1
|
Garcia Moreno SI, Limani F, Ludwig I, Gilbert C, Pifl C, Hnasko TS, Steinkellner T. Viral overexpression of human alpha-synuclein in mouse substantia nigra dopamine neurons results in hyperdopaminergia but no neurodegeneration. Exp Neurol 2024; 382:114959. [PMID: 39288832 DOI: 10.1016/j.expneurol.2024.114959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/19/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
Loss of select neuronal populations such as midbrain dopamine (DA) neurons is a pathological hallmark of Parkinson's disease (PD). The small neuronal protein α-synuclein has been related both genetically and neuropathologically to PD, yet how and if it contributes to selective vulnerability remains elusive. Here, we describe the generation of a novel adeno-associated viral vector (AAV) for Cre-dependent overexpression of wild-type human α-synuclein. Our strategy allows us to restrict α-synuclein to select neuronal populations and hence investigate the cell-autonomous effects of elevated α-synuclein in genetically-defined cell types. Since DA neurons in the substantia nigra pars compacta (SNc) are particularly vulnerable in PD, we investigated in more detail the effects of increased α-synuclein in these cells. AAV-mediated overexpression of wildtype human α-synuclein in SNc DA neurons increased the levels of α-synuclein within these cells and augmented phosphorylation of α-synuclein at serine-129, which is considered a pathological feature of PD and other synucleinopathies. However, despite abundant α-synuclein overexpression and hyperphosphorylation we did not observe any dopaminergic neurodegeneration up to 90 days post virus infusion. In contrast, we noticed that overexpression of α-synuclein resulted in increased locomotor activity and elevated striatal DA levels suggesting that α-synuclein enhanced dopaminergic activity. We therefore conclude that cell-autonomous effects of elevated α-synuclein are not sufficient to trigger acute dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Sofia Ines Garcia Moreno
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Fabian Limani
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Iina Ludwig
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Catherine Gilbert
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Christian Pifl
- Center for Brain Research, Medical University of Vienna, Vienna, Vienna, Austria
| | - Thomas S Hnasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Department of Veterans Affairs, San Diego Veterans Affairs Healthcare System, San Diego, CA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Thomas Steinkellner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
2
|
Morais LH, Boktor JC, MahmoudianDehkordi S, Kaddurah-Daouk R, Mazmanian SK. α-synuclein overexpression and the microbiome shape the gut and brain metabolome in mice. NPJ Parkinsons Dis 2024; 10:208. [PMID: 39477976 DOI: 10.1038/s41531-024-00816-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Pathological forms of α-synuclein contribute to synucleinopathies, including Parkinson's disease (PD). Most cases of PD arise from gene-environment interactions. Microbiome composition is altered in PD, and gut bacteria are causal to symptoms in animal models. We quantitatively profiled nearly 630 metabolites in the gut, plasma, and brain of α-synuclein-overexpressing (ASO) mice, compared to wild-type (WT) animals, and comparing germ-free (GF) to specific pathogen-free (SPF) animals (n = 5 WT-SPF; n = 6 ASO-SPF; n = 6 WT-GF; n = 6 ASO-GF). Many differentially expressed metabolites in ASO mice are also dysregulated in human PD patients, including amine oxides, bile acids and indoles. The microbial metabolite trimethylamine N-oxide (TMAO) strongly correlates from the gut to the plasma to the brain in mice, notable since TMAO is elevated in the blood and cerebrospinal fluid of PD patients. These findings uncover broad metabolomic changes that are influenced by the intersection of host genetics and microbiome in a mouse model of PD.
Collapse
Affiliation(s)
- Livia H Morais
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Joseph C Boktor
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | | | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA.
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA.
- Department of Medicine, Duke University, Durham, NC, USA.
| | - Sarkis K Mazmanian
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
3
|
Kim Y, McInnes J, Kim J, Liang YHW, Veeraragavan S, Garza AR, Belfort BDW, Arenkiel B, Samaco R, Zoghbi HY. Olfactory deficit and gastrointestinal dysfunction precede motor abnormalities in alpha-Synuclein G51D knock-in mice. Proc Natl Acad Sci U S A 2024; 121:e2406479121. [PMID: 39284050 PMCID: PMC11441490 DOI: 10.1073/pnas.2406479121] [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: 04/01/2024] [Accepted: 08/09/2024] [Indexed: 10/02/2024] Open
Abstract
Parkinson's disease (PD) is typically a sporadic late-onset disorder, which has made it difficult to model in mice. Several transgenic mouse models bearing mutations in SNCA, which encodes alpha-Synuclein (α-Syn), have been made, but these lines do not express SNCA in a physiologically accurate spatiotemporal pattern, which limits the ability of the mice to recapitulate the features of human PD. Here, we generated knock-in mice bearing the G51D SNCA mutation. After establishing that their motor symptoms begin at 9 mo of age, we then sought earlier pathologies. We assessed the phosphorylation at Serine 129 of α-Syn in different tissues and detected phospho-α-Syn in the olfactory bulb and enteric nervous system at 3 mo of age. Olfactory deficit and impaired gut transit followed at 6 mo, preceding motor symptoms. The SncaG51D mice thus parallel the progression of human PD and will enable us to study PD pathogenesis and test future therapies.
Collapse
Affiliation(s)
- YoungDoo Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030
| | - Joseph McInnes
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030
| | - Jiyoen Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030
| | - Yan Hong Wei Liang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030
| | - Surabi Veeraragavan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030
| | - Alexandra Rae Garza
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030
| | - Benjamin David Webst Belfort
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030
| | - Benjamin Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Rodney Samaco
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030
| | - Huda Yahya Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030
- HHMI, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
4
|
Soni D, Upadhayay S, Dhureja M, Arthur R, Kumar P. Crosstalk between gut-brain axis: unveiling the mysteries of gut ROS in progression of Parkinson's disease. Inflammopharmacology 2024:10.1007/s10787-024-01510-2. [PMID: 38992324 DOI: 10.1007/s10787-024-01510-2] [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: 01/22/2024] [Accepted: 06/06/2024] [Indexed: 07/13/2024]
Abstract
"Path to a good mood lies through the gut." This statement seems to imply that it has long been believed that the gut is connected with the brain. Research has shown that eating food activates the reward system and releases dopamine (DA), establishing a link between the peripheral and central nervous system. At the same time, researchers also trust that the gut is involved in the onset of many diseases, including Parkinson's disease (PD), in which gastrointestinal dysfunction is considered a prevalent symptom. Reports suggest that PD starts from the gut and reaches the brain via the vagus nerve. Recent studies have revealed an intriguing interaction between the gut and brain, which links gut dysbiosis to the etiology of PD. This review aims to explore the mechanistic pathway how reactive oxygen species (ROS) generation in the gut affects the makeup and operation of the dopamine circuitry in the brain. Our primary concern is ROS generation in the gut, which disrupts the gut microbiome (GM), causing α-synuclein accumulation and inflammation. This trio contributes to the loss of DA neurons in the brain, resulting in PD development. This review also compiles pre-clinical and clinical studies on antioxidants, demonstrating that antioxidants reduce ROS and increase DA levels. Collectively, the study highlights the necessity of comprehending the gut-brain axis for unraveling the riddles of PD pathogenesis and considering new therapeutic approaches.
Collapse
Affiliation(s)
- Divya Soni
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Shubham Upadhayay
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Maanvi Dhureja
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Richmond Arthur
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India.
| |
Collapse
|
5
|
Nakos Bimpos M, Karali K, Antoniou C, Palermos D, Fouka M, Delis A, Tzieras I, Chrousos GP, Koutmani Y, Stefanis L, Polissidis A. Alpha-synuclein-induced stress sensitivity renders the Parkinson's disease brain susceptible to neurodegeneration. Acta Neuropathol Commun 2024; 12:100. [PMID: 38886854 PMCID: PMC11181569 DOI: 10.1186/s40478-024-01797-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/12/2024] [Indexed: 06/20/2024] Open
Abstract
A link between chronic stress and Parkinson's disease (PD) pathogenesis is emerging. Ample evidence demonstrates that the presynaptic neuronal protein alpha-synuclein (asyn) is closely tied to PD pathogenesis. However, it is not known whether stress system dysfunction is present in PD, if asyn is involved, and if, together, they contribute to neurodegeneration. To address these questions, we assess stress axis function in transgenic rats overexpressing full-length wildtype human asyn (asyn BAC rats) and perform multi-level stress and PD phenotyping following chronic corticosterone administration. Stress signaling, namely corticotropin-releasing factor, glucocorticoid and mineralocorticoid receptor gene expression, is also examined in post-mortem PD patient brains. Overexpression of human wildtype asyn leads to HPA axis dysregulation in rats, while chronic corticosterone administration significantly aggravates nigrostriatal degeneration, serine129 phosphorylated asyn (pS129) expression and neuroinflammation, leading to phenoconversion from a prodromal to an overt motor PD phenotype. Interestingly, chronic corticosterone in asyn BAC rats induces a robust, twofold increase in pS129 expression in the hypothalamus, the master regulator of the stress response, while the hippocampus, both a regulator and a target of the stress response, also demonstrates elevated pS129 asyn levels and altered markers of stress signalling. Finally, defective hippocampal stress signalling is mirrored in human PD brains and correlates with asyn expression levels. Taken together, our results link brain stress system dysregulation with asyn and provide evidence that elevated circulating glucocorticoids can contribute to asyn-induced neurodegeneration, ultimately triggering phenoconversion from prodromal to overt PD.
Collapse
Affiliation(s)
- Modestos Nakos Bimpos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Katerina Karali
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- German Center for Neurodegenerative Diseases, Feodor-Lynen-Straße 17, 81377, Munich, Germany
- Athens International Master's Programme in Neurosciences, Department of Biology, National and Kapodistrian University of Athens, 15784, Illisia, Athens, Greece
| | - Christine Antoniou
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- Athens International Master's Programme in Neurosciences, Department of Biology, National and Kapodistrian University of Athens, 15784, Illisia, Athens, Greece
| | - Dionysios Palermos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Maria Fouka
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Anastasios Delis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Iason Tzieras
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - George Panagiotis Chrousos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- University Research Institute on Maternal and Child Health and Precision Medicine, and UNESCO Chair on Adolescent Health Care, Aghia Sophia Children's Hospital, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Yassemi Koutmani
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Leonidas Stefanis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- 1St Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Alexia Polissidis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece.
