1
|
Conrad WS, Oriol L, Kollman GJ, Faget L, Hnasko TS. Proportion and distribution of neurotransmitter-defined cell types in the ventral tegmental area and substantia nigra pars compacta. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582356. [PMID: 38464250 PMCID: PMC10925288 DOI: 10.1101/2024.02.28.582356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Most studies on the ventral tegmental area (VTA) and substantia nigra pars compacta (SNc) have focused on dopamine neurons and their role in processes such as motivation, learning, movement, and associated disorders such as addiction and Parkinson's disease. However there has been increasing attention on other VTA and SNc cell types that release GABA, glutamate, or a combination of neurotransmitters. Yet the relative distributions and proportions of neurotransmitter-defined cell types across VTA and SNc has remained unclear. Here, we used fluorescent in situ hybridization in male and female mice to label VTA and SNc neurons that expressed mRNA encoding the canonical vesicular transporters for dopamine, GABA, or glutamate: vesicular monoamine transporter (VMAT2), vesicular GABA transporter (VGAT), and vesicular glutamate transporter (VGLUT2). Within VTA, we found that no one type was particularly more abundant, instead we observed similar numbers of VMAT2+ (44%), VGAT+ (37%) and VGLUT2+ (41%) neurons. In SNc we found that a slight majority of neurons expressed VMAT2 (54%), fewer were VGAT+ (42%), and VGLUT2+ neurons were least abundant (16%). Moreover, 20% of VTA neurons and 10% of SNc neurons expressed more than one vesicular transporter, including 45% of VGLUT2+ neurons. We also assessed within VTA and SNc subregions and found remarkable heterogeneity in cell-type composition. And by quantifying density across both anterior-posterior and medial-lateral axes we generated heatmaps to visualize the distribution of each cell type. Our data complement recent single-cell RNAseq studies and support a more diverse landscape of neurotransmitter-defined cell types in VTA and SNc than is typically appreciated.
Collapse
|
2
|
Jordi L, Isacson O. Neuronal threshold functions: Determining symptom onset in neurological disorders. Prog Neurobiol 2024; 242:102673. [PMID: 39389338 DOI: 10.1016/j.pneurobio.2024.102673] [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: 05/22/2024] [Revised: 09/18/2024] [Accepted: 10/06/2024] [Indexed: 10/12/2024]
Abstract
Synaptic networks determine brain function. Highly complex interconnected brain synaptic networks provide output even under fluctuating or pathological conditions. Relevant to the treatment of brain disorders, understanding the limitations of such functional networks becomes paramount. Here we use the example of Parkinson's Disease (PD) as a system disorder, with PD symptomatology emerging only when the functional reserves of neurons, and their interconnected networks, are unable to facilitate effective compensatory mechanisms. We have denoted this the "threshold theory" to account for how PD symptoms develop in sequence. In this perspective, threshold functions are delineated in a quantitative, synaptic, and cellular network context. This provides a framework to discuss the development of specific symptoms. PD includes dysfunction and degeneration in many organ systems and both peripheral and central nervous system involvement. The threshold theory accounts for and explains the reasons why parallel gradually emerging pathologies in brain and peripheral systems generate specific symptoms only when functional thresholds are crossed, like tipping points. New and mounting evidence demonstrate that PD and related neurodegenerative diseases are multisystem disorders, which transcends the traditional brain-centric paradigm. We believe that representation of threshold functions will be helpful to develop new medicines and interventions that are specific for both pre- and post-symptomatic periods of neurodegenerative disorders.
Collapse
Affiliation(s)
- Luc Jordi
- Neuroregeneration Institute, McLean Hospital / Harvard Medical School, Belmont, MA 02478, USA.
| | - Ole Isacson
- Neuroregeneration Institute, McLean Hospital / Harvard Medical School, Belmont, MA 02478, USA; Department of Neurology and Program in Neuroscience, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Basso V, Döbrössy MD, Thompson LH, Kirik D, Fuller HR, Gates MA. State of the Art in Sub-Phenotyping Midbrain Dopamine Neurons. BIOLOGY 2024; 13:690. [PMID: 39336117 PMCID: PMC11428604 DOI: 10.3390/biology13090690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024]
Abstract
Dopaminergic neurons in the ventral tegmental area (VTA) and the substantia nigra pars compacta (SNpc) comprise around 75% of all dopaminergic neurons in the human brain. While both groups of dopaminergic neurons are in close proximity in the midbrain and partially overlap, development, function, and impairments in these two classes of neurons are highly diverse. The molecular and cellular mechanisms underlying these differences are not yet fully understood, but research over the past decade has highlighted the need to differentiate between these two classes of dopaminergic neurons during their development and in the mature brain. This differentiation is crucial not only for understanding fundamental circuitry formation in the brain but also for developing therapies targeted to specific dopaminergic neuron classes without affecting others. In this review, we summarize the state of the art in our understanding of the differences between the dopaminergic neurons of the VTA and the SNpc, such as anatomy, structure, morphology, output and input, electrophysiology, development, and disorders, and discuss the current technologies and methods available for studying these two classes of dopaminergic neurons, highlighting their advantages, limitations, and the necessary improvements required to achieve more-precise therapeutic interventions.
Collapse
Affiliation(s)
- Valentina Basso
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK
| | - Máté D Döbrössy
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional, Neurosurgery, Medical Center, University of Freiburg, 79106 Freiburg im Breisgau, Germany
- Department of Stereotactic and Functional Neurosurgery, Medical Center, University of Freiburg, 79106 Freiburg im Breisgau, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Lachlan H Thompson
- Charles Perkins Centre, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (B.R.A.I.N.S) Unit, Department of Experimental Medical Science, Lund University, BMC D11, 22184 Lund, Sweden
| | - Heidi R Fuller
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Monte A Gates
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK
| |
Collapse
|
4
|
Del Rey NLG, Hernández-Pinedo N, Carrillo M, Del Cerro M, Esteban-García N, Trigo-Damas I, Monje MHG, Lanciego JL, Cavada C, Obeso JA, Blesa J. Calbindin and Girk2/Aldh1a1 define resilient vs vulnerable dopaminergic neurons in a primate Parkinson's disease model. NPJ Parkinsons Dis 2024; 10:165. [PMID: 39223183 PMCID: PMC11369234 DOI: 10.1038/s41531-024-00777-0] [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: 04/17/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
The differential vulnerability of dopaminergic neurons of the substantia nigra pars compacta (SNc) is a critical and unresolved question in Parkinson´s disease. Studies in mice show diverse susceptibility of subpopulations of nigral dopaminergic neurons to various toxic agents. In the primate midbrain, the molecular phenotypes of dopaminergic neurons and their differential vulnerability are poorly characterized. We performed a detailed histological study to determine the anatomical distribution of different molecular phenotypes within identified midbrain neurons and their selective vulnerability in control and MPTP-treated monkeys. In the ventral tier of the SNc (nigrosome), neurons rich in Aldh1a1 and Girk2 are intermingled, whereas calbindin is the marker that best identifies the most resilient neurons located in the dorsal tier and ventral tegmental area, recapitulating the well-defined dorsoventral axis of susceptibility to degeneration of dopaminergic neurons. In particular, a loss of Aldh1a1+ neurons in the ventral SNc was observed in parallel to the progressive development of parkinsonism. Aldh1a1+ neurons were the main population of vulnerable dopaminergic nigrostriatal-projecting neurons, while Aldh1a1- neurons giving rise to nigropallidal projections remained relatively preserved. Moreover, bundles of entwined Aldh1a1+ dendrites with long trajectories extending towards the substantia nigra pars reticulata emerged from clusters of Aldh1a1+ neurons and colocalized with dense cannabinoid receptor 1 afferent fibers likely representing part of the striatonigral projection that is affected in human disorders, including Parkinson´s disease. In conclusion, vulnerable nigrostriatal-projecting neurons can be identified by using Aldh1a1 and Girk2. Further studies are needed to define the afferent/efferent projection patterns of these most vulnerable neurons.
Collapse
Affiliation(s)
- Natalia López-González Del Rey
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- PhD Program in Neuroscience Autónoma de Madrid University-Cajal Institute, Madrid, Spain
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Nagore Hernández-Pinedo
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | - Megan Carrillo
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - María Del Cerro
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | - Noelia Esteban-García
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- PhD Program in Neuroscience Autónoma de Madrid University-Cajal Institute, Madrid, Spain
| | - Inés Trigo-Damas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Madrid, Spain
| | - Mariana H G Monje
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
- Parkinson's Disease and Movement Disorders Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - José L Lanciego
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- CNS Gene Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Carmen Cavada
- PhD Program in Neuroscience Autónoma de Madrid University-Cajal Institute, Madrid, Spain
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Autónoma de Madrid University, Madrid, Spain
| | - José A Obeso
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain.
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.
| | - Javier Blesa
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain.
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain.
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Madrid, Spain.
| |
Collapse
|
5
|
Liu Q, Liu Z, Xie W, Li Y, Wang H, Zhang S, Wang W, Hao J, Geng D, Yang J, Wang L. Single-cell sequencing of the substantia nigra reveals microglial activation in a model of MPTP. Front Aging Neurosci 2024; 16:1390310. [PMID: 38952478 PMCID: PMC11215054 DOI: 10.3389/fnagi.2024.1390310] [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: 02/26/2024] [Accepted: 06/03/2024] [Indexed: 07/03/2024] Open
Abstract
Background N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a neurotoxin widely used to induce PD models, but the effect of MPTP on the cells and genes of PD has not been fully elucidated. Methods Single-nucleus RNA sequencing was performed in the Substantia Nigra (SN) of MPTP mice. UMAP analysis was used for the dimensionality reduction visualization of the SN in the MPTP mice. Known marker genes highly expressed genes in each cluster were used to annotate most clusters. Specific Differentially Expressed Genes (DEGs) and PD risk genes analysis were used to find MPTP-associated cells. GO, KEGG, PPI network, GSEA and CellChat analysis were used to reveal cell type-specific functional alterations and disruption of cell-cell communication networks. Subset reconstruction and pseudotime analysis were used to reveal the activation status of the cells, and to find the transcription factors with trajectory characterized. Results Initially, we observed specific DEGs and PD risk genes enrichment in microglia. Next, We obtained the functional phenotype changes in microglia and found that IGF, AGRN and PTN pathways were reduced in MPTP mice. Finally, we analyzed the activation state of microglia and revealed a pro-inflammatory trajectory characterized by transcription factors Nfe2l2 and Runx1. Conclusion Our work revealed alterations in microglia function, signaling pathways and key genes in the SN of MPTP mice.
Collapse
Affiliation(s)
- Qing Liu
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ziyu Liu
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wenmeng Xie
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yibo Li
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hongfang Wang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sanbing Zhang
- Department of Hand and Foot Surgery, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Wenyu Wang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiaxin Hao
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dandan Geng
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, Hebei, China
| | - Jing Yang
- Zhejiang Provincial Key Laboratory of Aging and Cancer Biology, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Lei Wang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Hand and Foot Surgery, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
6
|
Shi W, Zhang J, Huang S, Fan Q, Cao J, Zeng J, Wu L, Yang C. Next-Generation Sequencing-Based Spatial Transcriptomics: A Perspective from Barcoding Chemistry. JACS AU 2024; 4:1723-1743. [PMID: 38818076 PMCID: PMC11134576 DOI: 10.1021/jacsau.4c00118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 06/01/2024]
Abstract
Gene expression profiling of tissue cells with spatial context is in high demand to reveal cell types, locations, and intercellular or molecular interactions for physiological and pathological studies. With rapid advances in barcoding chemistry and sequencing chemistry, spatially resolved transcriptome (SRT) techniques have emerged to quantify spatial gene expression in tissue samples by correlating transcripts with their spatial locations using diverse strategies. These techniques provide both physical tissue structure and molecular characteristics and are poised to revolutionize many fields, such as developmental biology, neuroscience, oncology, and histopathology. In this context, this Perspective focuses on next-generation sequencing-based SRT methods, particularly highlighting spatial barcoding chemistry. It delves into optically manipulated spatial indexing methods and DNA array-barcoded spatial indexing methods by exploring current advances, challenges, and future development directions in this nascent field.
