, motivation, addiction and motor functioning. Although dopamine-producing midbrain structures are bordering, they show significant differences in structure and function that argue for a distinction when studying the functions of the dopaminergic midbrain, especially by means of neuroimaging. First, unlike the SNc, the VTA is not a nucleus, which makes it difficult to delineate the structure due to lack of clear anatomical borders. Second, there is no consensus in the literature about the anatomical nomenclature to describe the VTA. Third, these factors in combination with limitations in magnetic resonance imaging (MRI) complicate VTA visualization. We suggest that developing an MRI-compatible probabilistic atlas of the VTA will help to overcome these issues. Such an atlas can be used to identify the individual VTA and serve as region-of-interest for functional MRI.
Collapse
|
35
|
Wang Y, Zhou FM. Striatal But Not Extrastriatal Dopamine Receptors Are Critical to Dopaminergic Motor Stimulation. Front Pharmacol 2017; 8:935. [PMID: 29311936 PMCID: PMC5742616 DOI: 10.3389/fphar.2017.00935] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 12/11/2017] [Indexed: 11/19/2022] Open
Abstract
Dopamine (DA) is required for motor function in vertebrate animals including humans. The striatum, a key motor control center, receives a dense DA innervation and express high levels of DA D1 receptors (D1Rs) and D2 receptors (D2Rs). Other brain areas involved in motor function such as the globus pallidus external segment (GPe) and the substantia nigra pars reticulata (SNr) and the motor cortex (MC) also receive DA innervation and express DA receptors. Thus, the relative contribution of the striatal and extrastriatal DA systems to the motor function has been an important question critical for understanding the functional operation of the motor control circuits and also for therapeutic targeting. We have now experimentally addressed this question in the transcription factor Pitx3 null mutant (Pitx3Null) mice that have an autogenic and parkinsonian-like striatal DA denervation and hence supersensitive motor response to DA stimulation. Using DA agonist unilateral microinjection-induced rotation as a reliable readout of motor stimulation, our results show that L-dopa microinjection into the dorsal striatum (DS) induced 5–10 times more rotations than that induced by L-dopa microinjection into GPe and SNr, while L-dopa microinjection into the primary MC induced the least number of rotations. Furthermore, our results show that separate microinjection of the D1R-like agonist SKF81297 and the D2R-like agonist ropinirole into the DS each induced only modest numbers of rotation, whereas concurrent injection of the two agonists triggered more rotations than the sum of the rotations induced by each of these two agonists separately, indicating D1R–D2R synergy. These results suggest that the striatum, not GPe, SNr or MC, is the primary site for D1Rs and D2Rs to synergistically stimulate motor function in L-dopa treatment of Parkinson’s disease (PD). Our results also predict that non-selective, broad spectrum DA agonists activating both D1Rs and D2Rs are more efficacious anti-PD drugs than the current D2R agonists.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Pharmacology, University of Tennessee College of Medicine, Memphis, TN, United States
| | - Fu-Ming Zhou
- Department of Pharmacology, University of Tennessee College of Medicine, Memphis, TN, United States
| |
Collapse
|
36
|
Datta I, Ganapathy K, Razdan R, Bhonde R. Location and Number of Astrocytes Determine Dopaminergic Neuron Survival and Function Under 6-OHDA Stress Mediated Through Differential BDNF Release. Mol Neurobiol 2017; 55:5505-5525. [PMID: 28965325 DOI: 10.1007/s12035-017-0767-0] [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: 04/22/2017] [Accepted: 09/06/2017] [Indexed: 02/08/2023]
Abstract