- Department of Science and Mathematics, ACG-Research Center, Deree - American College of Greece, 15342, Athens, Greece.
| |
Collapse
|
6
|
Jin X, Si X, Lei X, Liu H, Shao A, Li L. Disruption of Dopamine Homeostasis Associated with Alteration of Proteins in Synaptic Vesicles: A Putative Central Mechanism of Parkinson's Disease Pathogenesis. Aging Dis 2024; 15:1204-1226. [PMID: 37815908 PMCID: PMC11081171 DOI: 10.14336/ad.2023.0821-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 08/21/2023] [Indexed: 10/12/2023] Open
Abstract
Vestigial dopaminergic cells in PD have selectivity for a sub-class of hypersensitive neurons with the nigrostriatal dopamine (DA) tract. DA is modulated in pre-synaptic nerve terminals to remain stable. To be specific, proteins at DA release sites that have a function of synthesizing and packing DA in cytoplasm, modulating release and reingestion, and changing excitability of neurons, display regional discrepancies that uncover relevancy of the observed sensitivity to neurodegenerative changes. Although the reasons of a majority of PD cases are still indistinct, heredity and environment are known to us to make significant influences. For decades, genetic analysis of PD patients with heredity in family have promoted our comprehension of pathogenesis to a great extent, which reveals correlative mechanisms including oxidative stress, abnormal protein homeostasis and mitochondrial dysfunction. In this review, we review the constitution of presynaptic vesicle related to DA homeostasis and describe the genetic and environmental evidence of presynaptic dysfunction that increase risky possibility of PD concerning intracellular vesicle transmission and their functional outcomes. We summarize alterations in synaptic vesicular proteins with great involvement in the reasons of some DA neurons highly vulnerable to neurodegenerative changes. We generalize different potential targets and therapeutic strategies for different pathogenic mechanisms, providing a reference for further studies of PD treatment in the future. But it remains to be further researched on this recently discovered and converging mechanism of vesicular dynamics and PD, which will provide a more profound comprehension and put up with new therapeutic tactics for PD patients.
Collapse
Affiliation(s)
- Xuanxiang Jin
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Xiaoli Si
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Xiaoguang Lei
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, the First School of Clinical Medicine, Kunming Medical University, Kunming, China.
| | - Huifang Liu
- Division of Neurology, Department of Medicine, University of Hong Kong, Hong Kong.
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Disease, Hangzhou, China.
| | - Lingfei Li
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
7
|
Moreno SIG, Limani F, Ludwig I, Gilbert C, Pifl C, Hnasko TS, Steinkellner T. Viral overexpression of human alpha-synuclein in mouse substantia nigra dopamine neurons results in hyperdopaminergia but no neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592188. [PMID: 38746104 PMCID: PMC11092628 DOI: 10.1101/2024.05.03.592188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Loss of select neuronal populations such as midbrain dopamine (DA) neurons is a pathological hallmark of Parkinson's disease (PD). The small neuronal protein α-synuclein has been related both genetically and neuropathologically to PD, yet how it contributes to selective vulnerability remains elusive. Here, we describe the generation of a novel adeno-associated viral vector (AAV) for Cre-dependent overexpression of wild-type human α-synuclein. Our strategy allows us to restrict α-synuclein to select neuronal populations and hence investigate the cell-autonomous effects of elevated α-synuclein in genetically-defined cell types. Since DA neurons in the substantia nigra pars compacta (SNc) are particularly vulnerable in PD, we investigated in more detail the effects of increased α-synuclein in these cells. AAV-mediated overexpression of wildtype human α-synuclein in SNc DA neurons increased the levels of α-synuclein within these cells and augmented phosphorylation of α-synuclein at serine-129, which is considered a pathological feature of PD and other synucleinopathies. However, despite abundant α-synuclein overexpression and hyperphosphorylation we did not observe any DA neurodegeneration up to 90 days post virus infusion. In contrast, we noticed that overexpression of α-synuclein resulted in increased locomotor activity and elevated striatal DA levels suggesting that α-synuclein enhanced dopaminergic activity. We therefore conclude that cell-autonomous effects of elevated α-synuclein are not sufficient to trigger acute DA neurodegeneration.
Collapse
|
8
|
Syvänen V, Koistinaho J, Lehtonen Š. Identification of the abnormalities in astrocytic functions as potential drug targets for neurodegenerative disease. Expert Opin Drug Discov 2024; 19:603-616. [PMID: 38409817 DOI: 10.1080/17460441.2024.2322988] [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: 10/26/2023] [Accepted: 02/21/2024] [Indexed: 02/28/2024]
Abstract
INTRODUCTION Historically, astrocytes were seen primarily as a supportive cell population within the brain; with neurodegenerative disease research focusing exclusively on malfunctioning neurons. However, astrocytes perform numerous tasks that are essential for maintenance of the central nervous system`s complex processes. Disruption of these functions can have negative consequences; hence, it is unsurprising to observe a growing amount of evidence for the essential role of astrocytes in the development and progression of neurodegenerative diseases. Targeting astrocytic functions may serve as a potential disease-modifying drug therapy in the future. AREAS COVERED The present review emphasizes the key astrocytic functions associated with neurodegenerative diseases and explores the possibility of pharmaceutical interventions to modify these processes. In addition, the authors provide an overview of current advancement in this field by including studies of possible drug candidates. EXPERT OPINION Glial research has experienced a significant renaissance in the last quarter-century. Understanding how disease pathologies modify or are caused by astrocyte functions is crucial when developing treatments for brain diseases. Future research will focus on building advanced models that can more precisely correlate to the state in the human brain, with the goal of routinely testing therapies in these models.
Collapse
Affiliation(s)
- Valtteri Syvänen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jari Koistinaho
- Neuroscience Center, Helsinki Institute of Life Science, and Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
| | - Šárka Lehtonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
9
|
Seiler JL, Zhuang X, Nelson AB, Lerner TN. Dopamine across timescales and cell types: Relevance for phenotypes in Parkinson's disease progression. Exp Neurol 2024; 374:114693. [PMID: 38242300 DOI: 10.1016/j.expneurol.2024.114693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
Dopamine neurons in the substantia nigra pars compacta (SNc) synthesize and release dopamine, a critical neurotransmitter for movement and learning. SNc dopamine neurons degenerate in Parkinson's Disease (PD), causing a host of motor and non-motor symptoms. Here, we review recent conceptual advances in our basic understanding of the dopamine system - including our rapidly advancing knowledge of dopamine neuron heterogeneity - with special attention to their importance for understanding PD. In PD patients, dopamine neuron degeneration progresses from lateral SNc to medial SNc, suggesting clinically relevant heterogeneity in dopamine neurons. With technical advances in dopamine system interrogation, we can understand the relevance of this heterogeneity for PD progression and harness it to develop new treatments.
Collapse
Affiliation(s)
- Jillian L Seiler
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Xiaowen Zhuang
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Alexandra B Nelson
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Talia N Lerner
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern University Interdepartmental Neuroscience Program (NUIN), Evanston, IL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
10
|
Patterson JR, Kochmanski J, Stoll AC, Kubik M, Kemp CJ, Duffy MF, Thompson K, Howe JW, Cole-Strauss A, Kuhn NC, Miller KM, Nelson S, Onyekpe CU, Beck JS, Counts SE, Bernstein AI, Steece-Collier K, Luk KC, Sortwell CE. Transcriptomic profiling of early synucleinopathy in rats induced with preformed fibrils. NPJ Parkinsons Dis 2024; 10:7. [PMID: 38172128 PMCID: PMC10764951 DOI: 10.1038/s41531-023-00620-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Examination of early phases of synucleinopathy when inclusions are present, but long before neurodegeneration occurs, is critical to both understanding disease progression and the development of disease modifying therapies. The rat alpha-synuclein (α-syn) preformed fibril (PFF) model induces synchronized synucleinopathy that recapitulates the pathological features of Parkinson's disease (PD) and can be used to study synucleinopathy progression. In this model, phosphorylated α-syn (pSyn) inclusion-containing neurons and reactive microglia (major histocompatibility complex-II immunoreactive) peak in the substantia nigra pars compacta (SNpc) months before appreciable neurodegeneration. However, it remains unclear which specific genes are driving these phenotypic changes. To identify transcriptional changes associated with early synucleinopathy, we used laser capture microdissection of the SNpc paired with RNA sequencing (RNASeq). Precision collection of the SNpc allowed for the assessment of differential transcript expression in the nigral dopamine neurons and proximal glia. Transcripts upregulated in early synucleinopathy were mainly associated with an immune response, whereas transcripts downregulated were associated with neurotransmission and the dopamine pathway. A subset of 29 transcripts associated with neurotransmission/vesicular release and the dopamine pathway were verified in a separate cohort of males and females to confirm reproducibility. Within this subset, fluorescent in situ hybridization (FISH) was used to localize decreases in the Syt1 and Slc6a3 transcripts to pSyn inclusion-containing neurons. Identification of transcriptional changes in early synucleinopathy provides insight into the molecular mechanisms driving neurodegeneration.
Collapse
Affiliation(s)
- Joseph R Patterson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA.
- Neuroscience Program, Michigan State University, East Lansing, MI, USA.
| | - Joseph Kochmanski
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Anna C Stoll
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Michael Kubik
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Christopher J Kemp
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Megan F Duffy
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Kajene Thompson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Jacob W Howe
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Allyson Cole-Strauss
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Nathan C Kuhn
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Kathryn M Miller
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Seth Nelson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Christopher U Onyekpe
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - John S Beck
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Scott E Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Alison I Bernstein
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, NJ, USA
| | - Kathy Steece-Collier
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Kelvin C Luk
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Caryl E Sortwell
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
11
|
Medina-Luque J, Piechocinski P, Feyen P, Sgobio C, Herms J. Striatal dopamine neurotransmission is altered in age- and region-specific manner in a Parkinson's disease transgenic mouse. Sci Rep 2024; 14:164. [PMID: 38167878 PMCID: PMC10761704 DOI: 10.1038/s41598-023-49600-5] [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: 10/01/2023] [Accepted: 12/10/2023] [Indexed: 01/05/2024] Open
Abstract
Dopamine (DA) plays a critical role in striatal motor control. The drop in DA level within the dorsal striatum is directly associated with the appearance of motor symptoms in Parkinson's disease (PD). The progression of the disease and inherent disruption of the DA neurotransmission has been closely related to accumulation of the synaptic protein α-synuclein. However, it is still unclear how α-synuclein affects dopaminergic terminals in different areas of dorsal striatum. Here we demonstrate that the overexpression of human α-synuclein (h-α-syn) interferes with the striatal DA neurotransmission in an age-dependent manner, preferentially in the dorsolateral striatum (DLS) of PDGF-h-α-syn mice. While 3-month-old mice showed an increase at the onset of h-α-syn accumulation in the DLS, 12-month-old mice revealed a decrease in electrically-evoked DA release. The enhanced DA release in 3-month-old mice coincided with better performance in a behavioural task. Notably, DA amplitude alterations were also accompanied by a delay in the DA clearance independently from the animal age. Structurally, dopamine transporter (DAT) was found to be redistributed in larger DAT-positive clumps only in the DLS of 3- and 12-month-old mice. Together, our data provide new insight into the vulnerability of DLS and suggest DAT-related dysfunctionalities from the very early stages of h-α-syn accumulation.