Collapse
Affiliation(s)
- Weixiong Shi
- Institute
of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry
and Nanomedicine, Renji Hospital, Shanghai
Jiao Tong University School of Medicine, Shanghai 200127, China
- The
MOE Key Laboratory of Spectrochemical Analysis & Instrumentation,
Discipline of Intelligent Instrument and Equipment, Department of
Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jing Zhang
- State
Key Laboratory of Cellular Stress Biology, School of Life Sciences,
Faculty of Medicine and Life Sciences, Xiamen
University, Xiamen 361102, China
| | - Shanqing Huang
- The
MOE Key Laboratory of Spectrochemical Analysis & Instrumentation,
Discipline of Intelligent Instrument and Equipment, Department of
Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Qian Fan
- Institute
of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry
and Nanomedicine, Renji Hospital, Shanghai
Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jiao Cao
- Institute
of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry
and Nanomedicine, Renji Hospital, Shanghai
Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jun Zeng
- Institute
of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry
and Nanomedicine, Renji Hospital, Shanghai
Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Lingling Wu
- Institute
of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry
and Nanomedicine, Renji Hospital, Shanghai
Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Chaoyong Yang
- Institute
of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry
and Nanomedicine, Renji Hospital, Shanghai
Jiao Tong University School of Medicine, Shanghai 200127, China
- The
MOE Key Laboratory of Spectrochemical Analysis & Instrumentation,
Discipline of Intelligent Instrument and Equipment, Department of
Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- State
Key Laboratory of Cellular Stress Biology, School of Life Sciences,
Faculty of Medicine and Life Sciences, Xiamen
University, Xiamen 361102, China
| |
Collapse
|
7
|
Kampmann M. Molecular and cellular mechanisms of selective vulnerability in neurodegenerative diseases. Nat Rev Neurosci 2024; 25:351-371. [PMID: 38575768 DOI: 10.1038/s41583-024-00806-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2024] [Indexed: 04/06/2024]
Abstract
The selective vulnerability of specific neuronal subtypes is a hallmark of neurodegenerative diseases. In this Review, I summarize our current understanding of the brain regions and cell types that are selectively vulnerable in different neurodegenerative diseases and describe the proposed underlying cell-autonomous and non-cell-autonomous mechanisms. I highlight how recent methodological innovations - including single-cell transcriptomics, CRISPR-based screens and human cell-based models of disease - are enabling new breakthroughs in our understanding of selective vulnerability. An understanding of the molecular mechanisms that determine selective vulnerability and resilience would shed light on the key processes that drive neurodegeneration and point to potential therapeutic strategies to protect vulnerable cell populations.
Collapse
Affiliation(s)
- Martin Kampmann
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
8
|
Prytkova I, Liu Y, Fernando M, Gameiro-Ros I, Popova D, Kamarajan C, Xuei X, Chorlian DB, Edenberg HJ, Tischfield JA, Porjesz B, Pang ZP, Hart RP, Goate A, Slesinger PA. Upregulated GIRK2 Counteracts Ethanol-Induced Changes in Excitability and Respiration in Human Neurons. J Neurosci 2024; 44:e0918232024. [PMID: 38350999 PMCID: PMC11026340 DOI: 10.1523/jneurosci.0918-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 02/17/2024] Open
Abstract
Genome-wide association studies (GWAS) of electroencephalographic endophenotypes for alcohol use disorder (AUD) has identified noncoding polymorphisms within the KCNJ6 gene. KCNJ6 encodes GIRK2, a subunit of a G-protein-coupled inwardly rectifying potassium channel that regulates neuronal excitability. We studied the effect of upregulating KCNJ6 using an isogenic approach with human glutamatergic neurons derived from induced pluripotent stem cells (male and female donors). Using multielectrode arrays, population calcium imaging, single-cell patch-clamp electrophysiology, and mitochondrial stress tests, we find that elevated GIRK2 acts in concert with 7-21 d of ethanol exposure to inhibit neuronal activity, to counteract ethanol-induced increases in glutamate response, and to promote an increase intrinsic excitability. Furthermore, elevated GIRK2 prevented ethanol-induced changes in basal and activity-dependent mitochondrial respiration. These data support a role for GIRK2 in mitigating the effects of ethanol and a previously unknown connection to mitochondrial function in human glutamatergic neurons.
Collapse
Affiliation(s)
- Iya Prytkova
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Yiyuan Liu
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Michael Fernando
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Isabel Gameiro-Ros
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Dina Popova
- Human Genetics Institute, Rutgers University, Piscataway, New Jersey 08854
| | - Chella Kamarajan
- Department of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York 11203
| | - Xiaoling Xuei
- Departments of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - David B Chorlian
- Department of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York 11203
| | - Howard J Edenberg
- Departments of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Jay A Tischfield
- Human Genetics Institute, Rutgers University, Piscataway, New Jersey 08854
| | - Bernice Porjesz
- Department of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York 11203
| | - Zhiping P Pang
- Human Genetics Institute, Rutgers University, Piscataway, New Jersey 08854
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901
| | - Ronald P Hart
- Human Genetics Institute, Rutgers University, Piscataway, New Jersey 08854
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, New Jersey 08854
| | - Alison Goate
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Paul A Slesinger
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| |
Collapse
|
9
|
Prytkova I, Liu Y, Fernando M, Gameiro-Ros I, Popova D, Kamarajan C, Xuei X, Chorlian DB, Edenberg HJ, Tischfield JA, Porjesz B, Pang ZP, Hart RP, Goate A, Slesinger PA. Upregulated GIRK2 counteracts ethanol-induced changes in excitability & respiration in human neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.22.533236. [PMID: 36993693 PMCID: PMC10055374 DOI: 10.1101/2023.03.22.533236] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Genome-wide association analysis (GWAS) of electroencephalographic endophenotypes for alcohol use disorder (AUD) has identified non-coding polymorphisms within the KCNJ6 gene. KCNJ6 encodes GIRK2, a subunit of a G protein-coupled inwardly-rectifying potassium channel that regulates neuronal excitability. How changes in GIRK2 affect human neuronal excitability and the response to repeated ethanol exposure is poorly understood. Here, we studied the effect of upregulating KCNJ6 using an isogenic approach with human glutamatergic neurons derived from induced pluripotent stem cells (male and female donors). Using multi-electrode-arrays, population calcium imaging, single-cell patch-clamp electrophysiology, and mitochondrial stress tests, we find that elevated GIRK2 acts in concert with 7-21 days of ethanol exposure to inhibit neuronal activity, to counteract ethanol-induced increases in glutamate response, and to promote an increase intrinsic excitability. Furthermore, elevated GIRK2 prevented ethanol-dependent changes in basal and activity-dependent mitochondrial respiration. These data support a role for GIRK2 in mitigating the effects of ethanol and a previously unknown connection to mitochondrial function in human glutamatergic neurons. SIGNIFICANCE STATEMENT Alcohol use disorder (AUD) is a major health problem that has worsened since COVID, affecting over 100 million people worldwide. While it is known that heritability contributes to AUD, specific genes and their role in neuronal function remain poorly understood, especially in humans. In the current manuscript, we focused on the inwardly-rectifying potassium channel GIRK2, which has been identified in an AUD-endophenotype genome-wide association study. We used human excitatory neurons derived from healthy donors to study the impact of GIRK2 expression. Our results reveal that elevated GIRK2 counteracts ethanol-induced increases in glutamate response and intracellular calcium, as well as deficits in activity-dependent mitochondrial respiration. The role of GIRK2 in mitigating ethanol-induced hyper-glutamatergic and mitochondrial offers therapeutic promise for treating AUD.
Collapse
|
10
|
Liu H, Gong Z, Li Z, Ye T, Cao A, He S, Lin S, Duan J, Lin X. Distribution, connection and function of ALDH1A1 +/TH + neurons in substantia nigra pars reticulata of mouse. Neurosci Lett 2024; 818:137555. [PMID: 37972684 DOI: 10.1016/j.neulet.2023.137555] [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: 09/18/2023] [Revised: 10/25/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023]
Abstract
The massive cell death of dopaminergic neurons (DANs) in substantia nigra pars compacta (SNC) is associated with motor diseases, such as Parkinson's disease. Moreover, as a subtype of DANs in SNC, ALDH1A1+ neurons show better resistance to PD related neurotoxin. DANs can also be found in the substantia nigra pars reticulata (SNR), however, whether they are ALDH1A1+ neurons are rarely reported, as well as their projection, function, and reaction in the PD pathology. We studied the distribution of ALDH1A1+ neurons and track their projection by injecting pAAV. We figured out that, in SNR, 87 % neurons are ALDH1A1+/TH+ in ALDH1A1+ cluster averagely, while ALDH1A1+/TH+: TH+ is 52 % averagely. There are two enrichment regions of ALDH1A1+/TH+ neurons at brgma -3.40 mm and brgma -3.70 mm in the SNR of the nTg mice. Nevertheless, in one type of PD-liked mice model, the proportion of ALDH1A1+/TH+: ALDH1A1+ neurons are 98 % averagely, while ALHD1A1+/TH+: TH+ is 57 %. Intriguingly, neuro-tracing discovered that there may be a previously unreported connection between SNR and anterior dorsal thalamus (ADT). The mouse received MPTP stereotactic injection to destroy TH+ neurons in SNR showed depression behavior, indicated the DANs death in SNR may contribute to depression behavior.
Collapse
Affiliation(s)
- Hao Liu
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China
| | - Zhuo Gong
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China
| | - Zhao Li
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China
| | - Tonglin Ye
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China
| | - Anqi Cao
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China
| | - Shuaiying He
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China
| | - Sijia Lin
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China
| | - Jinhai Duan
- Eastern Department of Neurology, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Xian Lin
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China; Department of Human Anatomy and physiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan 2(nd) Road, Guangzhou 510080, Guangdong, China.
| |
Collapse
|
11
|
Ye P, Fang Q, Hu X, Zou W, Huang M, Ke M, Li Y, Liu M, Cai X, Zhang C, Hua N, Al-Sheikh U, Liu X, Yu P, Jiang P, Pan PY, Luo J, Jiang LH, Xu S, Fang EF, Su H, Kang L, Yang W. TRPM2 as a conserved gatekeeper determines the vulnerability of DA neurons by mediating ROS sensing and calcium dyshomeostasis. Prog Neurobiol 2023; 231:102530. [PMID: 37739206 DOI: 10.1016/j.pneurobio.2023.102530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/01/2023] [Accepted: 09/17/2023] [Indexed: 09/24/2023]
Abstract
Different dopaminergic (DA) neuronal subgroups exhibit distinct vulnerability to stress, while the underlying mechanisms are elusive. Here we report that the transient receptor potential melastatin 2 (TRPM2) channel is preferentially expressed in vulnerable DA neuronal subgroups, which correlates positively with aging in Parkinson's Disease (PD) patients. Overexpression of human TRPM2 in the DA neurons of C. elegans resulted in selective death of ADE but not CEP neurons in aged worms. Mechanistically, TRPM2 activation mediates FZO-1/CED-9-dependent mitochondrial hyperfusion and mitochondrial permeability transition (MPT), leading to ADE death. In mice, TRPM2 knockout reduced vulnerable substantia nigra pars compacta (SNc) DA neuronal death induced by stress. Moreover, the TRPM2-mediated vulnerable DA neuronal death pathway is conserved from C. elegans to toxin-treated mice model and PD patient iPSC-derived DA neurons. The vulnerable SNc DA neuronal loss is the major symptom and cause of PD, and therefore the TRPM2-mediated pathway serves as a promising therapeutic target against PD.
Collapse
Affiliation(s)
- Peiwu Ye
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qiuyuan Fang
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xupang Hu
- Second Clinical Medical College, Affiliated Secondary Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310011, China
| | - Wenjuan Zou
- Department of Neurobiology and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang 310053, China
| | - Miaodan Huang
- Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Minjing Ke
- Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yunhao Li
- Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Min Liu
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaobo Cai
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Congyi Zhang
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ning Hua
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Umar Al-Sheikh
- Department of Neurobiology and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang 310053, China
| | - Xingyu Liu
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Peilin Yu
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Peiran Jiang
- School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ping-Yue Pan
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane, Piscataway, NJ 08854, USA
| | - Jianhong Luo
- School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lin-Hua Jiang
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK; Sino-UK Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang 453000, China; University of Leeds, Leeds LS2 9JT, UK
| | - Suhong Xu
- Center for Stem Cell and Regenerative Medicine and Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Evandro F Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Huanxing Su
- Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| | - Lijun Kang
- Second Clinical Medical College, Affiliated Secondary Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310011, China; School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Wei Yang
- Department of Biophysics, Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
12
|
Kreutz A, Hu G, Tokar E. Pluripotent Stem Cell-derived Dopaminergic Neurons for Studying Developmental Neurotoxicity. Stem Cell Rev Rep 2023; 19:2120-2130. [PMID: 37273171 PMCID: PMC10579108 DOI: 10.1007/s12015-023-10555-9] [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] [Accepted: 05/08/2023] [Indexed: 06/06/2023]
Abstract
With the vast number of chemicals in commerce, higher throughput strategies are needed to inform risk assessment. The field of toxicology is therefore moving away from traditional in vivo guideline studies towards in vitro new approach methodologies. There has been a great push for such a shift in the field of developmental neurotoxicity, where there is a particular lack of data. A battery of in vitro new approach methodologies has thus been developed to help fill this gap. Included in this battery are assays for numerous processes critical to neurodevelopment, such as proliferation, migration, and synaptogenesis. The current battery of developmental neurotoxicity new approach methodologies still lacks recapitulation of several critical neurodevelopmental processes, including development of neuronal subtypes. With their pluripotency, alongside other advantages, pluripotent stem cells (PSCs) are uniquely suited to address questions of developmental neurotoxicity, as they can recapitulate the different stages of human in vivo neurodevelopment. Among the various neuronal subtypes, development of dopaminergic neurons (DA) is perhaps the best understood and several approaches exist to differentiate PSCs into DA. Herein we review these approaches and propose utilizing PSCs for screening of the impact of environmental chemicals on development of DA. Related techniques and gaps in knowledge are also addressed.