While astrocytes throughout the CNS share many common traits, they exhibit significant differences in function and number among brain regions. The aim of the present study is to assess the effect of region-specificity and number of astrocytes on the survival of dopaminergic neurons under stress, and to understand the possible mechanism by which these astrocytes extend neuroprotection to dopaminergic neurons. Purified astrocytes obtained from forebrain, midbrain, and hindbrain region were characterized through FACS and immunofluorescence. Co-culture experiments (using trans-wells) were then performed to measure the effect of region-specificities and numbers of astrocytes on primary midbrain culture under 6-OHDA stress. Cell survival augmented with an increase in astrocyte seeding number and total cell survival was comparable among the different region-specific astrocytes for all numbers. However, striking differences were observed in dopaminergic neuronal (TH) cell survival in the presence of midbrain astrocytes in comparison to forebrain and hindbrain astrocytes at all seeding numbers. At 75 μM 6-OHDA insult, while cell survival was comparable in purified astrocytes from the different brain regions, a distinct increase in BDNF secretion (significantly higher than its constitutive release) was noted for midbrain astrocytes compared to forebrain and hindbrain astrocytes. The TH immunopositive population decreased when TrkB inhibitor was added to the co-culture under 6-OHDA toxicity, suggesting that BDNF released by co-cultured astrocytes plays a key role in the survival of dopaminergic neurons. This BDNF release decreased in presence of NO inhibitor and increased in the presence of NO donor (DETA/NO). We conclude that the BDNF released from astrocytes under 6-OHDA toxicity is mediated through NO release through both autocrine and paracrine signaling, and this BDNF release is primarily responsible for the differential effect of region-specific astrocytes on TH neuron survival under these conditions.
Collapse
Affiliation(s)
- Indrani Datta
- Department of Biophysics, National Institute of Mental Health and Neurosciences, an Institute of National Importance, P.B. No 2900, Hosur Road, Bengaluru, Karnataka, 560029, India.
| | - Kavina Ganapathy
- Department of Pharmacology, Al-Ameen College of Pharmacy, Bengaluru, Karnataka, India
| | - Rema Razdan
- School of Regenerative Medicine, Manipal University, Bengaluru, Karnataka, India
| | - Ramesh Bhonde
- Department of Pharmacology, Al-Ameen College of Pharmacy, Bengaluru, Karnataka, India
| |
Collapse
|
37
|
Feuerstein M, Chleilat E, Khakipoor S, Michailidis K, Ophoven C, Roussa E. Expression patterns of key Sonic Hedgehog signaling pathway components in the developing and adult mouse midbrain and in the MN9D cell line. Cell Tissue Res 2017; 370:211-225. [PMID: 28799057 PMCID: PMC5640734 DOI: 10.1007/s00441-017-2664-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 12/29/2022]
Abstract
The temporal dynamic expression of Sonic Hedgehog (SHH) and signaling during early midbrain dopaminergic (mDA) neuron development is one of the key players in establishing mDA progenitor diversity. However, whether SHH signaling is also required during later developmental stages and in mature mDA neurons is less understood. We study the expression of SHH receptors Ptch1 and Gas1 (growth arrest-specific 1) and of the transcription factors Gli1, Gli2 and Gli3 in mouse midbrain during embryonic development [embryonic day (E) 12.5 onwards)], in newborn and adult mice using in situ hybridization and immunohistochemistry. Moreover, we examine the expression and regulation of dopaminergic neuronal progenitor markers, midbrain dopaminergic neuronal markers and markers of the SHH signaling pathway in undifferentiated and butyric acid-treated (differentiated) MN9D cells in the presence or absence of exogenous SHH in vitro by RT-PCR, immunoblotting and immunocytochemistry. Gli1 was expressed in the lateral mesencephalic domains, whereas Gli2 and Gli3 were expressed dorsolaterally and complemented by ventrolateral expression of Ptch1. Co-localization with tyrosine hydroxylase could not be observed. GAS1 was exclusively expressed in the dorsal mesencephalon at E11.5 and co-localized with Ki67. In contrast, MN9D cells expressed all the genes investigated and treatment of the cells with butyric acid significantly upregulated their expression. The results suggest that SHH is only indirectly involved in the differentiation and survival of mDA neurons and that the MN9D cell line is a valuable model for investigating early development but not the differentiation and survival of mDA neurons.