Collapse
Affiliation(s)
- Jose Medina-Luque
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
| | | | - Paul Feyen
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Carmelo Sgobio
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Centre for Neuropathology and Prion Research, Ludwig-Maximilians University, Munich, Germany.
| | - Jochen Herms
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Centre for Neuropathology and Prion Research, Ludwig-Maximilians University, Munich, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
12
|
Flores AJ, Bartlett MJ, Seaton BT, Samtani G, Sexauer MR, Weintraub NC, Siegenthaler JR, Lu D, Heien ML, Porreca F, Sherman SJ, Falk T. Antagonism of kappa opioid receptors accelerates the development of L-DOPA-induced dyskinesia in a preclinical model of moderate dopamine depletion. Brain Res 2023; 1821:148613. [PMID: 37783263 PMCID: PMC10841913 DOI: 10.1016/j.brainres.2023.148613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023]
Abstract
Levels of the opioid peptide dynorphin, an endogenous ligand selective for kappa-opioid receptors (KORs), its mRNA and pro-peptide precursors are differentially dysregulated in Parkinson's disease (PD) and following the development of l-DOPA-induced dyskinesia (LID). It remains unclear whether these alterations contribute to the pathophysiological mechanisms underlying PD motor impairment and the subsequent development of LID, or whether they are part of compensatory mechanisms. We sought to investigate nor-BNI, a KOR antagonist, 1) in the dopamine (DA)-depleted PD state, 2) during the development phase of LID, and 3) via measuring of tonic levels of striatal DA. While nor-BNI (3 mg/kg; s.c.) did not lead to functional restoration in the DA-depleted state, it affected the dose-dependent development of abnormal voluntary movements (AIMs) in response to escalating doses of l-DOPA in a rat PD model with a moderate striatal 6-hydroxdopamine (6-OHDA) lesion. We tested five escalating doses of l-DOPA (6, 12, 24, 48, 72 mg/kg; i.p.), and nor-BNI significantly increased the development of AIMs at the 12 and 24 mg/kg l-DOPA doses. However, after reaching the 72 mg/kg l-DOPA, AIMs were not significantly different between control and nor-BNI groups. In summary, while blocking KORs significantly increased the rate of development of LID induced by chronic, escalating doses of l-DOPA in a moderate-lesioned rat PD model, it did not contribute further once the overall severity of LID was established. While we observed an increase of tonic DA levels in the moderately lesioned dorsolateral striatum, there was no tonic DA change following administration of nor-BNI.
Collapse
Affiliation(s)
- Andrew J Flores
- Department of Neurology, The University of Arizona, Tucson, AZ 85724, USA; Graduate Interdisciplinary Program in Physiological Sciences, The University of Arizona, Tucson, AZ 85724, USA
| | - Mitchell J Bartlett
- Department of Neurology, The University of Arizona, Tucson, AZ 85724, USA; Department of Pharmacology, The University of Arizona, Tucson, AZ 85724, USA
| | - Blake T Seaton
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
| | - Grace Samtani
- Department of Neurology, The University of Arizona, Tucson, AZ 85724, USA
| | - Morgan R Sexauer
- Department of Neurology, The University of Arizona, Tucson, AZ 85724, USA
| | - Nathan C Weintraub
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA; Department of Pharmacology, The University of Arizona, Tucson, AZ 85724, USA
| | - James R Siegenthaler
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
| | - Dong Lu
- Department of Pharmacology, The University of Arizona, Tucson, AZ 85724, USA
| | - Michael L Heien
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
| | - Frank Porreca
- Department of Pharmacology, The University of Arizona, Tucson, AZ 85724, USA
| | - Scott J Sherman
- Department of Neurology, The University of Arizona, Tucson, AZ 85724, USA
| | - Torsten Falk
- Department of Neurology, The University of Arizona, Tucson, AZ 85724, USA; Graduate Interdisciplinary Program in Physiological Sciences, The University of Arizona, Tucson, AZ 85724, USA; Department of Pharmacology, The University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
13
|
Martinez PA, Martinez VE, Rani S, Murrell M, Javors M, Gelfond J, Doorn JA, Fernandez E, Strong R. Impaired aldehyde detoxification exacerbates motor deficits in an alpha-synuclein mouse model of Parkinson's disease. Brain Behav 2023; 13:e3150. [PMID: 37452461 PMCID: PMC10498093 DOI: 10.1002/brb3.3150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 06/07/2023] [Indexed: 07/18/2023] Open
Abstract
INTRODUCTION The discovery of biogenic aldehydes in the postmortem parkinsonian brain and the ability of these aldehydes to modify and cross-link proteins has called attention to their possible role in Parkinson's disease. For example, many in vitro studies have found that the aldehyde metabolite of dopamine, 3,4-dihydroxyphenylacetaldehyde (DOPAL), induces the formation of stable, neurotoxic alpha-synuclein oligomers. METHODS To study this in vivo, mice deficient in the two aldehyde dehydrogenase enzymes (Aldh1a1 and Aldh2, DKO) primarily responsible for detoxification of DOPAL in the nigrostriatal pathway were crossed with mice that overexpress human wild-type alpha-synuclein. DKO overexpressing human wild-type alpha-synuclein (DKO/ASO) offspring were evaluated for impairment on motor tasks associated with Parkinsonism. RESULTS DKO/ASO mice developed severe motor deficits greater than that of mice overexpressing human wild-type alpha-synuclein alone. CONCLUSION These results provide evidence to support the idea that biogenic aldehydes such as DOPAL interact with human wild-type alpha-synuclein, directly or indirectly, in vivo to exacerbate locomotor deficits in Parkinson's disease.
Collapse
Affiliation(s)
- Paul Anthony Martinez
- Department of PharmacologyUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Center for Biomedical NeuroscienceUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - Vanessa Elia Martinez
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - Sheela Rani
- Department of PharmacologyUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - Meredith Murrell
- Department of PsychiatryUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Martin Javors
- Department of PharmacologyUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Department of PsychiatryUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Jonathan Gelfond
- Department of MedicineUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Department of Epidemiology & BiostatisticsUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - Jonathan Alan Doorn
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of PharmacyThe University of IowaIowa CityIowaUSA
| | - Elizabeth Fernandez
- Department of PharmacologyUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Geriatric Research, Education, and Clinical CenterSouth Texas Veterans Health. Care NetworkSan AntonioTexasUSA
| | - Randy Strong
- Department of PharmacologyUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Center for Biomedical NeuroscienceUniversity of Texas Health Science CenterSan AntonioTexasUSA
- Geriatric Research, Education, and Clinical CenterSouth Texas Veterans Health. Care NetworkSan AntonioTexasUSA
| |
Collapse
|
14
|
Videlock EJ, Xing T, Yehya AHS, Travagli RA. Experimental models of gut-first Parkinson's disease: A systematic review. Neurogastroenterol Motil 2023; 35:e14604. [PMID: 37125607 PMCID: PMC10524037 DOI: 10.1111/nmo.14604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND There is strong support from studies in humans and in animal models that Parkinson's disease (PD) may begin in the gut. This brings about a unique opportunity for researchers in the field of neurogastroenterology to contribute to advancing the field and making contributions that could lead to the ability to diagnose and treat PD in the premotor stages. Lack of familiarity with some of the aspects of the experimental approaches used in these studies may present a barrier for neurogastroenterology researchers to enter the field. Much remains to be understood about intestinal-specific components of gut-first PD pathogenesis and the field would benefit from contributions of enteric and central nervous system neuroscientists. PURPOSE To address these issues, we have conducted a systematic review of the two most frequently used experimental models of gut-first PD: transneuronal propagation of α-synuclein preformed fibrils and oral exposure to environmental toxins. We have reviewed the details of these studies and present methodological considerations for the use of these models. Our aim is that this review will serve as a framework and useful reference for neuroscientists, gastroenterologists, and neurologists interested in applying their expertise to advancing our understanding of gut-first PD.
Collapse
Affiliation(s)
- Elizabeth J. Videlock
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Tiaosi Xing
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Ashwaq Hamid Salem Yehya
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | | |
Collapse
|
15
|
Rocha E, Chamoli M, Chinta SJ, Andersen JK, Wallis R, Bezard E, Goldberg M, Greenamyre T, Hirst W, Kuan WL, Kirik D, Niedernhofer L, Rappley I, Padmanabhan S, Trudeau LE, Spillantini M, Scott S, Studer L, Bellantuono I, Mortiboys H. Aging, Parkinson's Disease, and Models: What Are the Challenges? AGING BIOLOGY 2023; 1:e20230010. [PMID: 38978807 PMCID: PMC11230631 DOI: 10.59368/agingbio.20230010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Parkinson's disease (PD) is a chronic, neurodegenerative condition characterized by motor symptoms such as bradykinesia, rigidity, and tremor, alongside multiple nonmotor symptoms. The appearance of motor symptoms is linked to progressive dopaminergic neuron loss within the substantia nigra. PD incidence increases sharply with age, suggesting a strong association between mechanisms driving biological aging and the development and progression of PD. However, the role of aging in the pathogenesis of PD remains understudied. Numerous models of PD, including cell models, toxin-induced models, and genetic models in rodents and nonhuman primates (NHPs), reproduce different aspects of PD, but preclinical studies of PD rarely incorporate age as a factor. Studies using patient neurons derived from stem cells via reprogramming methods retain some aging features, but their characterization, particularly of aging markers and reproducibility of neuron type, is suboptimal. Investigation of age-related changes in PD using animal models indicates an association, but this is likely in conjunction with other disease drivers. The biggest barrier to drawing firm conclusions is that each model lacks full characterization and appropriate time-course assessments. There is a need to systematically investigate whether aging increases the susceptibility of mouse, rat, and NHP models to develop PD and understand the role of cell models. We propose that a significant investment in time and resources, together with the coordination and sharing of resources, knowledge, and data, is required to accelerate progress in understanding the role of biological aging in PD development and improve the reliability of models to test interventions.
Collapse
Affiliation(s)
- Emily Rocha
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Shankar J Chinta
- Buck Institute for Research on Aging, Novato, CA, USA
- Touro University California, College of Pharmacy, Vallejo, CA, USA
| | | | - Ruby Wallis
- The Healthy Lifespan Institute, Sheffield, United Kingdom
| | | | | | - Tim Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - We-Li Kuan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (BRAINS), Lund, Sweden
| | - Laura Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Irit Rappley
- Recursion pharmaceuticals, Salt Lake City, UT, USA
| | | | - Louis-Eric Trudeau
- Department of pharmacology and physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Maria Spillantini
- Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | | | - Lorenz Studer
- The Center for Stem Cell Biology and Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Ilaria Bellantuono
- The Healthy Lifespan Institute, Sheffield, United Kingdom
- Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| | - Heather Mortiboys
- The Healthy Lifespan Institute, Sheffield, United Kingdom
- Department of Neuroscience, Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kindgom
| |
Collapse
|
16
|
Price DL, Khan A, Angers R, Cardenas A, Prato MK, Bani M, Bonhaus DW, Citron M, Biere AL. In vivo effects of the alpha-synuclein misfolding inhibitor minzasolmin supports clinical development in Parkinson's disease. NPJ Parkinsons Dis 2023; 9:114. [PMID: 37460603 PMCID: PMC10352257 DOI: 10.1038/s41531-023-00552-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 06/26/2023] [Indexed: 07/20/2023] Open
Abstract
Direct targeting of alpha-synuclein (ASYN) has emerged as a disease-modifying strategy for Parkinson's disease and other synucleinopathies which is being approached using both small molecule compounds and ASYN-targeted biologics. Minzasolmin (UCB0599) is an orally bioavailable and brain-penetrant small molecule ASYN misfolding inhibitor in clinical development as a disease-modifying therapeutic for Parkinson's disease. Herein the results of preclinical evaluations of minzasolmin that formed the basis for subsequent clinical development are described. Pharmacokinetic evaluations of intraperitoneal 1 and 5 mg/kg minzasolmin in wildtype mice revealed parallel and dose-proportional exposures in brain and plasma. Three-month administration studies in the Line 61 transgenic mouse model of PD were conducted to measure ASYN pathology and other PD-relevant endpoints including markers of CNS inflammation, striatal DAT labeling and gait. Reductions in ASYN pathology were correlated with improved aspects of gait and balance, reductions in CNS inflammation marker abundance, and normalized striatal DAT levels. These findings provide support for human dose determinations and have informed the translational strategy for clinical trial design and biomarker selection for the ongoing clinical studies of minzasolmin in patients living with early-stage Parkinson's disease (ClinicalTrials.gov ID: NCT04658186; EudraCT Number 2020-003265).