Collapse
Affiliation(s)
- Anna Kreutz
- Mechanistic Toxicology Branch, Division of Translational Toxicology, NIEHS, Research Triangle Park, Durham, NC, 27709, USA.
- Epigenetics and Stem Cell Biology Laboratory, Division of Intramural Research, NIEHS, Research Triangle Park, Durham, NC, 27709, USA.
| | - Guang Hu
- Epigenetics and Stem Cell Biology Laboratory, Division of Intramural Research, NIEHS, Research Triangle Park, Durham, NC, 27709, USA
| | - Erik Tokar
- Mechanistic Toxicology Branch, Division of Translational Toxicology, NIEHS, Research Triangle Park, Durham, NC, 27709, USA
| |
Collapse
|
13
|
Kato T, Nishimura K, Hirao M, Shimohama S, Takata K. Expression and role of nicotinic acetylcholine receptors during midbrain dopaminergic neuron differentiation from human induced pluripotent stem cells. Neuropsychopharmacol Rep 2023; 43:440-445. [PMID: 37366076 PMCID: PMC10496050 DOI: 10.1002/npr2.12361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/28/2023] Open
Abstract
AIM Nicotinic acetylcholine receptors (nAChRs) expressed in midbrain dopaminergic (mDA) neurons modulate mDA neuronal activity. However, their expression patterns and functional roles during mDA neuronal development remain unknown. Here, we profiled the expression and function of nAChR subtypes during mDA neuron differentiation from human induced pluripotent stem cells (hiPSCs). METHODS Midbrain dopaminergic neurons were differentiated from hiPSCs using a recently developed proprietary method that replicates midbrain development. The expression patterns of developmental marker proteins were monitored during mDA neuronal differentiation using immunohistochemical analysis. Gene expression of nAChR subtypes was analyzed by reverse transcription polymerase chain reaction. Pharmacological nAChR agonists and antagonists were used to reveal the role of the α6 nAChR subunit in the differentiation of mDA neurons from hiPSCs. RESULTS CHRNA4 expression was detected at the mDA neural progenitor stage, whereas CHRNA6 expression began during the mDA neuronal stage. CHRNA7 was expressed throughout the differentiation process, including in the undifferentiated hiPSCs. We also found that LMO3, a gene expressed in a subset of substantia nigra pars compacta (SNC) DA neurons in the midbrain, showed increased expression following nicotine treatment in a concentration-dependent manner. Additionally, 5-iodo A85380, a selective α6 nAChR agonist, also increased LMO3 expression in hiPSC-derived mDA neurons, and this increase was suppressed by simultaneous treatment with bPiDi, a selective α6 nAChR antagonist. CONCLUSION Our findings suggest that stimulating the α6 nAChR subunit on hiPSC-derived mDA neurons may induce neuronal maturation that is biased toward SNC DA neurons.
Collapse
Affiliation(s)
- Takeshi Kato
- Joint Research Laboratory, Division of Integrated Pharmaceutical ScienceKyoto Pharmaceutical UniversityKyotoJapan
| | - Kaneyasu Nishimura
- Joint Research Laboratory, Division of Integrated Pharmaceutical ScienceKyoto Pharmaceutical UniversityKyotoJapan
- Laboratory of Functional Brain Circuit Construction, Graduate School of Brain ScienceDoshisha UniversityKyotanabeJapan
| | - Masahiro Hirao
- Joint Research Laboratory, Division of Integrated Pharmaceutical ScienceKyoto Pharmaceutical UniversityKyotoJapan
| | - Shun Shimohama
- Department of Neurology, School of MedicineSapporo Medical UniversitySapporoJapan
- Jiseikai Dementia CenterItabashiJapan
- Jiseikai Nerima Takanodai HospitalNerimaJapan
| | - Kazuyuki Takata
- Joint Research Laboratory, Division of Integrated Pharmaceutical ScienceKyoto Pharmaceutical UniversityKyotoJapan
| |
Collapse
|
14
|
Fehér M, Márton Z, Szabó Á, Kocsa J, Kormos V, Hunyady Á, Kovács LÁ, Ujvári B, Berta G, Farkas J, Füredi N, Gaszner T, Pytel B, Reglődi D, Gaszner B. Downregulation of PACAP and the PAC1 Receptor in the Basal Ganglia, Substantia Nigra and Centrally Projecting Edinger-Westphal Nucleus in the Rotenone model of Parkinson's Disease. Int J Mol Sci 2023; 24:11843. [PMID: 37511603 PMCID: PMC10380602 DOI: 10.3390/ijms241411843] [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: 06/25/2023] [Revised: 07/19/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023] Open
Abstract
Numerous in vitro and in vivo models of Parkinson's disease (PD) demonstrate that pituitary adenylate cyclase-activating polypeptide (PACAP) conveys its strong neuroprotective actions mainly via its specific PAC1 receptor (PAC1R) in models of PD. We recently described the decrease in PAC1R protein content in the basal ganglia of macaques in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD that was partially reversed by levodopa therapy. In this work, we tested whether these observations occur also in the rotenone model of PD in the rat. The rotarod test revealed motor skill deterioration upon rotenone administration, which was reversed by benserazide/levodopa (B/L) treatment. The sucrose preference test suggested increased depression level while the open field test showed increased anxiety in rats rendered parkinsonian, regardless of the received B/L therapy. Reduced dopaminergic cell count in the substantia nigra pars compacta (SNpc) diminished the dopaminergic fiber density in the caudate-putamen (CPu) and decreased the peptidergic cell count in the centrally projecting Edinger-Westphal nucleus (EWcp), supporting the efficacy of rotenone treatment. RNAscope in situ hybridization revealed decreased PACAP mRNA (Adcyap1) and PAC1R mRNA (Adcyap1r1) expression in the CPu, globus pallidus, dopaminergic SNpc and peptidergic EWcp of rotenone-treated rats, but no remarkable downregulation occurred in the insular cortex. In the entopeduncular nucleus, only the Adcyap1r1 mRNA was downregulated in parkinsonian animals. B/L therapy attenuated the downregulation of Adcyap1 in the CPu only. Our current results further support the evolutionarily conserved role of the PACAP/PAC1R system in neuroprotection and its recruitment in the development/progression of neurodegenerative states such as PD.
Collapse
Affiliation(s)
- Máté Fehér
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Department of Neurosurgery, Kaposi Mór Teaching Hospital, Tallián Gy. u. 20-32, H-7400 Kaposvár, Hungary
| | - Zsombor Márton
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Ákos Szabó
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - János Kocsa
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Viktória Kormos
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Ágnes Hunyady
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - László Ákos Kovács
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Balázs Ujvári
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscopic Laboratory, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - József Farkas
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Nóra Füredi
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Tamás Gaszner
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Bence Pytel
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Dóra Reglődi
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- ELKH-PTE PACAP Research Group, Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Balázs Gaszner
- Department of Anatomy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Research Group for Mood Disorders, Centre for Neuroscience, University Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| |
Collapse
|
15
|
Hobson BD, Stanley AT, De Los Santos MB, Culbertson B, Mosharov EV, Sims PA, Sulzer D. Conserved and cell type-specific transcriptional responses to IFN-γ in the ventral midbrain. Brain Behav Immun 2023; 111:277-291. [PMID: 37100211 PMCID: PMC10460506 DOI: 10.1016/j.bbi.2023.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/28/2023] [Accepted: 04/23/2023] [Indexed: 04/28/2023] Open
Abstract
Dysregulated inflammation within the central nervous system (CNS) contributes to neuropathology in infectious, autoimmune, and neurodegenerative disease. With the exception of microglia, major histocompatibility complex (MHC) proteins are virtually undetectable in the mature, healthy central nervous system (CNS). Neurons have generally been considered incapable of antigen presentation, and although interferon gamma (IFN-γ) can elicit neuronal MHC class I (MHC-I) expression and antigen presentation in vitro, it has been unclear whether similar responses occur in vivo. Here we directly injected IFN-γ into the ventral midbrain of mature mice and analyzed gene expression profiles of specific CNS cell types. We found that IFN-γ upregulated MHC-I and associated mRNAs in ventral midbrain microglia, astrocytes, oligodendrocytes, and GABAergic, glutamatergic, and dopaminergic neurons. The core set of IFN-γ-induced genes and their response kinetics were similar in neurons and glia, but with a lower amplitude of expression in neurons. A diverse repertoire of genes was upregulated in glia, particularly microglia, which were the only cells to undergo cellular proliferation and express MHC classII (MHC-II) and associated genes. To determine if neurons respond directly via cell-autonomous IFN-γ receptor (IFNGR) signaling, we produced mutant mice with a deletion of the IFN-γ-binding domain of IFNGR1 in dopaminergic neurons, which resulted in a complete loss of dopaminergic neuronal responses to IFN-γ. Our results demonstrate that IFN-γ induces neuronal IFNGR signaling and upregulation of MHC-I and related genes in vivo, although the expression level is low compared to oligodendrocytes, astrocytes, and microglia.
Collapse
Affiliation(s)
- Benjamin D Hobson
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, United States; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States
| | - Adrien T Stanley
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States
| | - Mark B De Los Santos
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States
| | - Bruce Culbertson
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, United States; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Eugene V Mosharov
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, United States; Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY 10032, United States; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States.
| | - David Sulzer
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Pharmacology, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States.
| |
Collapse
|
16
|
Kamath T, Macosko EZ. Insights into Neurodegeneration in Parkinson's Disease from Single-Cell and Spatial Genomics. Mov Disord 2023; 38:518-525. [PMID: 36881930 PMCID: PMC11056908 DOI: 10.1002/mds.29374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/12/2023] [Accepted: 02/16/2023] [Indexed: 03/09/2023] Open
Abstract
Parkinson's disease (PD) is pathologically defined by the death of dopaminergic (DA) neurons within the pars compacta of the substantia nigra. To date, the cause of this multifaceted disease remains largely unclear, which may contribute in part to a current lack of disease-modifying therapies. Recent advances in single-cell and spatial genomic profiling tools have provided powerful new ways to measure cellular state changes in brain diseases. Here, we describe how these tools have offered insight into these complex disorders and highlight a recently performed comprehensive study of DA neuron susceptibility in PD. The data generated by this recent work provide evidence for the role of specific pathways and common genetic variants resulting in the loss of a critical DA subtype in PD. We conclude by outlining a set of basic and translational opportunities that arise from those data and insights gathered from this work. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Tushar Kamath
- Stanley Center for Psychiatric Research, Broad Institute, 75 Ames Street Cambridge, MA 02139
- Harvard Medical School and Harvard/MIT MD-PhD Program, Harvard University, Cambridge, MA 02139 USA
| | - Evan Z. Macosko
- Stanley Center for Psychiatric Research, Broad Institute, 75 Ames Street Cambridge, MA 02139
- Massachusetts General Hospital, Department of Psychiatry, Boston, MA USA
| |
Collapse
|
17
|
Abstract
The midbrain dopamine (mDA) system is composed of molecularly and functionally distinct neuron subtypes that mediate specific behaviours and are linked to various brain diseases. Considerable progress has been made in identifying mDA neuron subtypes, and recent work has begun to unveil how these neuronal subtypes develop and organize into functional brain structures. This progress is important for further understanding the disparate physiological functions of mDA neurons and their selective vulnerability in disease, and will ultimately accelerate therapy development. This Review discusses recent advances in our understanding of molecularly defined mDA neuron subtypes and their circuits, ranging from early developmental events, such as neuron migration and axon guidance, to their wiring and function, and future implications for therapeutic strategies.