Collapse
Affiliation(s)
- Melanie Feuerstein
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, D-79104, Freiburg, Germany
| | - Enaam Chleilat
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, D-79104, Freiburg, Germany
| | - Shokoufeh Khakipoor
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, D-79104, Freiburg, Germany
| | - Konstantinos Michailidis
- Neuroanatomy, Faculty of Medicine, University of Freiburg, Albertstrasse 17, D-79104, Freiburg, Germany
| | - Christian Ophoven
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, D-79104, Freiburg, Germany
| | - Eleni Roussa
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, D-79104, Freiburg, Germany. .,Neuroanatomy, Faculty of Medicine, University of Freiburg, Albertstrasse 17, D-79104, Freiburg, Germany.
| |
Collapse
|
38
|
|
39
|
Altered brain metabolic connectivity at multiscale level in early Parkinson's disease. Sci Rep 2017; 7:4256. [PMID: 28652595 PMCID: PMC5484707 DOI: 10.1038/s41598-017-04102-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/11/2017] [Indexed: 01/29/2023] Open
Abstract
To explore the effects of PD pathology on brain connectivity, we characterized with an emergent computational approach the brain metabolic connectome using [18F]FDG-PET in early idiopathic PD patients. We applied whole-brain and pathology-based connectivity analyses, using sparse-inverse covariance estimation in thirty-four cognitively normal PD cases and thirty-four age-matched healthy subjects for comparisons. Further, we assessed high-order resting state networks by interregional correlation analysis. Whole-brain analysis revealed altered metabolic connectivity in PD, with local decreases in frontolateral cortex and cerebellum and increases in the basal ganglia. Widespread long-distance decreases were present within the frontolateral cortex as opposed to connectivity increases in posterior cortical regions, all suggestive of a global-scale connectivity reconfiguration. The pathology-based analyses revealed significant connectivity impairment in the nigrostriatal dopaminergic pathway and in the regions early affected by α-synuclein pathology. Notably, significant connectivity changes were present in several resting state networks especially in frontal regions. These findings expand previous imaging evidence of altered connectivity in cognitively stable PD patients by showing pathology-based connectivity changes and disease-specific metabolic architecture reconfiguration at multiple scale levels, from the earliest PD phases. These alterations go well beyond the known striato-cortical connectivity derangement supporting in vivo an extended neural vulnerability in the PD synucleinopathy.
Collapse
|
40
|
Oliveira MAP, Balling R, Smidt MP, Fleming RMT. Embryonic development of selectively vulnerable neurons in Parkinson's disease. NPJ Parkinsons Dis 2017; 3:21. [PMID: 28685157 PMCID: PMC5484687 DOI: 10.1038/s41531-017-0022-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 05/24/2017] [Accepted: 06/01/2017] [Indexed: 02/07/2023] Open
Abstract
A specific set of brainstem nuclei are susceptible to degeneration in Parkinson's disease. We hypothesise that neuronal vulnerability reflects shared phenotypic characteristics that confer selective vulnerability to degeneration. Neuronal phenotypic specification is mainly the cumulative result of a transcriptional regulatory program that is active during the development. By manual curation of the developmental biology literature, we comprehensively reconstructed an anatomically resolved cellular developmental lineage for the adult neurons in five brainstem regions that are selectively vulnerable to degeneration in prodromal or early Parkinson's disease. We synthesised the literature on transcription factors that are required to be active, or required to be inactive, in the development of each of these five brainstem regions, and at least two differentially vulnerable nuclei within each region. Certain transcription factors, e.g., Ascl1 and Lmx1b, seem to be required for specification of many brainstem regions that are susceptible to degeneration in early Parkinson's disease. Some transcription factors can even distinguish between differentially vulnerable nuclei within the same brain region, e.g., Pitx3 is required for specification of the substantia nigra pars compacta, but not the ventral tegmental area. We do not suggest that Parkinson's disease is a developmental disorder. In contrast, we consider identification of shared developmental trajectories as part of a broader effort to identify the molecular mechanisms that underlie the phenotypic features that are shared by selectively vulnerable neurons. Systematic in vivo assessment of fate determining transcription factors should be completed for all neuronal populations vulnerable to degeneration in early Parkinson's disease.