Collapse
Affiliation(s)
| | - Asma Khan
- Neuropore Therapies, Inc., San Diego, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Montanari M, Imbriani P, Bonsi P, Martella G, Peppe A. Beyond the Microbiota: Understanding the Role of the Enteric Nervous System in Parkinson's Disease from Mice to Human. Biomedicines 2023; 11:1560. [PMID: 37371655 DOI: 10.3390/biomedicines11061560] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
The enteric nervous system (ENS) is a nerve network composed of neurons and glial cells that regulates the motor and secretory functions of the gastrointestinal (GI) tract. There is abundant evidence of mutual communication between the brain and the GI tract. Dysfunction of these connections appears to be involved in the pathophysiology of Parkinson's disease (PD). Alterations in the ENS have been shown to occur very early in PD, even before central nervous system (CNS) involvement. Post-mortem studies of PD patients have shown aggregation of α-synuclein (αS) in specific subtypes of neurons in the ENS. Subsequently, αS spreads retrogradely in the CNS through preganglionic vagal fibers to this nerve's dorsal motor nucleus (DMV) and other central nervous structures. Here, we highlight the role of the ENS in PD pathogenesis based on evidence observed in animal models and using a translational perspective. While acknowledging the putative role of the microbiome in the gut-brain axis (GBA), this review provides a comprehensive view of the ENS not only as a "second brain", but also as a window into the "first brain", a potentially crucial element in the search for new therapeutic approaches that can delay and even cure the disease.
Collapse
Affiliation(s)
- Martina Montanari
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Department of Systems Neuroscience, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Paola Imbriani
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Clinical Neuroscience, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Antonella Peppe
- Clinical Neuroscience, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| |
Collapse
|
18
|
Schidlitzki A, Stanojlovic M, Fournier C, Käufer C, Feja M, Gericke B, Garzotti M, Welford RWD, Steiner MA, Angot E, Richter F. Double-Edged Effects of Venglustat on Behavior and Pathology in Mice Overexpressing α-Synuclein. Mov Disord 2023. [PMID: 37050861 DOI: 10.1002/mds.29398] [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: 10/17/2022] [Revised: 03/01/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Venglustat is a brain-penetrant, small molecule inhibitor of glucosylceramide synthase used in clinical testing for treatment of Parkinson's disease (PD). Despite beneficial effects in certain cellular and rodent models, patients with PD with mutations in GBA, the gene for lysosomal glucocerebrosidase, experienced worsening of their motor function under venglustat treatment (NCT02906020, MOVES-PD, phase 2 trial). OBJECTIVE The objective of this study was to evaluate venglustat in mouse models of PD with overexpression of wild-type α-synuclein. METHODS Mice overexpressing α-synuclein (Thy1-aSyn line 61) or Gba-mutated mice with viral vector-induced overexpression of α-synuclein in the substantia nigra were administered venglustat as food admixture. Motor and cognitive performance, α-synuclein-related pathology, and microgliosis were compared with untreated controls. RESULTS Venglustat worsened motor function in Thy1-aSyn transgenics on the challenging beam and the pole test. Although venglustat did not alter the cognitive deficit in the Y-maze test, it alleviated anxiety-related behavior in the novel object recognition test. Venglustat reduced soluble and membrane-bound α-synuclein in the striatum and phosphorylated α-synuclein in limbic brain regions. Although venglustat reversed the loss of parvalbumin immunoreactivity in the basolateral amygdala, it tended to increase microgliosis and phosphorylated α-synuclein in the substantia nigra. Furthermore, venglustat also partially worsened motor performance and tended to increase neurofilament light chain in the cerebrospinal fluid in the Gba-deficient model with nigral α-synuclein overexpression and neurodegeneration. CONCLUSIONS Venglustat treatment in two mouse models of α-synuclein overexpression showed that glucosylceramide synthase inhibition had differential detrimental or beneficial effects on behavior and neuropathology possibly related to brain region-specific effects. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Alina Schidlitzki
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Milos Stanojlovic
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Céline Fournier
- CNS Pharmacology and Drug Discovery, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
- Roche Pharma Research and Early Development (pRED), F. Hoffman/La Roche Ltd, Basel, Switzerland
| | - Christopher Käufer
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Malte Feja
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Birthe Gericke
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Marco Garzotti
- CNS Pharmacology and Drug Discovery, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
- Matterhorn Biosciences AG, Basel, Switzerland
| | - Richard W D Welford
- CNS Pharmacology and Drug Discovery, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
- Roche Pharma Research and Early Development (pRED), F. Hoffman/La Roche Ltd, Basel, Switzerland
| | | | - Elodie Angot
- CNS Pharmacology and Drug Discovery, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
- Roche Pharma Research and Early Development (pRED), F. Hoffman/La Roche Ltd, Basel, Switzerland
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience Hannover, Hannover, Germany
| |
Collapse
|
19
|
Simons E, Fleming SM. Role of rodent models in advancing precision medicine for Parkinson's disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:3-16. [PMID: 36803818 DOI: 10.1016/b978-0-323-85555-6.00002-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
With a current lack of disease-modifying treatments, an initiative toward implementing a precision medicine approach for treating Parkinson's disease (PD) has emerged. However, challenges remain in how to define and apply precision medicine in PD. To accomplish the goal of optimally targeted and timed treatment for each patient, preclinical research in a diverse population of rodent models will continue to be an essential part of the translational path to identify novel biomarkers for patient diagnosis and subgrouping, understand PD disease mechanisms, identify new therapeutic targets, and screen therapeutics prior to clinical testing. This review highlights the most common rodent models of PD and discusses how these models can contribute to defining and implementing precision medicine for the treatment of PD.
Collapse
Affiliation(s)
- Emily Simons
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Sheila M Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States.
| |
Collapse
|
20
|
Hussein A, Guevara CA, Valle PD, Gupta S, Benson DL, Huntley GW. Non-Motor Symptoms of Parkinson's Disease: The Neurobiology of Early Psychiatric and Cognitive Dysfunction. Neuroscientist 2023; 29:97-116. [PMID: 33966533 PMCID: PMC9338765 DOI: 10.1177/10738584211011979] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that has been recognized for over 200 years by its clinically dominant motor system impairment. There are prominent non-motor symptoms as well, and among these, psychiatric symptoms of depression and anxiety and cognitive impairment are common and can appear earlier than motor symptoms. Although the neurobiology underlying these particular PD-associated non-motor symptoms is not completely understood, the identification of PARK genes that contribute to hereditary and sporadic PD has enabled genetic models in animals that, in turn, have fostered ever deepening analyses of cells, synapses, circuits, and behaviors relevant to non-motor psychiatric and cognitive symptoms of human PD. Moreover, while it has long been recognized that inflammation is a prominent component of PD, recent studies demonstrate that brain-immune signaling crosstalk has significant modulatory effects on brain cell and synaptic function in the context of psychiatric symptoms. This review provides a focused update on such progress in understanding the neurobiology of PD-related non-motor psychiatric and cognitive symptoms.
Collapse
Affiliation(s)
- Ayan Hussein
- Nash Family Department of Neuroscience and Friedman Brain Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher A. Guevara
- Nash Family Department of Neuroscience and Friedman Brain Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pamela Del Valle
- Nash Family Department of Neuroscience and Friedman Brain Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Swati Gupta
- Nash Family Department of Neuroscience and Friedman Brain Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deanna L. Benson
- Nash Family Department of Neuroscience and Friedman Brain Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George W. Huntley
- Nash Family Department of Neuroscience and Friedman Brain Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
21
|
Schümann F, Schmitt O, Wree A, Hawlitschka A. Distribution of Cleaved SNAP-25 in the Rat Brain, following Unilateral Injection of Botulinum Neurotoxin-A into the Striatum. Int J Mol Sci 2023; 24:1685. [PMID: 36675200 PMCID: PMC9865012 DOI: 10.3390/ijms24021685] [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: 10/21/2022] [Revised: 12/30/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
In Parkinson's disease, hypercholinism in the striatum occurs, with the consequence of disturbed motor functions. Direct application of Botulinum neurotoxin-A in the striatum of hemi-Parkinsonian rats might be a promising anticholinergic therapeutic option. Here, we aimed to determine the spread of intrastriatally injected BoNT-A in the brain as well as the duration of its action based on the distribution of cleaved SNAP-25. Rats were injected with 1 ng of BoNT-A into the right striatum and the brains were examined at different times up to one year after treatment. In brain sections immunohistochemically stained for BoNT-A, cleaved SNAP-25 area-specific densitometric analyses were performed. Increased immunoreactivity for cleaved SNAP-25 was found in brain regions other than the unilaterally injected striatum. Most cleaved SNAP-25-ir was found in widespread areas ipsilateral to the BoNT-A injection, in some regions, however, immunoreactivity was also measured in the contralateral hemisphere. There was a linear relationship between the distance of a special area from the injected striatum and the time until its maximum averaged immunoreactivity was reached. Moreover, we observed a positive relationship for the area-specific distance from the injected striatum and its maximum immunoreactivity as well as for the connection density with the striatum and its maximum immunoreactivity. The results speak for a bidirectional axonal transport of BoNT-A after its application into the striatum to its widespread connected parts of the brain. Even one year after BoNT-A injection, cleaved SNAP-25 could still be detected.
Collapse
Affiliation(s)
- Friederike Schümann
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstraße 9, 18057 Rostock, Germany
| | - Oliver Schmitt
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstraße 9, 18057 Rostock, Germany
- Medical School Hamburg, Am Kaiserkai 1, 20457 Hamburg, Germany
| | - Andreas Wree
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstraße 9, 18057 Rostock, Germany
| | - Alexander Hawlitschka
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstraße 9, 18057 Rostock, Germany
| |
Collapse
|
22
|
Richter F, Stanojlovic M, Käufer C, Gericke B, Feja M. A Mouse Model to Test Novel Therapeutics for Parkinson's Disease: an Update on the Thy1-aSyn ("line 61") Mice. Neurotherapeutics 2023; 20:97-116. [PMID: 36715870 PMCID: PMC10119371 DOI: 10.1007/s13311-022-01338-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2022] [Indexed: 01/31/2023] Open
Abstract
Development of neuroprotective therapeutics for Parkinson's disease (PD) is facing a lack of translation from pre-clinical to clinical trials. One strategy for improvement is to increase predictive validity of pre-clinical studies by using extensively characterized animal models with a comprehensive set of validated pharmacodynamic readouts. Mice over-expressing full-length, human, wild-type alpha-synuclein under the Thy-1 promoter (Thy1-aSyn line 61) reproduce key features of sporadic PD, such as progressive loss of striatal dopamine, alpha-synuclein pathology, deficits in motor and non-motor functions, and elevation of inflammatory markers. Extensive work with this model by multiple laboratories over the past decade further increased confidence in its robustness and validity, especially for analyzing pathomechanisms of alpha-synuclein pathology and down-stream pathways, and for pre-clinical drug testing. Interestingly, while postnatal transgene expression is widespread in central and peripheral neurons, the extent and progression of down-stream pathology differs between brain regions, thereby replicating the characteristic selective vulnerability of neurodegenerative diseases. In-depth characterization of these readouts in conjunction with behavioral deficits has led to more informative endpoints for pre-clinical trials. Each drug tested in Thy1-aSyn line 61 enhances knowledge on how molecular targets, pathology, and functional behavioral readouts are interconnected, thereby further optimizing the platform towards predictive validity for clinical trials. Here, we present the current state of the art using Thy1-aSyn line 61 for drug target discovery, validation, and pre-clinical testing.