Collapse
|
18
|
Bimpisidis Z, Serra GP, König N, Wallén-Mackenzie Å. Increased sucrose consumption in mice gene-targeted for Vmat2 selectively in NeuroD6-positive neurons of the ventral tegmental area. Front Mol Neurosci 2023; 16:1069834. [PMID: 36825278 PMCID: PMC9941196 DOI: 10.3389/fnmol.2023.1069834] [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: 10/14/2022] [Accepted: 01/11/2023] [Indexed: 02/10/2023] Open
Abstract
Ventral tegmental area (VTA) dopamine (DA) neurons are implicated in reward processing, motivation, reward prediction error, and in substance use disorder. Recent studies have identified distinct neuronal subpopulations within the VTA that can be clustered based on their molecular identity, neurotransmitter profile, physiology, projections and behavioral role. One such subpopulation is characterized by expression of the NeuroD6 gene, and projects primarily to the nucleus accumbens medial shell. We recently showed that optogenetic stimulation of these neurons induces real-time place preference while their targeted deletion of the Vmat2 gene caused altered response to rewarding substances, including ethanol and psychostimulants. Based on these recent findings, we wanted to further investigate the involvement of the NeuroD6-positive VTA subpopulation in reward processing. Using the same NeuroD6Cre+/wt ;Vmat2flox/flox mice as in our prior study, we now addressed the ability of the mice to process sucrose reward. In order to assess appetitive behavior and motivation to obtain sucrose reward, we tested conditional knockout (cKO) and control littermate mice in an operant sucrose self-administration paradigm. We observed that cKO mice demonstrate higher response rates to the operant task and consume more sucrose rewards than control mice. However, their motivation to obtain sucrose is identical to that of control mice. Our results highlight previous observations that appetitive behavior and motivation to obtain rewards can be served by distinct neuronal circuits, and demonstrate that the NeuroD6 VTA subpopulation is involved in mediating the former, but not the latter. Together with previous studies on the NeuroD6 subpopulation, our findings pinpoint the importance of unraveling the molecular and functional role of VTA subpopulations in order to better understand normal behavior and psychiatric disease.
Collapse
Affiliation(s)
| | - Gian Pietro Serra
- Unit of Comparative Physiology, Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| | - Niclas König
- Unit of Comparative Physiology, Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| | - Åsa Wallén-Mackenzie
- Unit of Comparative Physiology, Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
19
|
Jia E, Sheng Y, Shi H, Wang Y, Zhou Y, Liu Z, Qi T, Pan M, Bai Y, Zhao X, Ge Q. Spatial Transcriptome Profiling of Mouse Hippocampal Single Cell Microzone in Parkinson's Disease. Int J Mol Sci 2023; 24:ijms24031810. [PMID: 36768134 PMCID: PMC9915078 DOI: 10.3390/ijms24031810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/02/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
The hippocampus is an important part of the limbic system in the human brain that has essential roles in spatial navigation and cognitive functions. It is still unknown how gene expression changes in single-cell in different spatial locations of the hippocampus of Parkinson's disease. The purpose of this study was to analyze the gene expression features of single cells in different spatial locations of mouse hippocampus, and to explore the effects of gene expression regulation on learning and memory mechanisms. Here, we obtained 74 single-cell samples from different spatial locations in a mouse hippocampus through microdissection technology, and used single-cell RNA-sequencing and spatial transcriptome sequencing to visualize and quantify the single-cell transcriptome features of tissue sections. The results of differential expression analysis showed that the expression of Sv2b, Neurod6, Grp and Stk32b genes in a hippocampus single cell at different locations was significantly different, and the marker genes of CA1, CA3 and DG subregions were identified. The results of gene function enrichment analysis showed that the up-regulated differentially expressed genes Tubb2a, Eno1, Atp2b1, Plk2, Map4, Pex5l, Fibcd1 and Pdzd2 were mainly involved in neuron to neuron synapse, vesicle-mediated transport in synapse, calcium signaling pathway and neurodegenerative disease pathways, thus affecting learning and memory function. It revealed the transcriptome profile and heterogeneity of spatially located cells in the hippocampus of PD for the first time, and demonstrated that the impaired learning and memory ability of PD was affected by the synergistic effect of CA1 and CA3 subregions neuron genes. These results are crucial for understanding the pathological mechanism of the Parkinson's disease and making precise treatment plans.
Collapse
Affiliation(s)
- Erteng Jia
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
- Thoracic Surgery Laboratory, The First College of Clinical Medicine, Xuzhou Medical University, Xuzhou 221006, China
| | - Yuqi Sheng
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Huajuan Shi
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Ying Wang
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Ying Zhou
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhiyu Liu
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Ting Qi
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Min Pan
- School of Medicine, Southeast University, Nanjing 210097, China
| | - Yunfei Bai
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xiangwei Zhao
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
- Correspondence: (X.Z.); (Q.G.); Tel./Fax: +86-025-8379-2396 (Q.G.)
| | - Qinyu Ge
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
- Correspondence: (X.Z.); (Q.G.); Tel./Fax: +86-025-8379-2396 (Q.G.)
| |
Collapse
|
20
|
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
|
21
|
Martins-Oliveira M, Akerman S, Holland PR, Tavares I, Goadsby PJ. Pharmacological modulation of ventral tegmental area neurons elicits changes in trigeminovascular sensory processing and is accompanied by glycemic changes: Implications for migraine. Cephalalgia 2022; 42:1359-1374. [PMID: 36259130 DOI: 10.1177/03331024221110111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Imaging migraine premonitory studies show increased midbrain activation consistent with the ventral tegmental area, an area involved in pain modulation and hedonic feeding. We investigated ventral tegmental area pharmacological modulation effects on trigeminovascular processing and consequent glycemic levels, which could be involved in appetite changes in susceptible migraine patients. METHODS Serotonin and pituitary adenylate cyclase-activating polypeptide receptors immunohistochemistry was performed in ventral tegmental area parabrachial pigmented nucleus of male Sprague Dawley rats. In vivo trigeminocervical complex neuronal responses to dura mater nociceptive electrical stimulation, and facial mechanical stimulation of the ophthalmic dermatome were recorded. Changes in trigeminocervical complex responses following ventral tegmental area parabrachial pigmented nucleus microinjection of glutamate, bicuculline, naratriptan, pituitary adenylate cyclase-activating polypeptide-38 and quinpirole were measured, and blood glucose levels assessed pre- and post-microinjection. RESULTS Glutamatergic stimulation of ventral tegmental area parabrachial pigmented nucleus neurons reduced nociceptive and spontaneous trigeminocervical complex neuronal firing. Naratriptan, pituitary adenylate cyclase-activating polypeptide-38 and quinpirole inhibited trigeminovascular spontaneous activity, and trigeminocervical complex neuronal responses to dural-evoked electrical and mechanical noxious stimulation. Trigeminovascular sensory processing through modulation of the ventral tegmental area parabrachial pigmented nucleus resulted in reduced circulating glucose levels. CONCLUSION Pharmacological modulation of ventral tegmental area parabrachial pigmented nucleus neurons elicits changes in trigeminovascular sensory processing. The interplay between ventral tegmental area parabrachial pigmented nucleus activity and the sensory processing by the trigeminovascular system may be relevant to understand associated sensory and homeostatic symptoms in susceptible migraine patients.
Collapse
Affiliation(s)
- Margarida Martins-Oliveira
- Headache Group, Wolfson Centre for Age-Related Disease, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK.,Department of Nutrition and Metabolism, NOVA Medical School
- Faculdade de Ciências Médicas, NMS
- FCM, Universidade Nova de Lisboa; Lisboa, Portugal.,Department of Biomedicine, Faculty of Medicine of University of Porto, Porto, Portugal.,Institute of Investigation and Innovation in Health (i3S), University of Porto, Porto, Portugal
| | - Simon Akerman
- Department of Neural and Pain Sciences, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Philip R Holland
- Headache Group, Wolfson Centre for Age-Related Disease, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Isaura Tavares
- Department of Biomedicine, Faculty of Medicine of University of Porto, Porto, Portugal.,Institute of Investigation and Innovation in Health (i3S), University of Porto, Porto, Portugal
| | - Peter J Goadsby
- Headache Group, Wolfson Centre for Age-Related Disease, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK.,Department of Neurology, University of California, Los Angeles, Los Angeles CA USA
| |
Collapse
|
22
|
Zheng R, Yan Y, Pu J, Zhang B. Physiological and Pathological Functions of Neuronal Hemoglobin: A Key Underappreciated Protein in Parkinson's Disease. Int J Mol Sci 2022; 23:9088. [PMID: 36012351 PMCID: PMC9408843 DOI: 10.3390/ijms23169088] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
The expression of Hemoglobin (Hb) is not restricted to erythrocytes but is also present in neurons. Hb is selectively enriched in vulnerable mesencephalic dopaminergic neurons of Parkinson's disease (PD) instead of resistant neurons. Controversial results of neuronal Hb levels have been reported in postmortem brains of PD patients: although neuronal Hb levels may decline in PD patients, elderly men with higher Hb levels have an increased risk of developing PD. α-synuclein, a key protein involved in PD pathology, interacts directly with Hb protein and forms complexes in erythrocytes and brains of monkeys and humans. These complexes increase in erythrocytes and striatal cytoplasm, while they decrease in striatal mitochondria with aging. Besides, the colocalization of serine 129-phosphorylated (Pser129) α-synuclein and Hb β chains have been found in the brains of PD patients. Several underlying molecular mechanisms involving mitochondrial homeostasis, α-synuclein accumulation, iron metabolism, and hormone-regulated signaling pathways have been investigated to assess the relationship between neuronal Hb and PD development. The formation of fibrils with neuronal Hb in various neurodegenerative diseases may indicate a common fibrillization pathway and a widespread target that could be applied in neurodegeneration therapy.
Collapse
Affiliation(s)
| | | | - Jiali Pu
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Baorong Zhang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| |
Collapse
|
23
|
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: 1.5] [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
|
24
|
Mitra S, Basu S, Singh O, Srivastava A, Singru PS. Calcium-binding proteins typify the dopaminergic neuronal subtypes in the ventral tegmental area of zebra finch, Taeniopygia guttata. J Comp Neurol 2022; 530:2562-2586. [PMID: 35715989 DOI: 10.1002/cne.25352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 11/11/2022]
Abstract
Calcium-binding proteins (CBPs) regulate neuronal function in midbrain dopamine (DA)-ergic neurons in mammals by buffering and sensing the intracellular Ca2+ , and vesicular release. In birds, the equivalent set of neurons are important in song learning, directed singing, courtship, and energy balance, yet the status of CBPs in these neurons is unknown. Herein, for the first time, we probe the nature of CBPs, namely, Calbindin-, Calretinin-, Parvalbumin-, and Secretagogin-expressing DA neurons in the ventral tegmental area (VTA) and substantia nigra (SN) in the midbrain of zebra finch, Taeniopygia guttata. qRT-PCR analysis of ventral midbrain tissue fragment revealed higher Calbindin- and Calretinin-mRNA levels compared to Parvalbumin and Secretagogin. Application of immunofluorescence showed CBP-immunoreactive (-i) neurons in VTA (anterior [VTAa], mid [VTAm], caudal [VTAc]), SN (compacta [SNc], and reticulata [SNr]). Compared to VTAa, higher Calbindin- and Parvalbumin-immunoreactivity (-ir), and lower Calretinin-ir were observed in VTAm and VTAc. Secretagogin-ir was highly localized to VTAa. In SN, Calbindin- and Calretinin-ir were higher in SNc, SNr was Parvalbumin enriched, and Secretagogin-ir was not detected. Weak, moderate, and intense tyrosine hydroxylase (TH)-i VTA neurons were demarcated as subtypes 1, 2, and 3, respectively. While subtype 1 TH-i neurons were neither Calbindin- nor Calretinin-i, ∼80 and ∼65% subtype 2 and ∼30 and ∼45% subtype 3 TH-i neurons co-expressed Calbindin and Calretinin, respectively. All TH-i neuronal subtypes co-expressed Parvalbumin with reciprocal relationship with TH-ir. We suggest that the CBPs may determine VTA DA neuronal heterogeneity and differentially regulate their activity in T. guttata.