Collapse
Affiliation(s)
- Miguel A. P. Oliveira
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, Belvaux, L-4362 Luxembourg
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, Belvaux, L-4362 Luxembourg
| | - Marten P. Smidt
- Department of Molecular Neuroscience, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Sciencepark 904, 1098 XH Amsterdam, The Netherlands
| | - Ronan M. T. Fleming
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, Belvaux, L-4362 Luxembourg
| |
Collapse
|
41
|
Caminiti SP, Presotto L, Baroncini D, Garibotto V, Moresco RM, Gianolli L, Volonté MA, Antonini A, Perani D. Axonal damage and loss of connectivity in nigrostriatal and mesolimbic dopamine pathways in early Parkinson's disease. NEUROIMAGE-CLINICAL 2017; 14:734-740. [PMID: 28409113 PMCID: PMC5379906 DOI: 10.1016/j.nicl.2017.03.011] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 03/13/2017] [Accepted: 03/24/2017] [Indexed: 12/20/2022]
Abstract
A progressive loss of dopamine neurons in the substantia nigra (SN) is considered the main feature of idiopathic Parkinson's disease (PD). Recent neuropathological evidence however suggests that the axons of the nigrostriatal dopaminergic system are the earliest target of α-synuclein accumulation in PD, thus the principal site for vulnerability. Whether this applies to in vivo PD, and also to the mesolimbic system has not been investigated yet. We used [11C]FeCIT PET to measure presynaptic dopamine transporter (DAT) activity in both nigrostriatal and mesolimbic systems, in 36 early PD patients (mean disease duration in months ± SD 21.8 ± 10.7) and 14 healthy controls similar for age. We also performed anatomically-driven partial correlation analysis to evaluate possible changes in the connectivity within both the dopamine networks at an early clinical phase. In the nigrostriatal system, we found a severe DAT reduction in the afferents to the dorsal putamen (DPU) (η2 = 0.84), whereas the SN was the less affected region (η2 = 0.31). DAT activity in the ventral tegmental area (VTA) and the ventral striatum (VST) were also reduced in the patient group, but to a lesser degree (VST η2 = 0.71 and VTA η2 = 0.31). In the PD patients compared to the controls, there was a marked decrease in dopamine network connectivity between SN and DPU nodes, supporting the significant derangement in the nigrostriatal pathway. These results suggest that neurodegeneration in the dopamine pathways is initially more prominent in the afferent axons and more severe in the nigrostriatal system. Considering PD as a disconnection syndrome starting from the axons, it would justify neuroprotective interventions even if patients have already manifested clinical symptoms. In vivo study of mesolimbic and nigrostriatal dopamine systems in early iPD Evidence for a severe axonal damage with relative sparing of SN Evidence for a moderate damage of the mesolimbic pathway in early iPD Significant reduction of molecular connectivity between nigrostriatal nodes Justification for neuroprotective interventions in early-iPD phase
Collapse
Affiliation(s)
- Silvia Paola Caminiti
- Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina, 58, 20132 Milan, Italy
| | - Luca Presotto
- Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina, 58, 20132 Milan, Italy
| | - Damiano Baroncini
- Department of Neurology, San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy
| | - Valentina Garibotto
- Department of Medical Imaging, Geneva University, Geneva University Hospitals, Geneva, Switzerland
| | - Rosa Maria Moresco
- IBFM-CNR, Segrate, Italy, Tecnomed Foundation, Department of Health Sciences, University of Milan-Bicocca, Monza, Italy
| | - Luigi Gianolli
- Nuclear Medicine Unit, San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy
| | | | - Angelo Antonini
- Parkinson's Disease and Movement Disorders Unit, I.R.C.C.S Hospital San Camillo, Via Alberoni 70, 30126 Venice, Italy.,Department of Neurosciences (DNS), University of Padua, Via Giustiniani 5, 35128 Padova, Italy