Collapse
Affiliation(s)
- Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany.
- Center for Systems Neuroscience Hannover, Hannover, Germany.
| | - Milos Stanojlovic
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
| | - Christopher Käufer
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
| | - Birthe Gericke
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
- Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Malte Feja
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
- Center for Systems Neuroscience Hannover, Hannover, Germany
| |
Collapse
|
23
|
A A, W C, N N, L M, M D, Zhang DD. α-Syn overexpression, NRF2 suppression, and enhanced ferroptosis create a vicious cycle of neuronal loss in Parkinson's disease. Free Radic Biol Med 2022; 192:130-140. [PMID: 36150560 PMCID: PMC9841923 DOI: 10.1016/j.freeradbiomed.2022.09.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/08/2022] [Accepted: 09/16/2022] [Indexed: 01/18/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder, affecting millions each year. Most PD cases (∼90%) are sporadic, resulting from the age-dependent accumulation of pathogenic effects. One key pathological hallmark of PD progression is the accumulation of alpha-synuclein (α-syn), which has been shown to negatively affect neuronal function and viability. Here, using 3- and 6-month-old Nrf2+/+ and Nrf2-/- mice overexpressing human α-syn (PD model), we show that loss of NRF2 increases markers of ferroptosis across PD-relevant brain regions. Increased ferroptosis was associated with an age- and genotype-dependent increase in α-syn pathology and behavioral deficits. Finally, we demonstrate that α-syn overexpression sensitizes neuronal cells and ex vivo brain slices to ferroptosis induction, which may be due to α-syn decreasing NRF2 protein levels. Altogether, these results indicate that NRF2 is a critical anti-ferroptotic mediator of neuronal survival, and that the vicious cycle of α-syn overexpression and NRF2 suppression, leading to enhanced neuronal ferroptotic cell death, could represent a targetable and currently untapped means of preventing PD onset and progression.
Collapse
Affiliation(s)
- Anandhan A
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Chen W
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Nguyen N
- Department of Neurology, University of Arizona, Tucson, AZ, USA
| | - Madhavan L
- Department of Neurology, University of Arizona, Tucson, AZ, USA; Evelyn F McKnight Brain Institute and Bio5 Institute, University of Arizona, Tucson, AZ, USA
| | - Dodson M
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA.
| | - D D Zhang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA; Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA.
| |
Collapse
|
24
|
Beserra-Filho JIA, Maria-Macêdo A, Silva-Martins S, Custódio-Silva AC, Soares-Silva B, Silva SP, Lambertucci RH, de Souza Araújo AA, Lucchese AM, Quintans-Júnior LJ, Santos JR, Silva RH, Ribeiro AM. Lippia grata essential oil complexed with β-cyclodextrin ameliorates biochemical and behavioral deficits in an animal model of progressive parkinsonism. Metab Brain Dis 2022; 37:2331-2347. [PMID: 35779151 DOI: 10.1007/s11011-022-01032-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 06/13/2022] [Indexed: 10/17/2022]
Abstract
Parkinson's disease (PD) is identified by the loss of dopaminergic neurons in the Substantia Nigra pars compacta (SNpc), and is correlated to aggregates of proteins such as α-synuclein, Lewy's bodies. Although the PD etiology remains poorly understood, evidence suggests a main role of oxidative stress on this process. Lippia grata Schauer, known as "alecrim-do-mato", "alecrim-de-vaqueiro", "alecrim-da-chapada", is a native bush from tropical areas mainly distributed throughout the Central and South America. This plant species is commonly used in traditional medicine for relief of pain and inflammation conditions, and that has proven antioxidant effects. We evaluated the effects of essential oil of the L. grata after its complexed with β-cyclodextrin (LIP) on PD animal model induced by reserpine (RES). Behavioral assessments were performed across the treatment. Upon completion the treatment, the animals were euthanized, afterwards their brains were isolated and processed for immunohistochemical and oxidative stress analysis. The LIP treatment delayed the onset of the behavior of catalepsy, decreased the number of oral movements and prevented the memory impairment on the novel object recognition task. In addition, the treatment with LIP protected against dopaminergic depletion in the SNpc and dorsal striatum (STRd), and decreased the α-syn immunoreactivity in the SNpc and hippocampus (HIP). Moreover, there was reduction of the oxidative stability index. These findings demonstrated that the LIP treatment has neuroprotective effect in a progressive parkinsonism model, suggesting that LIP could be an important source for novel treatment approaches in PD.
Collapse
Affiliation(s)
- Jose Ivo A Beserra-Filho
- Departament of Biosciences, Universidade Federal de São Paulo, Santos, São Paulo, Brazil
- Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Amanda Maria-Macêdo
- Departament of Biosciences, Universidade Federal de São Paulo, Santos, São Paulo, Brazil
| | - Suellen Silva-Martins
- Departament of Biosciences, Universidade Federal de São Paulo, Santos, São Paulo, Brazil
| | | | - Beatriz Soares-Silva
- Departament of Biosciences, Universidade Federal de São Paulo, Santos, São Paulo, Brazil
| | - Sara Pereira Silva
- Departament of Biosciences, Universidade Federal de São Paulo, Santos, São Paulo, Brazil
| | | | | | - Angélica Maria Lucchese
- Graduate Programm in Biotechnology, Universidade Estadual de Feira de Santana, Feira de Santana, Bahia, Brazil
| | | | - José Ronaldo Santos
- Department of Biosciences, Universidade Federal de Sergipe, Itabaiana, Sergipe, Brazil
| | - Regina H Silva
- Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Alessandra M Ribeiro
- Departament of Biosciences, Universidade Federal de São Paulo, Santos, São Paulo, Brazil.
| |
Collapse
|
25
|
Blok LER, Boon M, van Reijmersdal B, Höffler KD, Fenckova M, Schenck A. Genetics, molecular control and clinical relevance of habituation learning. Neurosci Biobehav Rev 2022; 143:104883. [PMID: 36152842 DOI: 10.1016/j.neubiorev.2022.104883] [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: 04/22/2022] [Revised: 08/08/2022] [Accepted: 08/30/2022] [Indexed: 11/29/2022]
Abstract
Habituation is the most fundamental form of learning. As a firewall that protects our brain from sensory overload, it is indispensable for cognitive processes. Studies in humans and animal models provide increasing evidence that habituation is affected in autism and related monogenic neurodevelopmental disorders (NDDs). An integrated application of habituation assessment in NDDs and their animal models has unexploited potential for neuroscience and medical care. With the aim to gain mechanistic insights, we systematically retrieved genes that have been demonstrated in the literature to underlie habituation. We identified 258 evolutionarily conserved genes across species, describe the biological processes they converge on, and highlight regulatory pathways and drugs that may alleviate habituation deficits. We also summarize current habituation paradigms and extract the most decisive arguments that support the crucial role of habituation for cognition in health and disease. We conclude that habituation is a conserved, quantitative, cognition- and disease-relevant process that can connect preclinical and clinical work, and hence is a powerful tool to advance research, diagnostics, and treatment of NDDs.
Collapse
Affiliation(s)
- Laura Elisabeth Rosalie Blok
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Marina Boon
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Boyd van Reijmersdal
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Kira Daniela Höffler
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Michaela Fenckova
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands; Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia in Ceske Budejovice, Branisovska 31, 37005, Ceske Budejovice, Czech Republic.
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| |
Collapse
|
26
|
Iba M, McDevitt RA, Kim C, Roy R, Sarantopoulou D, Tommer E, Siegars B, Sallin M, Kwon S, Sen JM, Sen R, Masliah E. Aging exacerbates the brain inflammatory micro-environment contributing to α-synuclein pathology and functional deficits in a mouse model of DLB/PD. Mol Neurodegener 2022; 17:60. [PMID: 36064424 PMCID: PMC9447339 DOI: 10.1186/s13024-022-00564-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 08/19/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Although ɑ-synuclein (ɑ-syn) spreading in age-related neurodegenerative diseases such as Parkinson's disease (PD) and Dementia with Lewy bodies (DLB) has been extensively investigated, the role of aging in the manifestation of disease remains unclear. METHODS We explored the role of aging and inflammation in the pathogenesis of synucleinopathies in a mouse model of DLB/PD initiated by intrastriatal injection of ɑ-syn preformed fibrils (pff). RESULTS We found that aged mice showed more extensive accumulation of ɑ-syn in selected brain regions and behavioral deficits that were associated with greater infiltration of T cells and microgliosis. Microglial inflammatory gene expression induced by ɑ-syn-pff injection in young mice had hallmarks of aged microglia, indicating that enhanced age-associated pathologies may result from inflammatory synergy between aging and the effects of ɑ-syn aggregation. Based on the transcriptomics analysis projected from Ingenuity Pathway Analysis, we found a network that included colony stimulating factor 2 (CSF2), LPS related genes, TNFɑ and poly rl:rC-RNA as common regulators. CONCLUSIONS We propose that aging related inflammation (eg: CSF2) influences outcomes of pathological spreading of ɑ-syn and suggest that targeting neuro-immune responses might be important in developing treatments for DLB/PD.
Collapse
Affiliation(s)
- Michiyo Iba
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ross A McDevitt
- Mouse Phenotyping Unit, Comparative Medicine Section, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Changyoun Kim
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Roshni Roy
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Dimitra Sarantopoulou
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Ella Tommer
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Byron Siegars
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Michelle Sallin
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Somin Kwon
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jyoti Misra Sen
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
- Immunology Program, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21224, USA
| | - Ranjan Sen
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
- Immunology Program, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21224, USA
| | - Eliezer Masliah
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA.
- Division of Neuroscience, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20814, USA.
| |
Collapse
|
27
|
Aniszewska A, Bergström J, Ingelsson M, Ekmark-Lewén S. Modeling Parkinson's disease-related symptoms in alpha-synuclein overexpressing mice. Brain Behav 2022; 12:e2628. [PMID: 35652155 PMCID: PMC9304846 DOI: 10.1002/brb3.2628] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/08/2022] [Accepted: 04/15/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Intracellular deposition of alpha-synuclein (α-syn) as Lewy bodies and Lewy neurites is a central event in the pathogenesis of Parkinson's disease (PD) and other α-synucleinopathies. Transgenic mouse models overexpressing human α-syn, are useful research tools in preclinical studies of pathogenetic mechanisms. Such mice develop α-syn inclusions as well as neurodegeneration with a topographical distribution that varies depending on the choice of promoter and which form of α-syn that is overexpressed. Moreover, they display motor symptoms and cognitive disturbances that to some extent resemble the human conditions. PURPOSE One of the main motives for assessing behavior in these mouse models is to evaluate the potential of new treatment strategies, including their impact on motor and cognitive symptoms. However, due to a high within-group variability with respect to such features, the behavioral studies need to be applied with caution. In this review, we discuss how to make appropriate choices in the experimental design and which tests that are most suitable for the evaluation of PD-related symptoms in such studies. METHODS We have evaluated published results on two selected transgenic mouse models overexpressing wild type (L61) and mutated (A30P) α-syn in the context of their validity and utility for different types of behavioral studies. CONCLUSIONS By applying appropriate behavioral tests, α-syn transgenic mouse models provide an appropriate experimental platform for studies of symptoms related to PD and other α-synucleinopathies.