Collapse
Affiliation(s)
- Saptarsi Mitra
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sumela Basu
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Omprakash Singh
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Abhinav Srivastava
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Praful S Singru
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
25
|
Horvath G, Reglodi D, Fabian E, Opper B. Effects of Pituitary Adenylate Cyclase Activating Polypeptide on Cell Death. Int J Mol Sci 2022; 23:ijms23094953. [PMID: 35563353 PMCID: PMC9100246 DOI: 10.3390/ijms23094953] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 12/18/2022] Open
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) was first isolated as a hypothalamic peptide based on its efficacy to increase adenylate cyclase (AC) activity. It has a widespread distribution throughout the body including the nervous system and peripheral organs, where PACAP exerts protective effects both in vivo and in vitro through its anti-apoptotic, anti-inflammatory, and antioxidant functions. The aim of the present paper was to review the currently available literature regarding the effects of PACAP on cell death in vitro in neural and non-neural cells. Among others, its effect on apoptosis can be detected in cerebellar granule cells against different toxic stimuli. Different neural cell types from the cerebral cortex are also prevented from cell death. PACAP also shows effects on cell death in cells belonging to the peripheral nervous system and protects both neural and non-neural cells of sensory organs. In addition, cell survival-promoting effect can be observed in different peripheral organ systems including cardiovascular, immune, respiratory, gastrointestinal, urinary, and reproductive systems. The studies summarized here indicate its noteworthy effect on cell death in different in vitro models, suggesting PACAP’s potential therapeutic usage in several pathological conditions.
Collapse
|
26
|
Ni A, Ernst C. Evidence That Substantia Nigra Pars Compacta Dopaminergic Neurons Are Selectively Vulnerable to Oxidative Stress Because They Are Highly Metabolically Active. Front Cell Neurosci 2022; 16:826193. [PMID: 35308118 PMCID: PMC8931026 DOI: 10.3389/fncel.2022.826193] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/28/2022] [Indexed: 12/21/2022] Open
Abstract
There are 400–500 thousand dopaminergic cells within each side of the human substantia nigra pars compacta (SNpc) making them a minuscule portion of total brain mass. These tiny clusters of cells have an outsized impact on motor output and behavior as seen in disorders such as Parkinson’s disease (PD). SNpc dopaminergic neurons are more vulnerable to oxidative stress compared to other brain cell types, but the reasons for this are not precisely known. Here we provide evidence to support the hypothesis that this selective vulnerability is because SNpc neurons sustain high metabolic rates compared to other neurons. A higher baseline requirement for ATP production may lead to a selective vulnerability to impairments in oxidative phosphorylation (OXPHOS) or genetic insults that impair Complex I of the electron transport chain. We suggest that the energy demands of the unique morphological and electrophysiological properties of SNpc neurons may be one reason these cells produce more ATP than other cells. We further provide evidence to support the hypothesis that transcription factors (TFs) required to drive induction, differentiation, and maintenance of midbrain dopaminergic neural progenitor cells which give rise to terminally differentiated SNpc neurons are uniquely involved in both developmental patterning and metabolism, a dual function unlike other TFs that program neurons in other brain regions. The use of these TFs during induction and differentiation may program ventral midbrain progenitor cells metabolically to higher ATP levels, allowing for the development of those specialized cell processes seen in terminally differentiated cells. This paper provides a cellular and developmental framework for understanding the selective vulnerability of SNpc dopaminergic cells to oxidative stress.
Collapse
|
27
|
Examination of pituitary adenylate cyclase-activating polypeptide in Parkinson’s disease focusing on correlations with motor symptoms. GeroScience 2022; 44:785-803. [PMID: 35220508 PMCID: PMC9135934 DOI: 10.1007/s11357-022-00530-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/15/2022] [Indexed: 12/16/2022] Open
Abstract
The neuroprotective effects of pituitary adenylate cyclase-activating polypeptide (PACAP) have been shown in numerous in vitro and in vivo models of Parkinson’s disease (PD) supporting the theory that PACAP could have an important role in the pathomechanism of the disorder affecting mostly older patients. Earlier studies found changes in PACAP levels in neurological disorders; therefore, the aim of our study was to examine PACAP in plasma samples of PD patients. Peptide levels were measured with ELISA and correlated with clinical parameters, age, stage of the disorder based on the Hoehn and Yahr (HY) scale, subtype of the disease, treatment, and specific scores measuring motor and non-motor symptoms, such as movement disorder society-unified Parkinson’s disease rating scale (MDS-UPDRS), Epworth sleepiness scale (ESS), Parkinson’s disease sleep scale (PDSS-2), and Beck depression inventory (BDI). Our results showed significantly decreased PACAP levels in PD patients without deep brain stimulation (DBS) therapy and in akinetic-rigid subtype; additionally we also observed a further decrease in the HY stage 3 and 4. Elevated PACAP levels were found in patients with DBS. There were no significant correlations between PACAP level with MDS-UPDRS, type of pharmacological treatment, PDSS-2 sleepiness, or depression (BDI) scales, but we found increased PACAP level in patients with more severe sleepiness problems based on the ESS scale. Based on these results, we suggest that following the alterations of PACAP with other frequently used clinical biomarkers in PD patients might improve strategic planning of further therapeutic interventions and help to provide a clearer prognosis regarding the future perspective of the disease.
Collapse
|
28
|
Solés-Tarrés I, Cabezas-Llobet N, Lefranc B, Leprince J, Alberch J, Vaudry D, Xifró X. Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Protects Striatal Cells and Improves Motor Function in Huntington’s Disease Models: Role of PAC1 Receptor. Front Pharmacol 2022; 12:797541. [PMID: 35153755 PMCID: PMC8832515 DOI: 10.3389/fphar.2021.797541] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/31/2021] [Indexed: 12/21/2022] Open
Abstract
Huntington’s disease (HD) is a hereditary neurodegenerative disorder caused by the expression of mutant huntingtin (mHtt). One of the main features of HD is the degeneration of the striatum that leads to motor discoordination. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that acts through three receptors named PAC1R, VPAC1R, and VPAC2R. In the present study, we first investigated the effect of PACAP on STHdhQ7/Q7 and STHdhQ111/Q111 cells that express wild-type Htt with 7 and mHtt with 111 glutamines, respectively. Then we explored the capacity of PACAP to rescue motor symptoms in the R6/1, a murine model of HD. We found that PACAP treatment (10–7 M) for 24 h protects STHdhQ111/Q111 cells from mHtt-induced apoptosis. This effect is associated with an increase in PAC1R transcription, phosphorylation of ERK and Akt, and an increase of intracellular c-fos, egr1, CBP, and BDNF protein content. Moreover, the use of pharmacological inhibitors revealed that activation of ERK and Akt mediates these antiapoptotic and neurotrophic effects of PACAP. To find out PAC1R implication, we treated STHdh cells with vasoactive intestinal peptide (VIP), which exhibits equal affinity for VPAC1R and VPAC2R, but lower affinity for PAC1R, in contrast to PACAP which has same affinity for the three receptors. VIP reduced cleaved caspase-3 protein level, without promoting the expression of c-fos, egr1, CBP, and the neurotrophin BDNF. We next measured the protein level of PACAP receptors in the striatum and cortex of R6/1 mice. We observed a specific reduction of PAC1R at the onset of motor symptoms. Importantly, the intranasal administration of PACAP to R6/1 animals restored the motor function and increased the striatal levels of PAC1R, CBP, and BDNF. In conclusion, PACAP exerts antiapoptotic and neurotrophic effects in striatal neurons mainly through PAC1R. This effect in HD striatum allows the recovery of motor function and point out PAC1R as a therapeutic target for treatment of HD.
Collapse
Affiliation(s)
- Irene Solés-Tarrés
- New Therapeutic Targets Group, Department of Medical Science, Faculty of Medicine, University of Girona, Girona, Spain
| | - Núria Cabezas-Llobet
- New Therapeutic Targets Group, Department of Medical Science, Faculty of Medicine, University of Girona, Girona, Spain
| | - Benjamin Lefranc
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, UNIROUEN, Inserm, Normandie University, Rouen, France
- Regional Cell Imaging Platform of Normandy (PRIMACEN), UNIROUEN, Normandie University, Rouen, France
| | - Jérôme Leprince
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, UNIROUEN, Inserm, Normandie University, Rouen, France
- Regional Cell Imaging Platform of Normandy (PRIMACEN), UNIROUEN, Normandie University, Rouen, France
| | - Jordi Alberch
- Departament de Biomedicina, Institut de Neurociències, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
- Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - David Vaudry
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, UNIROUEN, Inserm, Normandie University, Rouen, France
- Regional Cell Imaging Platform of Normandy (PRIMACEN), UNIROUEN, Normandie University, Rouen, France
| | - Xavier Xifró
- New Therapeutic Targets Group, Department of Medical Science, Faculty of Medicine, University of Girona, Girona, Spain
- *Correspondence: Xavier Xifró,
| |
Collapse
|
29
|
Wong-Guerra M, Montano-Peguero Y, Ramírez-Sánchez J, Jiménez-Martin J, Fonseca-Fonseca LA, Hernández-Enseñat D, Nonose Y, Valdés O, Mondelo-Rodriguez A, Ortiz-Miranda Y, Bergado G, Carmenate T, Soto Del Valle RM, Pardo-Andreu G, Outeiro TF, Padrón-Yaquis AS, Martimbianco de Assis A, O Souza D, Nuñez-Figueredo Y. JM-20 treatment prevents neuronal damage and memory impairment induced by aluminum chloride in rats. Neurotoxicology 2021; 87:70-85. [PMID: 34481871 DOI: 10.1016/j.neuro.2021.08.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 02/05/2023]
Abstract
The number of people with dementia worldwide is estimated at 50 million by 2018 and continues to rise mainly due to increasing aging and population growth. Clinical impact of current interventions remains modest and all efforts aimed at the identification of new therapeutic approaches are therefore critical. Previously, we showed that JM-20, a dihydropyridine-benzodiazepine hybrid molecule, protected memory processes against scopolamine-induced cholinergic dysfunction. In order to gain further insight into the therapeutic potential of JM-20 on cognitive decline and Alzheimer's disease (AD) pathology, here we evaluated its neuroprotective effects after chronic aluminum chloride (AlCl3) administration to rats and assessed possible alterations in several types of episodic memory and associated pathological mechanisms. Oral administration of aluminum to rodents recapitulates several neuropathological alterations and cognitive impairment, being considered a convenient tool for testing the efficacy of new therapies for dementia. We used behavioral tasks to test spatial, emotional- associative and novel object recognition memory, as well as molecular, enzymatic and histological assays to evaluate selected biochemical parameters. Our study revealed that JM-20 prevented memory decline alongside the inhibition of AlCl3 -induced oxidative stress, increased AChE activity, TNF-α and pro-apoptotic proteins (like Bax, caspase-3, and 8) levels. JM-20 also protected against neuronal damage in the hippocampus and prefrontal cortex. Our findings expanded our understanding of the ability of JM-20 to preserve memory in rats under neurotoxic conditions and confirm its potential capacity to counteract cognitive impairment and etiological factors of AD by breaking the progression of key steps associated with neurodegeneration.
Collapse
Affiliation(s)
- Maylin Wong-Guerra
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26, No.1605, e/Boyeros y Puentes Grandes, CP10600, La Habana, Cuba
| | - Yanay Montano-Peguero
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26, No.1605, e/Boyeros y Puentes Grandes, CP10600, La Habana, Cuba
| | - Jeney Ramírez-Sánchez
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26, No.1605, e/Boyeros y Puentes Grandes, CP10600, La Habana, Cuba
| | - Javier Jiménez-Martin
- Department of Physiology, Otago School of Medical Sciences, University of Otago, P.O. Box 913, Dunedin, 9016, New Zealand
| | - Luis Arturo Fonseca-Fonseca
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26, No.1605, e/Boyeros y Puentes Grandes, CP10600, La Habana, Cuba
| | - Daniela Hernández-Enseñat
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26, No.1605, e/Boyeros y Puentes Grandes, CP10600, La Habana, Cuba
| | - Yasmine Nonose
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil
| | - Odalys Valdés
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26, No.1605, e/Boyeros y Puentes Grandes, CP10600, La Habana, Cuba
| | - Abel Mondelo-Rodriguez
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26, No.1605, e/Boyeros y Puentes Grandes, CP10600, La Habana, Cuba
| | - Yaquelin Ortiz-Miranda
- Centro de Inmunología Molecular, Calle 216 esq 15, Atabey, Playa, PO Box 16040, Havana, Cuba
| | - Gretchen Bergado
- Centro de Inmunología Molecular, Calle 216 esq 15, Atabey, Playa, PO Box 16040, Havana, Cuba
| | - Tania Carmenate
- Centro de Inmunología Molecular, Calle 216 esq 15, Atabey, Playa, PO Box 16040, Havana, Cuba
| | | | - Gilberto Pardo-Andreu
- Centro de Estudio para las Investigaciones y Evaluaciones Biológicas, Instituto de Farmacia y Alimentos, Universidad de La Habana, Calle 222, No. 2317, e/ 23 y 31, La Coronela, La Lisa, CP 13600, La Habana, Cuba
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Gottingen, Göttingen, Germany; Max Planck Institute for Experimental Medicine, Goettingen, Germany; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle, UK
| | - Alejandro Saúl Padrón-Yaquis
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26, No.1605, e/Boyeros y Puentes Grandes, CP10600, La Habana, Cuba
| | - Adriano Martimbianco de Assis
- University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK; Post-graduate Program in Health and Behavior, Health Sciences Centre, Universidade Católica de Pelotas, Pelotas, Brazil
| | - Diogo O Souza
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul-UFRGS, Porto Alegre, Brazil
| | - Yanier Nuñez-Figueredo
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26, No.1605, e/Boyeros y Puentes Grandes, CP10600, La Habana, Cuba.