| | - Daniela Perani
- Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina, 58, 20132 Milan, Italy.,Nuclear Medicine Unit, San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy
| |
Collapse
|
42
|
Richter F, Gabby L, McDowell KA, Mulligan CK, De La Rosa K, Sioshansi PC, Mortazavi F, Cely I, Ackerson LC, Tsan L, Murphy NP, Maidment NT, Chesselet MF. Effects of decreased dopamine transporter levels on nigrostriatal neurons and paraquat/maneb toxicity in mice. Neurobiol Aging 2016; 51:54-66. [PMID: 28038352 DOI: 10.1016/j.neurobiolaging.2016.11.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 01/19/2023]
Abstract
How genetic variations in the dopamine transporter (DAT) combined with exposure to environmental toxins modulate the risk of Parkinson's disease remains unclear. Using unbiased stereology in DAT knock-down mice (DAT-KD) and wild-type (WT) littermates, we found that decreased DAT caused a loss of tyrosine hydroxylase-positive (dopaminergic) neurons in subregions of the substantia nigra pars compacta at 3-4 days, 5 weeks, and 18 months of age. Both genotypes lost dopaminergic neurons with age and remaining neurons at 11 months were resilient to paraquat/maneb. In 5-week-old mice, the toxins decreased substantia nigra pars compacta dopaminergic neurons in both genotypes but less in DAT-KD. Regional analysis revealed striking differences in the subsets of neurons affected by low DAT, paraquat/maneb, and aging. In particular, we show that a potentially protective effect of low DAT against toxin exposure is not sufficient to reduce death of all nigrostriatal dopaminergic neurons. Thus, different regional vulnerability of nigrostriatal dopaminergic neurons may contribute to an increased risk of developing Parkinson's disease when multiple factors are combined.
Collapse
Affiliation(s)
| | - Lauryn Gabby
- Department of Neurology, UCLA, Los Angeles, CA, USA
| | | | | | | | | | | | - Ingrid Cely
- Hatos Center, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA
| | - Larry C Ackerson
- Hatos Center, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA
| | - Linda Tsan
- Hatos Center, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA
| | - Niall P Murphy
- Hatos Center, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA
| | - Nigel T Maidment
- Hatos Center, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA
| | | |
Collapse
|
43
|
Peris J, MacFadyen K, Smith JA, de Kloet AD, Wang L, Krause EG. Oxytocin receptors are expressed on dopamine and glutamate neurons in the mouse ventral tegmental area that project to nucleus accumbens and other mesolimbic targets. J Comp Neurol 2016; 525:1094-1108. [PMID: 27615433 DOI: 10.1002/cne.24116] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/24/2016] [Accepted: 09/04/2016] [Indexed: 12/25/2022]
Abstract
The mesolimbic dopamine (DA) circuitry determines which behaviors are positively reinforcing and therefore should be encoded in the memory to become a part of the behavioral repertoire. Natural reinforcers, like food and sex, activate this pathway, thereby increasing the likelihood of further consummatory, social, and sexual behaviors. Oxytocin (OT) has been implicated in mediating natural reward and OT-synthesizing neurons project to the ventral tegmental area (VTA) and nucleus accumbens (NAc); however, direct neuroanatomical evidence of OT regulation of DA neurons within the VTA is sparse. To phenotype OT-receptor (OTR) expressing neurons originating within the VTA, we delivered Cre-inducible adeno-associated virus that drives the expression of fluorescent marker into the VTA of male mice that had Cre-recombinase driven by OTR gene expression. OTR-expressing VTA neurons project to NAc, prefrontal cortex, the extended amygdala, and other forebrain regions but less than 10% of these OTR-expressing neurons were identified as DA neurons (defined by tyrosine hydroxylase colocalization). Instead, almost 50% of OTR-expressing cells in the VTA were glutamate (GLU) neurons, as indicated by expression of mRNA for the vesicular GLU transporter (vGluT). About one-third of OTR-expressing VTA neurons did not colocalize with either DA or GLU phenotypic markers. Thus, OTR expression by VTA neurons implicates that OT regulation of reward circuitry is more complex than a direct action on DA neurotransmission. J. Comp. Neurol. 525:1094-1108, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Joanna Peris