Collapse
Affiliation(s)
- Agata Aniszewska
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Joakim Bergström
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden.,Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada.,Department of Medicine and Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Ontario, Canada
| | - Sara Ekmark-Lewén
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| |
Collapse
|
28
|
Rauschenberger L, Behnke J, Grotemeyer A, Knorr S, Volkmann J, Ip CW. Age-dependent neurodegeneration and neuroinflammation in a genetic A30P/A53T double-mutated α-synuclein mouse model of Parkinson's disease. Neurobiol Dis 2022; 171:105798. [PMID: 35750147 DOI: 10.1016/j.nbd.2022.105798] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/31/2022] [Accepted: 06/17/2022] [Indexed: 11/28/2022] Open
Abstract
The pathogenesis of Parkinson's disease (PD) is closely interwoven with the process of aging. Moreover, increasing evidence from human postmortem studies and from animal models for PD point towards inflammation as an additional factor in disease development. We here assessed the impact of aging and inflammation on dopaminergic neurodegeneration in the hm2α-SYN-39 mouse model of PD that carries the human, A30P/A53T double-mutated α-synuclein gene. At 2-3 months of age, no significant differences were observed comparing dopaminergic neuron numbers of the substantia nigra (SN) pars compacta of hm2α-SYN-39 mice with wildtype controls. At an age of 16-17 months, however, hm2α-SYN-39 mice revealed a significant loss of dopaminergic SN neurons, of dopaminergic terminals in the striatum as well as a reduction of striatal dopamine levels compared to young, 2-3 months transgenic mice and compared to 16-17 months old wildtype littermates. A significant age-related correlation of infiltrating CD4+ and CD8+ T cell numbers with dopaminergic terminal loss of the striatum was found in hm2α-SYN-39 mice, but not in wildtype controls. In the striatum of 16-17 months old wildtype mice a slightly elevated CD8+ T cell count and CD11b+ microglia cell count was observed compared to younger aged mice. Additional analyses of neuroinflammation in the nigrostriatal tract of wildtype mice did not yield any significant age-dependent changes of CD4+, CD8+ T cell and B220+ B cell numbers, respectively. In contrast, a significant age-dependent increase of CD8+ T cells, GFAP+ astrocytes as well as a pronounced increase of CD11b+ microglia numbers were observed in the SN of hm2α-SYN-39 mice pointing towards a neuroinflammatory processes in this genetic mouse model for PD. The findings in the hm2α-SYN-39 mouse model strengthen the evidence that T cell and glial cell responses are involved in the age-related neurodegeneration in PD. The slow and age-dependent progression of neurodegeneration and neuroinflammation in the hm2α-SYN-39 PD rodent model underlines its translational value and makes it suitable for studying anti-inflammatory therapies.
Collapse
Affiliation(s)
- Lisa Rauschenberger
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Jennifer Behnke
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Alexander Grotemeyer
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Susanne Knorr
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Jens Volkmann
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Chi Wang Ip
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany.
| |
Collapse
|
29
|
Cocaine Modulates the Neuronal Endosomal System and Extracellular Vesicles in a Sex-Dependent Manner. Neurochem Res 2022; 47:2263-2277. [PMID: 35501523 PMCID: PMC9352616 DOI: 10.1007/s11064-022-03612-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/15/2022] [Accepted: 04/16/2022] [Indexed: 01/08/2023]
Abstract
In multiple neurodevelopmental and neurodegenerative disorders, endosomal changes correlate with changes in exosomes. We examined this linkage in the brain of mice that received cocaine injections for two weeks starting at 2.5 months of age. Cocaine caused a decrease in the number of both neuronal early and late endosomes and exosomes in the brains of male but not female mice. The response to cocaine in ovariectomized females mirrored male, demonstrating that these sex-differences in response to cocaine are driven by hormonal differences. Moreover, cocaine increased the amount of α-synuclein per exosome in the brain of females but did not affect exosomal α-synuclein content in the brain of males, a sex-difference eliminated by ovariectomy. Enhanced packaging of α-synuclein into female brain exosomes with the potential for propagation of pathology throughout the brain suggests a mechanism for the different response of females to chronic cocaine exposure as compared to males.
Collapse
|
30
|
Glycation modulates glutamatergic signaling and exacerbates Parkinson's disease-like phenotypes. NPJ Parkinsons Dis 2022; 8:51. [PMID: 35468899 PMCID: PMC9038780 DOI: 10.1038/s41531-022-00314-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/31/2022] [Indexed: 01/17/2023] Open
Abstract
Alpha-synuclein (aSyn) is a central player in the pathogenesis of synucleinopathies due to its accumulation in typical protein aggregates in the brain. However, it is still unclear how it contributes to neurodegeneration. Type-2 diabetes mellitus is a risk factor for Parkinson's disease (PD). Interestingly, a common molecular alteration among these disorders is the age-associated increase in protein glycation. We hypothesized that glycation-induced neuronal dysfunction is a contributing factor in synucleinopathies. Here, we dissected the impact of methylglyoxal (MGO, a glycating agent) in mice overexpressing aSyn in the brain. We found that MGO-glycation potentiates motor, cognitive, olfactory, and colonic dysfunction in aSyn transgenic (Thy1-aSyn) mice that received a single dose of MGO via intracerebroventricular injection. aSyn accumulates in the midbrain, striatum, and prefrontal cortex, and protein glycation is increased in the cerebellum and midbrain. SWATH mass spectrometry analysis, used to quantify changes in the brain proteome, revealed that MGO mainly increase glutamatergic-associated proteins in the midbrain (NMDA, AMPA, glutaminase, VGLUT and EAAT1), but not in the prefrontal cortex, where it mainly affects the electron transport chain. The glycated proteins in the midbrain of MGO-injected Thy1-aSyn mice strongly correlate with PD and dopaminergic pathways. Overall, we demonstrated that MGO-induced glycation accelerates PD-like sensorimotor and cognitive alterations and suggest that the increase of glutamatergic signaling may underly these events. Our study sheds new light into the enhanced vulnerability of the midbrain in PD-related synaptic dysfunction and suggests that glycation suppressors and anti-glutamatergic drugs may hold promise as disease-modifying therapies for synucleinopathies.
Collapse
|
31
|
Age-dependent alterations in key components of the nigrostriatal dopaminergic system and distinct motor phenotypes. Acta Pharmacol Sin 2022; 43:862-875. [PMID: 34244603 PMCID: PMC8975991 DOI: 10.1038/s41401-021-00713-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 06/06/2021] [Indexed: 02/06/2023] Open
Abstract
The nigrostriatal dopaminergic (DA) system, which includes DA neurons in the ventral and dorsal tiers of the substantia nigra pars compacta (vSNc, dSNc) and DA terminals in the dorsal striatum, is critically implicated in motor control. Accumulating studies demonstrate that both the nigrostriatal DA system and motor function are impaired in aged subjects. However, it is unknown whether dSNc and vSNc DA neurons and striatal DA terminals age in similar patterns, and whether these changes parallel motor deficits. To address this, we performed ex vivo patch-clamp recordings in dSNc and vSNc DA neurons, measured striatal dopamine release, and analyzed motor behaviors in rodents. Spontaneous firing in dSNc and vSNc DA neurons and depolarization-evoked firing in dSNc DA neurons showed inverse V-shaped changes with age. But depolarization-evoked firing in vSNc DA neurons increased with age. In the dorsal striatum, dopamine release declined with age. In locomotor tests, 12-month-old rodents showed hyperactive exploration, relative to 6- and 24-month-old rodents. Additionally, aged rodents showed significant deficits in coordination. Elevating dopamine levels with a dopamine transporter inhibitor improved both locomotion and coordination. Therefore, key components in the nigrostriatal DA system exhibit distinct aging patterns and may contribute to age-related alterations in locomotion and coordination.
Collapse
|
32
|
Merino-Galan L, Jimenez-Urbieta H, Zamarbide M, Rodríguez-Chinchilla T, Belloso-Iguerategui A, Santamaria E, Fernández-Irigoyen J, Aiastui A, Doudnikoff E, Bézard E, Ouro A, Knafo S, Gago B, Quiroga-Varela A, Rodríguez-Oroz MC. Striatal synaptic bioenergetic and autophagic decline in premotor experimental parkinsonism. Brain 2022; 145:2092-2107. [PMID: 35245368 PMCID: PMC9460676 DOI: 10.1093/brain/awac087] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/31/2022] [Accepted: 02/20/2022] [Indexed: 12/02/2022] Open
Abstract
Synaptic impairment might precede neuronal degeneration in Parkinson’s disease. However, the intimate mechanisms altering synaptic function by the accumulation of presynaptic α-synuclein in striatal dopaminergic terminals before dopaminergic death occurs, have not been elucidated. Our aim is to unravel the sequence of synaptic functional and structural changes preceding symptomatic dopaminergic cell death. As such, we evaluated the temporal sequence of functional and structural changes at striatal synapses before parkinsonian motor features appear in a rat model of progressive dopaminergic death induced by overexpression of the human mutated A53T α-synuclein in the substantia nigra pars compacta, a protein transported to these synapses. Sequential window acquisition of all theoretical mass spectra proteomics identified deregulated proteins involved first in energy metabolism and later, in vesicle cycling and autophagy. After protein deregulation and when α-synuclein accumulated at striatal synapses, alterations to mitochondrial bioenergetics were observed using a Seahorse XF96 analyser. Sustained dysfunctional mitochondrial bioenergetics was followed by a decrease in the number of dopaminergic terminals, morphological and ultrastructural alterations, and an abnormal accumulation of autophagic/endocytic vesicles inside the remaining dopaminergic fibres was evident by electron microscopy. The total mitochondrial population remained unchanged whereas the number of ultrastructurally damaged mitochondria increases as the pathological process evolved. We also observed ultrastructural signs of plasticity within glutamatergic synapses before the expression of motor abnormalities, such as a reduction in axospinous synapses and an increase in perforated postsynaptic densities. Overall, we found that a synaptic energetic failure and accumulation of dysfunctional organelles occur sequentially at the dopaminergic terminals as the earliest events preceding structural changes and cell death. We also identify key proteins involved in these earliest functional abnormalities that may be modulated and serve as therapeutic targets to counterbalance the degeneration of dopaminergic cells to delay or prevent the development of Parkinson’s disease.