| |
Collapse
|
30
|
Serra GP, Guillaumin A, Dumas S, Vlcek B, Wallén-Mackenzie Å. Midbrain Dopamine Neurons Defined by TrpV1 Modulate Psychomotor Behavior. Front Neural Circuits 2021; 15:726893. [PMID: 34858142 PMCID: PMC8632262 DOI: 10.3389/fncir.2021.726893] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Dopamine (DA) neurons of the ventral tegmental area (VTA) continue to gain attention as far more heterogeneous than previously realized. Within the medial aspect of the VTA, the unexpected presence of TrpV1 mRNA has been identified. TrpV1 encodes the Transient Receptor Potential cation channel subfamily V member 1, TRPV1, also known as the capsaicin receptor, well recognized for its role in heat and pain processing by peripheral neurons. In contrast, the brain distribution of TrpV1 has been debated. Here, we hypothesized that the TrpV1+ identity defines a distinct subpopulation of VTA DA neurons. To explore these brain TrpV1+ neurons, histological analyses and Cre-driven mouse genetics were employed. TrpV1 mRNA was most strongly detected at the perinatal stage forming a band of scattered neurons throughout the medial VTA, reaching into the posterior hypothalamus. Within the VTA, the majority of TrpV1 co-localized with both Tyrosine hydroxylase (Th) and Vesicular monoamine transporter 2 (Vmat2), confirming a DA phenotype. However, TrpV1 also co-localized substantially with Vesicular glutamate transporter 2 (Vglut2), representing the capacity for glutamate (GLU) release. These TrpV1+/Th+/Vglut2+/Vmat2+ neurons thus constitute a molecularly and anatomically distinct subpopulation of DA-GLU co-releasing neurons. To assess behavioral impact, a TrpV1Cre -driven strategy targeting the Vmat2 gene in mice was implemented. This manipulation was sufficient to alter psychomotor behavior induced by amphetamine. The acute effect of the drug was accentuated above control levels, suggesting super-sensitivity in the drug-na ve state resembling a "pre-sensitized" phenotype. However, no progressive increase with repeated injections was observed. This study identifies a distinct TrpV1+ VTA subpopulation as a critical modulatory component in responsiveness to amphetamine. Moreover, expression of the gene encoding TRPV1 in selected VTA neurons opens up for new possibilities in pharmacological intervention of this heterogeneous, but clinically important, brain area.
Collapse
Affiliation(s)
- Gian Pietro Serra
- Unit of Comparative Physiology, Department of Organism Biology, Uppsala University, Uppsala, Sweden
| | - Adriane Guillaumin
- Unit of Comparative Physiology, Department of Organism Biology, Uppsala University, Uppsala, Sweden
| | | | - Bianca Vlcek
- Unit of Comparative Physiology, Department of Organism Biology, Uppsala University, Uppsala, Sweden
| | - Åsa Wallén-Mackenzie
- Unit of Comparative Physiology, Department of Organism Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
31
|
Iarkov A, Mendoza C, Echeverria V. Cholinergic Receptor Modulation as a Target for Preventing Dementia in Parkinson's Disease. Front Neurosci 2021; 15:665820. [PMID: 34616271 PMCID: PMC8488354 DOI: 10.3389/fnins.2021.665820] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 08/26/2021] [Indexed: 12/20/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative condition characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) in the midbrain resulting in progressive impairment in cognitive and motor abilities. The physiological and molecular mechanisms triggering dopaminergic neuronal loss are not entirely defined. PD occurrence is associated with various genetic and environmental factors causing inflammation and mitochondrial dysfunction in the brain, leading to oxidative stress, proteinopathy, and reduced viability of dopaminergic neurons. Oxidative stress affects the conformation and function of ions, proteins, and lipids, provoking mitochondrial DNA (mtDNA) mutation and dysfunction. The disruption of protein homeostasis induces the aggregation of alpha-synuclein (α-SYN) and parkin and a deficit in proteasome degradation. Also, oxidative stress affects dopamine release by activating ATP-sensitive potassium channels. The cholinergic system is essential in modulating the striatal cells regulating cognitive and motor functions. Several muscarinic acetylcholine receptors (mAChR) and nicotinic acetylcholine receptors (nAChRs) are expressed in the striatum. The nAChRs signaling reduces neuroinflammation and facilitates neuronal survival, neurotransmitter release, and synaptic plasticity. Since there is a deficit in the nAChRs in PD, inhibiting nAChRs loss in the striatum may help prevent dopaminergic neurons loss in the striatum and its pathological consequences. The nAChRs can also stimulate other brain cells supporting cognitive and motor functions. This review discusses the cholinergic system as a therapeutic target of cotinine to prevent cognitive symptoms and transition to dementia in PD.
Collapse
Affiliation(s)
- Alexandre Iarkov
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
| | - Cristhian Mendoza
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
| | - Valentina Echeverria
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile.,Research & Development Service, Bay Pines VA Healthcare System, Bay Pines, FL, United States
| |
Collapse
|
32
|
Carmichael K, Sullivan B, Lopez E, Sun L, Cai H. Diverse midbrain dopaminergic neuron subtypes and implications for complex clinical symptoms of Parkinson's disease. AGEING AND NEURODEGENERATIVE DISEASES 2021; 1. [PMID: 34532720 PMCID: PMC8442626 DOI: 10.20517/and.2021.07] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Parkinson’s disease (PD), the most common degenerative movement disorder, is clinically manifested with various motor and non-motor symptoms. Degeneration of midbrain substantia nigra pas compacta (SNc) dopaminergic neurons (DANs) is generally attributed to the motor syndrome. The underlying neuronal mechanisms of non-motor syndrome are largely unexplored. Besides SNc, midbrain ventral tegmental area (VTA) DANs also produce and release dopamine and modulate movement, reward, motivation, and memory. Degeneration of VTA DANs also occurs in postmortem brains of PD patients, implying an involvement of VTA DANs in PD-associated non-motor symptoms. However, it remains to be established that there is a distinct segregation of different SNc and VTA DAN subtypes in regulating different motor and non-motor functions, and that different DAN subpopulations are differentially affected by normal ageing or PD. Traditionally, the distinction among different DAN subtypes was mainly based on the location of cell bodies and axon terminals. With the recent advance of single cell RNA sequencing technology, DANs can be readily classified based on unique gene expression profiles. A combination of specific anatomic and molecular markers shows great promise to facilitate the identification of DAN subpopulations corresponding to different behavior modules under normal and disease conditions. In this review, we first summarize the recent progress in characterizing genetically, anatomically, and functionally diverse midbrain DAN subtypes. Then, we provide perspectives on how the preclinical research on the connectivity and functionality of DAN subpopulations improves our current understanding of cell-type and circuit specific mechanisms of the disease, which could be critically informative for designing new mechanistic treatments.
Collapse
Affiliation(s)
- Kathleen Carmichael
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.,The Graduate Partnership Program of NIH and Brown University, National Institutes of Health, Bethesda, MD 20892, USA
| | - Breanna Sullivan
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elena Lopez
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lixin Sun
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
33
|
Aguila J, Cheng S, Kee N, Cao M, Wang M, Deng Q, Hedlund E. Spatial RNA Sequencing Identifies Robust Markers of Vulnerable and Resistant Human Midbrain Dopamine Neurons and Their Expression in Parkinson's Disease. Front Mol Neurosci 2021; 14:699562. [PMID: 34305528 PMCID: PMC8297217 DOI: 10.3389/fnmol.2021.699562] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/08/2021] [Indexed: 01/26/2023] Open
Abstract
Defining transcriptional profiles of substantia nigra pars compacta (SNc) and ventral tegmental area (VTA) dopamine neurons is critical to understanding their differential vulnerability in Parkinson’s Disease (PD). Here, we determine transcriptomes of human SNc and VTA dopamine neurons using LCM-seq on a large sample cohort. We apply a bootstrapping strategy as sample input to DESeq2 and identify 33 stably differentially expressed genes (DEGs) between these two subpopulations. We also compute a minimal sample size for identification of stable DEGs, which highlights why previous reported profiles from small sample sizes display extensive variability. Network analysis reveal gene interactions unique to each subpopulation and highlight differences in regulation of mitochondrial stability, apoptosis, neuronal survival, cytoskeleton regulation, extracellular matrix modulation as well as synapse integrity, which could explain the relative resilience of VTA dopamine neurons. Analysis of PD tissues showed that while identified stable DEGs can distinguish the subpopulations also in disease, the SNc markers SLIT1 and ATP2A3 were down-regulated and thus appears to be biomarkers of disease. In summary, our study identifies human SNc and VTA marker profiles, which will be instrumental for studies aiming to modulate dopamine neuron resilience and to validate cell identity of stem cell-derived dopamine neurons.
Collapse
Affiliation(s)
- Julio Aguila
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Shangli Cheng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Nigel Kee
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Ming Cao
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Menghan Wang
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Eva Hedlund
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| |
Collapse
|
34
|
Zachariou V, Bauer CE, Seago ER, Panayiotou G, Hall ED, Butterfield DA, Gold BT. Healthy dietary intake moderates the effects of age on brain iron concentration and working memory performance. Neurobiol Aging 2021; 106:183-196. [PMID: 34284261 DOI: 10.1016/j.neurobiolaging.2021.06.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/31/2021] [Accepted: 06/16/2021] [Indexed: 11/18/2022]
Abstract
Age-related brain iron accumulation is linked with oxidative stress, neurodegeneration and cognitive decline. Certain nutrients can reduce brain iron concentration in animal models, however, this association is not well established in humans. Moreover, it remains unknown if nutrition can moderate the effects of age on brain iron concentration and/or cognition. Here, we explored these issues in a sample of 73 healthy older adults (61-86 years old), while controlling for several factors such as age, gender, years of education, physical fitness and alcohol-intake. Quantitative susceptibility mapping was used for assessment of brain iron concentration and participants performed an N-Back paradigm to evaluate working memory performance. Nutritional-intake was assessed via a validated questionnaire. Nutrients were grouped into nutrition factors based on previous literature and factor analysis. One factor, comprised of vitamin E, lysine, DHA omega-3 and LA omega-6 PUFA, representing food groups such as nuts, healthy oils and fish, moderated the effects of age on both brain iron concentration and working memory performance, suggesting that these nutrients may slow the rate of brain iron accumulation and working memory declines in aging.
Collapse
Affiliation(s)
- Valentinos Zachariou
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA.
| | - Christopher E Bauer
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Elayna R Seago
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Georgia Panayiotou
- Department of Psychology and Center for Applied Neuroscience, University of Cyprus, Nicosia, Cyprus
| | - Edward D Hall
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - D Allan Butterfield
- Department of Chemistry, College of Medicine, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Brian T Gold
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA; Magnetic Resonance Imaging and Spectroscopy Center, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
35
|
Fiorenzano A, Sozzi E, Parmar M, Storm P. Dopamine Neuron Diversity: Recent Advances and Current Challenges in Human Stem Cell Models and Single Cell Sequencing. Cells 2021; 10:cells10061366. [PMID: 34206038 PMCID: PMC8226961 DOI: 10.3390/cells10061366] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 12/12/2022] Open
Abstract
Human midbrain dopamine (DA) neurons are a heterogeneous group of cells that share a common neurotransmitter phenotype and are in close anatomical proximity but display different functions, sensitivity to degeneration, and axonal innervation targets. The A9 DA neuron subtype controls motor function and is primarily degenerated in Parkinson’s disease (PD), whereas A10 neurons are largely unaffected by the condition, and their dysfunction is associated with neuropsychiatric disorders. Currently, DA neurons can only be reliably classified on the basis of topographical features, including anatomical location in the midbrain and projection targets in the forebrain. No systematic molecular classification at the genome-wide level has been proposed to date. Although many years of scientific efforts in embryonic and adult mouse brain have positioned us to better understand the complexity of DA neuron biology, many biological phenomena specific to humans are not amenable to being reproduced in animal models. The establishment of human cell-based systems combined with advanced computational single-cell transcriptomics holds great promise for decoding the mechanisms underlying maturation and diversification of human DA neurons, and linking their molecular heterogeneity to functions in the midbrain. Human pluripotent stem cells have emerged as a useful tool to recapitulate key molecular features of mature DA neuron subtypes. Here, we review some of the most recent advances and discuss the current challenges in using stem cells, to model human DA biology. We also describe how single cell RNA sequencing may provide key insights into the molecular programs driving DA progenitor specification into mature DA neuron subtypes. Exploiting the state-of-the-art approaches will lead to a better understanding of stem cell-derived DA neurons and their use in disease modeling and regenerative medicine.