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida, USA
| | - Kaley MacFadyen
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida, USA
| | - Justin A Smith
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida, USA
| | - Annette D de Kloet
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| | - Lei Wang
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida, USA
| | - Eric G Krause
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
44
|
Williams MJ, Perland E, Eriksson MM, Carlsson J, Erlandsson D, Laan L, Mahebali T, Potter E, Frediksson R, Benedict C, Schiöth HB. Recurrent Sleep Fragmentation Induces Insulin and Neuroprotective Mechanisms in Middle-Aged Flies. Front Aging Neurosci 2016; 8:180. [PMID: 27531979 PMCID: PMC4969361 DOI: 10.3389/fnagi.2016.00180] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/13/2016] [Indexed: 11/24/2022] Open
Abstract
Lack of quality sleep increases central nervous system oxidative stress and impairs removal of neurotoxic soluble metabolites from brain parenchyma. During aging poor sleep quality, caused by sleep fragmentation, increases central nervous system cellular stress. Currently, it is not known how organisms offset age-related cytotoxic metabolite increases in order to safeguard neuronal survival. Furthermore, it is not understood how age and sleep fragmentation interact to affect oxidative stress protection pathways. We demonstrate sleep fragmentation increases systems that protect against oxidative damage and neuroprotective endoplasmic reticulum molecular chaperones, as well as neuronal insulin and dopaminergic expression in middle-aged Drosophila males. Interestingly, even after sleep recovery the expression of these genes was still upregulated in middle-aged flies. Finally, sleep fragmentation generates higher levels of reactive oxygen species (ROS) in middle-aged flies and after sleep recovery these levels remain significantly higher than in young flies. The fact that neuroprotective pathways remain upregulated in middle-aged flies beyond sleep fragmentation suggests it might represent a strong stressor for the brain during later life.
Collapse
Affiliation(s)
- Michael J Williams
- Functional Pharmacology, Department of Neuroscience, Uppsala University Uppsala, Sweden
| | - Emelie Perland
- Functional Pharmacology, Department of Neuroscience, Uppsala University Uppsala, Sweden
| | - Mikaela M Eriksson
- Functional Pharmacology, Department of Neuroscience, Uppsala University Uppsala, Sweden
| | - Josef Carlsson
- Functional Pharmacology, Department of Neuroscience, Uppsala University Uppsala, Sweden
| | - Daniel Erlandsson
- Functional Pharmacology, Department of Neuroscience, Uppsala University Uppsala, Sweden
| | - Loora Laan
- Functional Pharmacology, Department of Neuroscience, Uppsala University Uppsala, Sweden
| | - Tabusi Mahebali
- Functional Pharmacology, Department of Neuroscience, Uppsala University Uppsala, Sweden
| | - Ella Potter
- Functional Pharmacology, Department of Neuroscience, Uppsala University Uppsala, Sweden
| | - Robert Frediksson
- Functional Pharmacology, Department of Neuroscience, Uppsala University Uppsala, Sweden
| | - Christian Benedict
- Functional Pharmacology, Department of Neuroscience, Uppsala University Uppsala, Sweden
| | - Helgi B Schiöth
- Functional Pharmacology, Department of Neuroscience, Uppsala University Uppsala, Sweden
| |
Collapse
|