Collapse
Affiliation(s)
- Leyre Merino-Galan
- Neuroscience Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, 31008 Pamplona, Spain.,Neuroscience Department, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Haritz Jimenez-Urbieta
- Cell culture Platform, Biodonostia Health Research Institute, San Sebastian, 20014 Donostia, Spain
| | - Marta Zamarbide
- Neuroscience Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, 31008 Pamplona, Spain
| | | | | | - Enrique Santamaria
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Ana Aiastui
- Cell culture Platform, Biodonostia Health Research Institute, San Sebastian, 20014 Donostia, Spain
| | - Evelyne Doudnikoff
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33076 Bordeaux, France
| | - Erwan Bézard
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33076 Bordeaux, France
| | - Alberto Ouro
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Shira Knafo
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The National Institute for Biotechnology in the Negev, and The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, 8410501 Beer-Sheva, Israel.,Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Basque Foundation for Science, IKERBASQUE, 48940 Leioa, Spain
| | - Belén Gago
- Faculty of Medicine, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29016 Málaga, Spain
| | - Ana Quiroga-Varela
- Neuroscience Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, 31008 Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - María Cruz Rodríguez-Oroz
- Neuroscience Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, 31008 Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,Neurology Department, Clínica Universidad de Navarra (CUN), 31008 Pamplona, Spain
| |
Collapse
|
33
|
Bancroft EA, Srinivasan R. Emerging Roles for Aberrant Astrocytic Calcium Signals in Parkinson's Disease. Front Physiol 2022; 12:812212. [PMID: 35087422 PMCID: PMC8787054 DOI: 10.3389/fphys.2021.812212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/09/2021] [Indexed: 11/25/2022] Open
Abstract
Astrocytes display a plethora of spontaneous Ca2+ signals that modulate vital functions of the central nervous system (CNS). This suggests that astrocytic Ca2+ signals also contribute to pathological processes in the CNS. In this context, the molecular mechanisms by which aberrant astrocytic Ca2+ signals trigger dopaminergic neuron loss during Parkinson's disease (PD) are only beginning to emerge. Here, we provide an evidence-based perspective on potential mechanisms by which aberrant astrocytic Ca2+ signals can trigger dysfunction in three distinct compartments of the brain, viz., neurons, microglia, and the blood brain barrier, thereby leading to PD. We envision that the coming decades will unravel novel mechanisms by which aberrant astrocytic Ca2+ signals contribute to PD and other neurodegenerative processes in the CNS.
Collapse
Affiliation(s)
- Eric A. Bancroft
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, United States
| | - Rahul Srinivasan
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, United States
- Texas A&M Institute for Neuroscience (TAMIN), College Station, TX, United States
| |
Collapse
|
34
|
Pirooznia SK, Rosenthal LS, Dawson VL, Dawson TM. Parkinson Disease: Translating Insights from Molecular Mechanisms to Neuroprotection. Pharmacol Rev 2021; 73:33-97. [PMID: 34663684 DOI: 10.1124/pharmrev.120.000189] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson disease (PD) used to be considered a nongenetic condition. However, the identification of several autosomal dominant and recessive mutations linked to monogenic PD has changed this view. Clinically manifest PD is then thought to occur through a complex interplay between genetic mutations, many of which have incomplete penetrance, and environmental factors, both neuroprotective and increasing susceptibility, which variably interact to reach a threshold over which PD becomes clinically manifested. Functional studies of PD gene products have identified many cellular and molecular pathways, providing crucial insights into the nature and causes of PD. PD originates from multiple causes and a range of pathogenic processes at play, ultimately culminating in nigral dopaminergic loss and motor dysfunction. An in-depth understanding of these complex and possibly convergent pathways will pave the way for therapeutic approaches to alleviate the disease symptoms and neuroprotective strategies to prevent disease manifestations. This review is aimed at providing a comprehensive understanding of advances made in PD research based on leveraging genetic insights into the pathogenesis of PD. It further discusses novel perspectives to facilitate identification of critical molecular pathways that are central to neurodegeneration that hold the potential to develop neuroprotective and/or neurorestorative therapeutic strategies for PD. SIGNIFICANCE STATEMENT: A comprehensive review of PD pathophysiology is provided on the complex interplay of genetic and environmental factors and biologic processes that contribute to PD pathogenesis. This knowledge identifies new targets that could be leveraged into disease-modifying therapies to prevent or slow neurodegeneration in PD.
Collapse
Affiliation(s)
- Sheila K Pirooznia
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Liana S Rosenthal
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| |
Collapse
|
35
|
Torres ERS, Stanojlovic M, Zelikowsky M, Bonsberger J, Hean S, Mulligan C, Baldauf L, Fleming S, Masliah E, Chesselet MF, Fanselow MS, Richter F. Alpha-synuclein pathology, microgliosis, and parvalbumin neuron loss in the amygdala associated with enhanced fear in the Thy1-aSyn model of Parkinson's disease. Neurobiol Dis 2021; 158:105478. [PMID: 34390837 PMCID: PMC8447919 DOI: 10.1016/j.nbd.2021.105478] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/20/2021] [Accepted: 08/09/2021] [Indexed: 11/17/2022] Open
Abstract
In Parkinson's disease (PD), the second most common neurodegenerative disorder, non-motor symptoms often precede the development of debilitating motor symptoms and present a severe impact on the quality of life. Lewy bodies containing misfolded α-synuclein progressively develop in neurons throughout the peripheral and central nervous system, which may be correlated with the early development of non-motor symptoms. Among those, increased fear and anxiety is frequent in PD and thought to result from pathology outside the dopaminergic system, which has been the focus of symptomatic treatment to alleviate motor symptoms. Alpha-synuclein accumulation has been reported in the amygdala of PD patients, a brain region critically involved in fear and anxiety. Here we asked whether α-synuclein overexpression alone is sufficient to induce an enhanced fear phenotype in vivo and which pathological mechanisms are involved. Transgenic mice expressing human wild-type α-synuclein (Thy1-aSyn), a well-established model of PD, were subjected to fear conditioning followed by extinction and then tested for extinction memory retention followed by histopathological analysis. Thy1-aSyn mice showed enhanced tone fear across acquisition and extinction compared to wild-type littermates, as well as a trend to less retention of fear extinction. Immunohistochemical analysis of the basolateral nucleus of the amygdala, a nucleus critically involved in tone fear learning, revealed extensive α-synuclein pathology, with accumulation, phosphorylation, and aggregation of α-synuclein in transgenic mice. This pathology was accompanied by microgliosis and parvalbumin neuron loss in this nucleus, which could explain the enhanced fear phenotype. Importantly, this non-motor phenotype was detected in the pre-clinical phase, prior to dopamine loss in Thy1-aSyn mice, thus replicating observations in patients. Results obtained in this study suggest a possible mechanism by which increased anxiety and maladaptive fear processing may occur in PD, opening a door for therapeutic options and further early biomarker research.
Collapse
Affiliation(s)
- Eileen Ruth S Torres
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Milos Stanojlovic
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Moriel Zelikowsky
- Department of Psychology, Staglin Center for Brain and Behavioral Health, UCLA, Los Angeles, CA 90095, USA; Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Jana Bonsberger
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Sindalana Hean
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Caitlin Mulligan
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Leonie Baldauf
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Sheila Fleming
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Eliezer Masliah
- Department of Neurosciences, UCSD School of Medicine, La Jolla, CA 92093, USA
| | | | - Michael S Fanselow
- Department of Psychology, Staglin Center for Brain and Behavioral Health, UCLA, Los Angeles, CA 90095, USA
| | - Franziska Richter
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, Hanover, Germany.
| |
Collapse
|
36
|
Dagra A, Miller DR, Lin M, Gopinath A, Shaerzadeh F, Harris S, Sorrentino ZA, Støier JF, Velasco S, Azar J, Alonge AR, Lebowitz JJ, Ulm B, Bu M, Hansen CA, Urs N, Giasson BI, Khoshbouei H. α-Synuclein-induced dysregulation of neuronal activity contributes to murine dopamine neuron vulnerability. NPJ Parkinsons Dis 2021; 7:76. [PMID: 34408150 PMCID: PMC8373893 DOI: 10.1038/s41531-021-00210-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 07/09/2021] [Indexed: 02/07/2023] Open
Abstract
Pathophysiological damages and loss of function of dopamine neurons precede their demise and contribute to the early phases of Parkinson's disease. The presence of aberrant intracellular pathological inclusions of the protein α-synuclein within ventral midbrain dopaminergic neurons is one of the cardinal features of Parkinson's disease. We employed molecular biology, electrophysiology, and live-cell imaging to investigate how excessive α-synuclein expression alters multiple characteristics of dopaminergic neuronal dynamics and dopamine transmission in cultured dopamine neurons conditionally expressing GCaMP6f. We found that overexpression of α-synuclein in mouse (male and female) dopaminergic neurons altered neuronal firing properties, calcium dynamics, dopamine release, protein expression, and morphology. Moreover, prolonged exposure to the D2 receptor agonist, quinpirole, rescues many of the alterations induced by α-synuclein overexpression. These studies demonstrate that α-synuclein dysregulation of neuronal activity contributes to the vulnerability of dopaminergic neurons and that modulation of D2 receptor activity can ameliorate the pathophysiology. These findings provide mechanistic insights into the insidious changes in dopaminergic neuronal activity and neuronal loss that characterize Parkinson's disease progression with significant therapeutic implications.
Collapse
Affiliation(s)
- Abeer Dagra
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Douglas R. Miller
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Min Lin
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Adithya Gopinath
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Fatemeh Shaerzadeh
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Sharonda Harris
- grid.15276.370000 0004 1936 8091Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL USA
| | - Zachary A. Sorrentino
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Jonatan Fullerton Støier
- grid.5254.60000 0001 0674 042XMolecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sophia Velasco
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Janelle Azar
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Adetola R. Alonge
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Joseph J. Lebowitz
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Brittany Ulm
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Mengfei Bu
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Carissa A. Hansen
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Nikhil Urs
- grid.15276.370000 0004 1936 8091Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL USA
| | - Benoit I. Giasson
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Habibeh Khoshbouei
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| |
Collapse
|
37
|
Pan PY, Sheehan P, Wang Q, Zhu X, Zhang Y, Choi I, Li X, Saenz J, Zhu J, Wang J, El Gaamouch F, Zhu L, Cai D, Yue Z. Synj1 haploinsufficiency causes dopamine neuron vulnerability and alpha-synuclein accumulation in mice. Hum Mol Genet 2021; 29:2300-2312. [PMID: 32356558 DOI: 10.1093/hmg/ddaa080] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/24/2020] [Accepted: 04/24/2020] [Indexed: 12/31/2022] Open
Abstract
Synaptojanin1 (synj1) is a phosphoinositide phosphatase with dual SAC1 and 5'-phosphatase enzymatic activities in regulating phospholipid signaling. The brain-enriched isoform has been shown to participate in synaptic vesicle (SV) recycling. More recently, recessive human mutations were identified in the two phosphatase domains of SYNJ1, including R258Q, R459P and R839C, which are linked to rare forms of early-onset Parkinsonism. We now demonstrate that Synj1 heterozygous deletion (Synj1+/-), which is associated with an impaired 5'-phosphatase activity, also leads to Parkinson's disease (PD)-like pathologies in mice. We report that male Synj1+/- mice display age-dependent motor function abnormalities as well as alpha-synuclein accumulation, impaired autophagy and dopaminergic terminal degeneration. Synj1+/- mice contain elevated 5'-phosphatase substrate, PI(4,5)P2, particularly in the midbrain neurons. Moreover, pharmacological elevation of membrane PI(4,5)P2 in cultured neurons impairs SV endocytosis, specifically in midbrain neurons, and further exacerbates SV trafficking defects in Synj1+/- midbrain neurons. We demonstrate down-regulation of SYNJ1 transcript in a subset of sporadic PD brains, implicating a potential role of Synj1 deficiency in the decline of dopaminergic function during aging.