Collapse
|
36
|
Zheng Y, Zhang L, Xie J, Shi L. The Emerging Role of Neuropeptides in Parkinson's Disease. Front Aging Neurosci 2021; 13:646726. [PMID: 33762925 PMCID: PMC7982480 DOI: 10.3389/fnagi.2021.646726] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/10/2021] [Indexed: 12/11/2022] Open
Abstract
Parkinson’s disease (PD), the second most common age-related neurodegenerative disease, results from the loss of dopamine neurons in the substantia nigra. This disease is characterized by cardinal non-motor and motor symptoms. Several studies have demonstrated that neuropeptides, such as ghrelin, neuropeptide Y, pituitary adenylate cyclase-activating polypeptide, substance P, and neurotensin, are related to the onset of PD. This review mainly describes the changes in these neuropeptides and their receptors in the substantia nigra-striatum system as well as the other PD-related brain regions. Based on several in vitro and in vivo studies, most neuropeptides play a significant neuroprotective role in PD by preventing caspase-3 activation, decreasing mitochondrial-related oxidative stress, increasing mitochondrial biogenesis, inhibiting microglial activation, and anti-autophagic activity. Thus, neuropeptides may provide a new strategy for PD therapy.
Collapse
Affiliation(s)
- Yanan Zheng
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Linlin Zhang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Junxia Xie
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, China.,Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Limin Shi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, China.,Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| |
Collapse
|
37
|
Thomas R, Moloney EB, Macbain ZK, Hallett PJ, Isacson O. Fibroblasts from idiopathic Parkinson's disease exhibit deficiency of lysosomal glucocerebrosidase activity associated with reduced levels of the trafficking receptor LIMP2. Mol Brain 2021; 14:16. [PMID: 33468204 PMCID: PMC7816505 DOI: 10.1186/s13041-020-00712-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/07/2020] [Indexed: 12/25/2022] Open
Abstract
Lysosomal dysfunction is a central pathway associated with Parkinson's disease (PD) pathogenesis. Haploinsufficiency of the lysosomal hydrolase GBA (encoding glucocerebrosidase (GCase)) is one of the largest genetic risk factors for developing PD. Deficiencies in the activity of the GCase enzyme have been observed in human tissues from both genetic (harboring mutations in the GBA gene) and idiopathic forms of the disease. To understand the mechanisms behind the deficits of lysosomal GCase enzyme activity in idiopathic PD, this study utilized a large cohort of fibroblast cells from control subjects and PD patients with and without mutations in the GBA gene (N370S mutation) (control, n = 15; idiopathic PD, n = 31; PD with GBA N370S mutation, n = 6). The current data demonstrates that idiopathic PD fibroblasts devoid of any mutations in the GBA gene also exhibit reduction in lysosomal GCase activity, similar to those with the GBA N370S mutation. This reduced GCase enzyme activity in idiopathic PD cells was accompanied by decreased expression of the GBA trafficking receptor, LIMP2, and increased ER retention of the GBA protein in these cells. Importantly, in idiopathic PD fibroblasts LIMP2 protein levels correlated significantly with GCase activity, which was not the case in control subjects or in genetic PD GBA N370S cells. In conclusion, idiopathic PD fibroblasts have decreased GCase activity primarily driven by altered LIMP2-mediated transport of GBA to lysosome and the reduced GCase activity exhibited by the genetic GBA N370S derived PD fibroblasts occurs through a different mechanism.
Collapse
Affiliation(s)
- Ria Thomas
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA
| | - Elizabeth B Moloney
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA
| | - Zachary K Macbain
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA
| | - Penelope J Hallett
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA.
| | - Ole Isacson
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA.
| |
Collapse
|
38
|
Single-Cell Technologies in Parkinson׳s Disease. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11613-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
39
|
Muddapu VR, Chakravarthy VS. A Multi-Scale Computational Model of Excitotoxic Loss of Dopaminergic Cells in Parkinson's Disease. Front Neuroinform 2020; 14:34. [PMID: 33101001 PMCID: PMC7555610 DOI: 10.3389/fninf.2020.00034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/14/2020] [Indexed: 11/30/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder caused by loss of dopaminergic neurons in substantia nigra pars compacta (SNc). Although the exact cause of cell death is not clear, the hypothesis that metabolic deficiency is a key factor has been gaining attention in recent years. In the present study, we investigated this hypothesis using a multi-scale computational model of the subsystem of the basal ganglia comprising the subthalamic nucleus (STN), globus pallidus externa (GPe), and SNc. The proposed model is a multiscale model in that interaction among the three nuclei are simulated using more abstract Izhikevich neuron models, while the molecular pathways involved in cell death of SNc neurons are simulated in terms of detailed chemical kinetics. Simulation results obtained from the proposed model showed that energy deficiencies occurring at cellular and network levels could precipitate the excitotoxic loss of SNc neurons in PD. At the subcellular level, the models show how calcium elevation leads to apoptosis of SNc neurons. The therapeutic effects of several neuroprotective interventions are also simulated in the model. From neuroprotective studies, it was clear that glutamate inhibition and apoptotic signal blocker therapies were able to halt the progression of SNc cell loss when compared to other therapeutic interventions, which only slowed down the progression of SNc cell loss.
Collapse
Affiliation(s)
- Vignayanandam Ravindernath Muddapu
- Laboratory for Computational Neuroscience, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - V Srinivasa Chakravarthy
- Laboratory for Computational Neuroscience, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
40
|
Fougère M, van der Zouwen CI, Boutin J, Ryczko D. Heterogeneous expression of dopaminergic markers and Vglut2 in mouse mesodiencephalic dopaminergic nuclei A8-A13. J Comp Neurol 2020; 529:1273-1292. [PMID: 32869307 DOI: 10.1002/cne.25020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022]
Abstract
Co-transmission of glutamate by brain dopaminergic (DA) neurons was recently proposed as a potential factor influencing cell survival in models of Parkinson's disease. Intriguingly, brain DA nuclei are differentially affected in Parkinson's disease. Whether this is associated with different patterns of co-expression of the glutamatergic phenotype along the rostrocaudal brain axis is unknown in mammals. We hypothesized that, as in zebrafish, the glutamatergic phenotype is present preferentially in the caudal mesodiencephalic DA nuclei. Here, we used in mice a cell fate mapping strategy based on reporter protein expression (ZsGreen) consecutive to previous expression of the vesicular glutamate transporter 2 (Vglut2) gene, coupled with immunofluorescence experiments against tyrosine hydroxylase (TH) or dopamine transporter (DAT). We found three expression patterns in DA cells, organized along the rostrocaudal brain axis. The first pattern (TH-positive, DAT-positive, ZsGreen-positive) was found in A8-A10. The second pattern (TH-positive, DAT-negative, ZsGreen-positive) was found in A11. The third pattern (TH-positive, DAT-negative, ZsGreen-negative) was found in A12-A13. These patterns should help to refine the establishment of the homology of DA nuclei between vertebrate species. Our results also uncover that Vglut2 is expressed at some point during cell lifetime in DA nuclei known to degenerate in Parkinson's disease and largely absent from those that are preserved, suggesting that co-expression of the glutamatergic phenotype in DA cells influences their survival in Parkinson's disease.
Collapse
Affiliation(s)
- Maxime Fougère
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de La Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Cornelis Immanuel van der Zouwen
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de La Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Joël Boutin
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de La Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Dimitri Ryczko
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de La Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
- Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada
- Centre d'Excellence en Neurosciences de l'Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
41
|
Maugeri G, D'Amico AG, Morello G, Reglodi D, Cavallaro S, D'Agata V. Differential Vulnerability of Oculomotor Versus Hypoglossal Nucleus During ALS: Involvement of PACAP. Front Neurosci 2020; 14:805. [PMID: 32848572 PMCID: PMC7432287 DOI: 10.3389/fnins.2020.00805] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive multifactorial disease characterized by the loss of motor neurons (MNs). Not all MNs undergo degeneration: neurons of the oculomotor nucleus, which regulate eye movements, are less vulnerable compared to hypoglossal nucleus MNs. Several molecular studies have been performed to understand the different vulnerability of these MNs. By analyzing postmortem samples from ALS patients to other unrelated decedents, the differential genomic pattern between the two nuclei has been profiled. Among identified genes, adenylate cyclase activating polypeptide 1 (ADCYAP1) gene, encoding for pituitary adenylate cyclase-activating polypeptide (PACAP), was found significantly up-regulated in the oculomotor versus hypoglossal nucleus suggesting that it could play a trophic effect on MNs in ALS. In the present review, some aspects regarding the different vulnerability of oculomotor and hypoglossal nucleus to degeneration will be summarized. The distribution and potential role of PACAP on these MNs as studied largely in an animal model of ALS compared to controls, will be discussed.
Collapse
Affiliation(s)
- Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, Catania, Italy
| | | | - Giovanna Morello
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Catania, Italy
| | - Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
| | - Sebastiano Cavallaro
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Catania, Italy
| | - Velia D'Agata
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, Catania, Italy
| |
Collapse
|
42
|
Kim TW, Koo SY, Studer L. Pluripotent Stem Cell Therapies for Parkinson Disease: Present Challenges and Future Opportunities. Front Cell Dev Biol 2020; 8:729. [PMID: 32903681 PMCID: PMC7438741 DOI: 10.3389/fcell.2020.00729] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/15/2020] [Indexed: 12/16/2022] Open
Abstract
In Parkinson's disease (PD), there are currently no effective therapies to prevent or slow down disease progression. Cell replacement therapy using human pluripotent stem cell (hPSC)-derived dopamine neurons holds considerable promise. It presents a novel, regenerative strategy, building on the extensive history of fetal tissue grafts and capturing the potential of hPSCs to serve as a scalable and standardized cell source. Progress in establishing protocols for the direct differentiation to midbrain dopamine (mDA) neurons from hPSC have catalyzed the development of cell-based therapies for PD. Consequently, several groups have derived clinical-grade mDA neuron precursors under clinical good manufacture practice condition, which are progressing toward clinical testing in PD patients. Here we will review the current status of the field, discuss the remaining key challenges, and highlight future areas for further improvements of hPSC-based technologies in the clinical translation to PD.
Collapse
Affiliation(s)
- Tae Wan Kim
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, United States.,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, United States
| | - So Yeon Koo
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, United States.,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, United States.,Neuroscience Graduate Program of Weill Cornell Graduate School of Biomedical Sciences, Weill Cornell Medicine, New York, NY, United States
| | - Lorenz Studer
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, United States.,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, United States
| |
Collapse
|
43
|
Solés-Tarrés I, Cabezas-Llobet N, Vaudry D, Xifró X. Protective Effects of Pituitary Adenylate Cyclase-Activating Polypeptide and Vasoactive Intestinal Peptide Against Cognitive Decline in Neurodegenerative Diseases. Front Cell Neurosci 2020; 14:221. [PMID: 32765225 PMCID: PMC7380167 DOI: 10.3389/fncel.2020.00221] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/22/2020] [Indexed: 12/23/2022] Open
Abstract
Cognitive impairment is one of the major symptoms in most neurodegenerative disorders such as Alzheimer’s (AD), Parkinson (PD), and Huntington diseases (HD), affecting millions of people worldwide. Unfortunately, there is no treatment to cure or prevent the progression of those diseases. Cognitive impairment has been related to neuronal cell death and/or synaptic plasticity alteration in important brain regions, such as the cerebral cortex, substantia nigra, striatum, and hippocampus. Therefore, compounds that can act to protect the neuronal loss and/or to reestablish the synaptic activity are needed to prevent cognitive decline in neurodegenerative diseases. Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are two highly related multifunctional neuropeptides widely distributed in the central nervous system (CNS). PACAP and VIP exert their action through two common receptors, VPAC1 and VPAC2, while PACAP has an additional specific receptor, PAC1. In this review article, we first presented evidence showing the therapeutic potential of PACAP and VIP to fight the cognitive decline observed in models of AD, PD, and HD. We also reviewed the main transduction pathways activated by PACAP and VIP receptors to reduce cognitive dysfunction. Furthermore, we identified the therapeutic targets of PACAP and VIP, and finally, we evaluated different novel synthetic PACAP and VIP analogs as promising pharmacological tools.
Collapse
Affiliation(s)
- Irene Solés-Tarrés
- New Therapeutic Targets Group (TargetsLab), Department of Medical Science, Faculty of Medicine, Universitat de Girona, Girona, Spain
| | - Núria Cabezas-Llobet
- New Therapeutic Targets Group (TargetsLab), Department of Medical Science, Faculty of Medicine, Universitat de Girona, Girona, Spain
| | - David Vaudry
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, Normandie University, UNIROUEN, Inserm, Rouen, France
| | - Xavier Xifró
- New Therapeutic Targets Group (TargetsLab), Department of Medical Science, Faculty of Medicine, Universitat de Girona, Girona, Spain
| |
Collapse
|
44
|
Thomas R, Hallett PJ, Isacson O. Experimental studies of mitochondrial and lysosomal function in in vitro and in vivo models relevant to Parkinson's disease genetic risk. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:279-302. [PMID: 32739007 DOI: 10.1016/bs.irn.2020.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Several studies have identified the involvement of mitochondrial and lysosomal dysfunction in Parkinson's disease (PD) pathology. In this review we discuss recent work that has identified deficits in mitophagy, mitochondrial network formation, increased sensitivity to mitochondrial stressors and alterations in proteins regulating mitochondrial fission and fusion associated with patient-derived fibroblasts harboring mutations in LRRK2 gene and from sporadic PD patient cells. We further focus on alterations of lysosomal enzymes, in particular glucocerebrosidase activity, and resultant lipid dyshomeostasis in PD and aging, in human tissue and in vivo rodent models. Future studies aimed at understanding the convergence of mitochondrial and lysosomal pathways will be of essence for the identification of unique cellular defects in PD and for the development of new treatments.
Collapse
Affiliation(s)
- Ria Thomas
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, United States.
| | - Penelope J Hallett
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, United States.
| | - Ole Isacson
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, United States.
| |
Collapse
|
45
|
Cardoso T, Lévesque M. Toward Generating Subtype-Specific Mesencephalic Dopaminergic Neurons in vitro. Front Cell Dev Biol 2020; 8:443. [PMID: 32626706 PMCID: PMC7311634 DOI: 10.3389/fcell.2020.00443] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022] Open
Abstract
Mesencephalic dopaminergic (mDA) neurons derived from pluripotent stem cells (PSCs) have proven to be pivotal for disease modeling studies and as a source of transplantable tissue for regenerative therapies in Parkinson's disease (PD). Current differentiation protocols can generate standardized and reproducible cell products of dopaminergic neurons that elicit the characteristic transcriptional profile of ventral midbrain. Nonetheless, dopamine neurons residing in the mesencephalon comprise distinct groups of cells within diffusely defined anatomical boundaries and with distinct functional, electrophysiological, and molecular properties. Here we review recent single cell sequencing studies that are shedding light on the neuronal heterogeneity within the mesencephalon and discuss how resolving the complex molecular profile of distinct sub-populations within this region could help refine patterning and quality control assessment of PSC-derived mDA neurons to subtype-specificity in vitro. In turn, such advances would have important impact in improving cell replacement therapy, disease mechanistic studies and drug screening in PD.
Collapse
Affiliation(s)
- Tiago Cardoso
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.,CERVO Brain Research Center, Université Laval, Québec, QC, Canada
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.,CERVO Brain Research Center, Université Laval, Québec, QC, Canada
| |
Collapse
|
46
|
Korecka JA, Thomas R, Christensen DP, Hinrich AJ, Ferrari EJ, Levy SA, Hastings ML, Hallett PJ, Isacson O. Mitochondrial clearance and maturation of autophagosomes are compromised in LRRK2 G2019S familial Parkinson's disease patient fibroblasts. Hum Mol Genet 2020; 28:3232-3243. [PMID: 31261377 DOI: 10.1093/hmg/ddz126] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 04/16/2019] [Accepted: 06/07/2019] [Indexed: 12/13/2022] Open
Abstract
This study utilized human fibroblasts as a preclinical discovery and diagnostic platform for identification of cell biological signatures specific for the LRRK2 G2019S mutation producing Parkinson's disease (PD). Using live cell imaging with a pH-sensitive Rosella biosensor probe reflecting lysosomal breakdown of mitochondria, mitophagy rates were found to be decreased in fibroblasts carrying the LRRK2 G2019S mutation compared to cells isolated from healthy subject (HS) controls. The mutant LRRK2 increased kinase activity was reduced by pharmacological inhibition and targeted antisense oligonucleotide treatment, which normalized mitophagy rates in the G2019S cells and also increased mitophagy levels in HS cells. Detailed mechanistic analysis showed a reduction of mature autophagosomes in LRRK2 G2019S fibroblasts, which was rescued by LRRK2 specific kinase inhibition. These findings demonstrate an important role for LRRK2 protein in regulation of mitochondrial clearance by the lysosomes, which is hampered in PD with the G2019S mutation. The current results are relevant for cell phenotypic diagnostic approaches and potentially for stratification of PD patients for targeted therapy.
Collapse
Affiliation(s)
- Joanna A Korecka
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Ria Thomas
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Dan P Christensen
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Anthony J Hinrich
- Center for Genetic Diseases, Department of Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Eliza J Ferrari
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Simon A Levy
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Michelle L Hastings
- Center for Genetic Diseases, Department of Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Penelope J Hallett
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Ole Isacson
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| |
Collapse
|
47
|
Delic V, Beck KD, Pang KCH, Citron BA. Biological links between traumatic brain injury and Parkinson's disease. Acta Neuropathol Commun 2020; 8:45. [PMID: 32264976 PMCID: PMC7137235 DOI: 10.1186/s40478-020-00924-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 03/26/2020] [Indexed: 12/19/2022] Open
Abstract
Parkinson's Disease (PD) is a progressive neurodegenerative disorder with no cure. Clinical presentation is characterized by postural instability, resting tremors, and gait problems that result from progressive loss of A9 dopaminergic neurons in the substantia nigra pars compacta. Traumatic brain injury (TBI) has been implicated as a risk factor for several neurodegenerative diseases, but the strongest evidence is linked to development of PD. Mild TBI (mTBI), is the most common and is defined by minimal, if any, loss of consciousness and the absence of significant observable damage to the brain tissue. mTBI is responsible for a 56% higher risk of developing PD in U.S. Veterans and the risk increases with severity of injury. While the mounting evidence from human studies suggests a link between TBI and PD, fundamental questions as to whether TBI nucleates PD pathology or accelerates PD pathology in vulnerable populations remains unanswered. Several promising lines of research point to inflammation, metabolic dysregulation, and protein accumulation as potential mechanisms through which TBI can initiate or accelerate PD. Amyloid precursor protein (APP), alpha synuclein (α-syn), hyper-phosphorylated Tau, and TAR DNA-binding protein 43 (TDP-43), are some of the most frequently reported proteins upregulated following a TBI and are also closely linked to PD. Recently, upregulation of Leucine Rich Repeat Kinase 2 (LRRK2), has been found in the brain of mice following a TBI. Subset of Rab proteins were identified as biological substrates of LRRK2, a protein also extensively linked to late onset PD. Inhibition of LRRK2 was found to be neuroprotective in PD and TBI models. The goal of this review is to survey current literature concerning the mechanistic overlap between TBI and PD with a particular focus on inflammation, metabolic dysregulation, and aforementioned proteins. This review will also cover the application of rodent TBI models to further our understanding of the relationship between TBI and PD.
Collapse
Affiliation(s)
- Vedad Delic
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA.
- NeuroBehavioral Research Laboratory, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA.
| | - Kevin D Beck
- NeuroBehavioral Research Laboratory, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers- New Jersey Medical School, Newark, NJ, 07103, USA
| | - Kevin C H Pang
- NeuroBehavioral Research Laboratory, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers- New Jersey Medical School, Newark, NJ, 07103, USA
| | - Bruce A Citron
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research and Development (Mailstop 15), 385 Tremont Ave, East Orange, NJ, 07018, USA
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers- New Jersey Medical School, Newark, NJ, 07103, USA
| |
Collapse
|
48
|
Alexoudi A, Deftereos S. CGRP Antagonists: Side Effects and Potential Parkinson’s Disease Development. Headache 2020; 60:789-790. [DOI: 10.1111/head.13770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 01/06/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Athanasia Alexoudi
- Department of Neurosurgery National & Kapodistrian University of Athens Evangelismos Hospital Athens Greece
| | | |
Collapse
|
49
|
Muddapu VR, Dharshini SAP, Chakravarthy VS, Gromiha MM. Neurodegenerative Diseases - Is Metabolic Deficiency the Root Cause? Front Neurosci 2020; 14:213. [PMID: 32296300 PMCID: PMC7137637 DOI: 10.3389/fnins.2020.00213] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/26/2020] [Indexed: 01/31/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer, Parkinson, Huntington, and amyotrophic lateral sclerosis, are a prominent class of neurological diseases currently without a cure. They are characterized by an inexorable loss of a specific type of neurons. The selective vulnerability of specific neuronal clusters (typically a subcortical cluster) in the early stages, followed by the spread of the disease to higher cortical areas, is a typical pattern of disease progression. Neurodegenerative diseases share a range of molecular and cellular pathologies, including protein aggregation, mitochondrial dysfunction, glutamate toxicity, calcium load, proteolytic stress, oxidative stress, neuroinflammation, and aging, which contribute to neuronal death. Efforts to treat these diseases are often limited by the fact that they tend to address any one of the above pathological changes while ignoring others. Lack of clarity regarding a possible root cause that underlies all the above pathologies poses a significant challenge. In search of an integrative theory for neurodegenerative pathology, we hypothesize that metabolic deficiency in certain vulnerable neuronal clusters is the common underlying thread that links many dimensions of the disease. The current review aims to present an outline of such an integrative theory. We present a new perspective of neurodegenerative diseases as metabolic disorders at molecular, cellular, and systems levels. This helps to understand a common underlying mechanism of the many facets of the disease and may lead to more promising disease-modifying therapeutic interventions. Here, we briefly discuss the selective metabolic vulnerability of specific neuronal clusters and also the involvement of glia and vascular dysfunctions. Any failure in satisfaction of the metabolic demand by the neurons triggers a chain of events that precipitate various manifestations of neurodegenerative pathology.
Collapse
Affiliation(s)
- Vignayanandam Ravindernath Muddapu
- Laboratory for Computational Neuroscience, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - S. Akila Parvathy Dharshini
- Protein Bioinformatics Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - V. Srinivasa Chakravarthy
- Laboratory for Computational Neuroscience, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - M. Michael Gromiha
- Protein Bioinformatics Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
50
|
Monzón-Sandoval J, Poggiolini I, Ilmer T, Wade-Martins R, Webber C, Parkkinen L. Human-Specific Transcriptome of Ventral and Dorsal Midbrain Dopamine Neurons. Ann Neurol 2020; 87:853-868. [PMID: 32167609 PMCID: PMC8651008 DOI: 10.1002/ana.25719] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 02/01/2023]
Abstract
Objective Neuronal loss in the substantia nigra pars compacta (SNpc) in Parkinson disease (PD) is not uniform, as dopamine neurons from the ventral tier are lost more rapidly than those of the dorsal tier. Identifying the intrinsic differences that account for this differential vulnerability may provide a key for developing new treatments for PD. Methods Here, we compared the RNA‐sequenced transcriptomes of ~100 laser captured microdissected SNpc neurons from each tier from 7 healthy controls. Results Expression levels of dopaminergic markers were similar across the tiers, whereas markers specific to the neighboring ventral tegmental area were virtually undetected. After accounting for unwanted sources of variation, we identified 106 differentially expressed genes (DEGs) between the SNpc tiers. The genes higher in the dorsal/resistant SNpc tier neurons displayed coordinated patterns of expression across the human brain, their protein products had more interactions than expected by chance, and they demonstrated evidence of functional convergence. No significant shared functionality was found for genes higher in the ventral/vulnerable SNpc tier. Surprisingly but importantly, none of the identified DEGs was among the familial PD genes or genome‐wide associated loci. Finally, we found some DEGs in opposite tier orientation between human and analogous mouse populations. Interpretation Our results highlight functional enrichments of vesicular trafficking, ion transport/homeostasis and oxidative stress genes showing higher expression in the resistant neurons of the SNpc dorsal tier. Furthermore, the comparison of gene expression variation in human and mouse SNpc populations strongly argues for the need of human‐focused omics studies. ANN NEUROL 2020;87:853–868
Collapse
Affiliation(s)
- Jimena Monzón-Sandoval
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford.,UK Dementia Research Institute, Cardiff University, Cardiff
| | - Ilaria Poggiolini
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford
| | - Tobias Ilmer
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford.,Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Caleb Webber
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford.,UK Dementia Research Institute, Cardiff University, Cardiff.,Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Laura Parkkinen
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford.,Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford
| |
Collapse
|