Collapse
Affiliation(s)
- Ping-Yue Pan
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854 USA
| | - Patricia Sheehan
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Qian Wang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Xinyu Zhu
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854 USA
| | - Yuanxi Zhang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Insup Choi
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Xianting Li
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Jacqueline Saenz
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854 USA
| | - Justin Zhu
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854 USA
| | - Jing Wang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Farida El Gaamouch
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,James J Peters VA Medical Center, Research & Development, Bronx, NY 10468 USA
| | - Li Zhu
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,James J Peters VA Medical Center, Research & Development, Bronx, NY 10468 USA
| | - Dongming Cai
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,James J Peters VA Medical Center, Research & Development, Bronx, NY 10468 USA
| | - Zhenyu Yue
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.,The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| |
Collapse
|
38
|
Anandhan A, Nguyen N, Syal A, Dreher LA, Dodson M, Zhang DD, Madhavan L. NRF2 Loss Accentuates Parkinsonian Pathology and Behavioral Dysfunction in Human α-Synuclein Overexpressing Mice. Aging Dis 2021; 12:964-982. [PMID: 34221542 PMCID: PMC8219498 DOI: 10.14336/ad.2021.0511] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 05/11/2021] [Indexed: 12/14/2022] Open
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (NRF2) is a central regulator of cellular stress responses and its transcriptional activation promotes multiple cellular defense and survival mechanisms. The loss of NRF2 has been shown to increase oxidative and proteotoxic stress, two key pathological features of neurodegenerative disorders such as Parkinson's disease (PD). Moreover, compromised redox homeostasis and protein quality control can cause the accumulation of pathogenic proteins, including alpha-synuclein (α-Syn) which plays a key role in PD. However, despite this link, the precise mechanisms by which NRF2 may regulate PD pathology is not clear. In this study, we generated a humanized mouse model to study the importance of NRF2 in the context of α-Syn-driven neuropathology in PD. Specifically, we developed NRF2 knockout and wild-type mice that overexpress human α-Syn (hα-Syn+/Nrf2-/- and hα-Syn+/Nrf2+/+ respectively) and tested changes in their behavior through nest building, challenging beam, and open field tests at three months of age. Cellular and molecular alterations in α-Syn, including phosphorylation and subsequent oligomerization, as well as changes in oxidative stress, inflammation, and autophagy were also assessed across multiple brain regions. It was observed that although monomeric α-Syn levels did not change, compared to their wild-type counterparts, hα-Syn+/Nrf2-/- mice exhibited increased phosphorylation and oligomerization of α-Syn. This was associated with a loss of tyrosine hydroxylase expressing dopaminergic neurons in the substantia nigra, and more pronounced behavioral deficits reminiscent of early-stage PD, in the hα-Syn+/Nrf2-/- mice. Furthermore, hα-Syn+/Nrf2-/- mice showed significantly amplified oxidative stress, greater expression of inflammatory markers, and signs of increased autophagic burden, especially in the midbrain, striatum and cortical brain regions. These results support an important role for NRF2, early in PD progression. More broadly, it indicates NRF2 biology as fundamental to PD pathogenesis and suggests that targeting NRF2 activation may delay the onset and progression of PD.
Collapse
Affiliation(s)
- Annadurai Anandhan
- Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA.
- Department of Neurology, University of Arizona, Tucson, AZ, USA.
| | - Nhat Nguyen
- Physiology Undergraduate Program, Tucson, AZ, USA.
| | - Arjun Syal
- Neuroscience and Cognitive Science Undergraduate Program, Tucson, AZ, USA.
| | - Luke A Dreher
- Ecology and Evolutionary Biology Undergraduate Program, Tucson, AZ, USA.
| | - Matthew Dodson
- Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA.
| | - Donna D Zhang
- Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA.
| | - Lalitha Madhavan
- Department of Neurology, University of Arizona, Tucson, AZ, USA.
- Evelyn F McKnight Brain Institute and Bio5 Institute, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
39
|
Huntington TE, Srinivasan R. Adeno-Associated Virus Expression of α-Synuclein as a Tool to Model Parkinson's Disease: Current Understanding and Knowledge Gaps. Aging Dis 2021; 12:1120-1137. [PMID: 34221553 PMCID: PMC8219504 DOI: 10.14336/ad.2021.0517] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/16/2021] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder in the aging population and is characterized by a constellation of motor and non-motor symptoms. The abnormal aggregation and spread of alpha-synuclein (α-syn) is thought to underlie the loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc), leading to the development of PD. It is in this context that the use of adeno-associated viruses (AAVs) to express a-syn in the rodent midbrain has become a popular tool to model SNc DA neuron loss during PD. In this review, we summarize results from two decades of experiments using AAV-mediated a-syn expression in rodents to model PD. Specifically, we outline aspects of AAV vectors that are particularly relevant to modeling a-syn dysfunction in rodent models of PD such as changes in striatal neurochemistry, a-syn biochemistry, and PD-related behaviors resulting from AAV-mediated a-syn expression in the midbrain. Finally, we discuss the emerging role of astrocytes in propagating a-syn pathology, and point to future directions for employing AAVs as a tool to better understand how astrocytes contribute to a-syn pathology during the development of PD. We envision that lessons learned from two decades of utilizing AAVs to express a-syn in the rodent brain will enable us to develop an optimized set of parameters for gaining a better understanding of how a-syn leads to the development of PD.
Collapse
Affiliation(s)
- Taylor E Huntington
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University College of Medicine, 8447 Riverside Pkwy, Bryan, TX 77807, USA.
- Texas A&M Institute for Neuroscience (TAMIN), College Station, TX 77843, USA
| | - Rahul Srinivasan
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University College of Medicine, 8447 Riverside Pkwy, Bryan, TX 77807, USA.
- Texas A&M Institute for Neuroscience (TAMIN), College Station, TX 77843, USA
| |
Collapse
|
40
|
Ferreira N, Gram H, Sorrentino ZA, Gregersen E, Schmidt SI, Reimer L, Betzer C, Perez-Gozalbo C, Beltoja M, Nagaraj M, Wang J, Nowak JS, Dong M, Willén K, Cholak E, Bjerregaard-Andersen K, Mendez N, Rabadia P, Shahnawaz M, Soto C, Otzen DE, Akbey Ü, Meyer M, Giasson BI, Romero-Ramos M, Jensen PH. Multiple system atrophy-associated oligodendroglial protein p25α stimulates formation of novel α-synuclein strain with enhanced neurodegenerative potential. Acta Neuropathol 2021; 142:87-115. [PMID: 33978813 PMCID: PMC8217051 DOI: 10.1007/s00401-021-02316-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 01/08/2023]
Abstract
Pathology consisting of intracellular aggregates of alpha-Synuclein (α-Syn) spread through the nervous system in a variety of neurodegenerative disorders including Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. The discovery of structurally distinct α-Syn polymorphs, so-called strains, supports a hypothesis where strain-specific structures are templated into aggregates formed by native α-Syn. These distinct strains are hypothesised to dictate the spreading of pathology in the tissue and the cellular impact of the aggregates, thereby contributing to the variety of clinical phenotypes. Here, we present evidence of a novel α-Syn strain induced by the multiple system atrophy-associated oligodendroglial protein p25α. Using an array of biophysical, biochemical, cellular, and in vivo analyses, we demonstrate that compared to α-Syn alone, a substoichiometric concentration of p25α redirects α-Syn aggregation into a unique α-Syn/p25α strain with a different structure and enhanced in vivo prodegenerative properties. The α-Syn/p25α strain induced larger inclusions in human dopaminergic neurons. In vivo, intramuscular injection of preformed fibrils (PFF) of the α-Syn/p25α strain compared to α-Syn PFF resulted in a shortened life span and a distinct anatomical distribution of inclusion pathology in the brain of a human A53T transgenic (line M83) mouse. Investigation of α-Syn aggregates in brain stem extracts of end-stage mice demonstrated that the more aggressive phenotype of the α-Syn/p25α strain was associated with an increased load of α-Syn aggregates based on a Förster resonance energy transfer immunoassay and a reduced α-Syn aggregate seeding activity based on a protein misfolding cyclic amplification assay. When injected unilaterally into the striata of wild-type mice, the α-Syn/p25α strain resulted in a more-pronounced motoric phenotype than α-Syn PFF and exhibited a "tropism" for nigro-striatal neurons compared to α-Syn PFF. Overall, our data support a hypothesis whereby oligodendroglial p25α is responsible for generating a highly prodegenerative α-Syn strain in multiple system atrophy.
Collapse
|
41
|
Khan A, Johnson R, Wittmer C, Maile M, Tatsukawa K, Wong JL, Gill MB, Stocking EM, Natala SR, Paulino AD, Bowden-Verhoek JK, Wrasidlo W, Masliah E, Bonhaus DW, Price DL. NPT520-34 improves neuropathology and motor deficits in a transgenic mouse model of Parkinson's disease. Brain 2021; 144:3692-3709. [PMID: 34117864 DOI: 10.1093/brain/awab214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/12/2021] [Accepted: 05/11/2021] [Indexed: 12/09/2022] Open
Abstract
NPT520-34 is a clinical-stage, small molecule being developed for the treatment of Parkinson's disease and other neurodegenerative disorders. The therapeutic potential of NPT520-34 was first suggested by findings from cell-based assays of alpha-synuclein (ASYN) clearance. As reported here, NPT520-34 was subsequently evaluated for therapeutically relevant actions in a transgenic animal model of Parkinson's disease that overexpresses human ASYN and in an acute lipopolysaccharide (LPS)-challenge model using wild-type mice. Daily administration of NPT520-34 to mThy1-ASYN (Line 61) transgenic mice for one or three months resulted in reduced ASYN pathology, reduced expression of markers of neuroinflammation, and improvements in multiple indices of motor function. In an LPS-challenge model using wild-type mice, a single-dose of NPT520-34 reduced LPS-evoked increases in the expression of several pro-inflammatory cytokines in plasma. These findings demonstrate the beneficial effects of NPT520-34 on both inflammation and protein-pathology endpoints, with consequent improvements in motor function in an animal model of Parkinson's disease. These findings further suggest that NPT520-34 may have two complementary actions: (1) to increase the clearance of neurotoxic protein aggregates and (2) to directly attenuate inflammation. NPT520-34 treatment may thereby address two of the predominate underlying pathophysiological aspects of neurodegenerative disorders such as Parkinson's disease.
Collapse
Affiliation(s)
- Asma Khan
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Robert Johnson
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Carrie Wittmer
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Michelle Maile
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Keith Tatsukawa
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Julian L Wong
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Martin B Gill
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Emily M Stocking
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Srinivasa R Natala
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Amy D Paulino
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Jon K Bowden-Verhoek
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Wolfgang Wrasidlo
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Eliezer Masliah
- Departments of Neuroscience and Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Douglas W Bonhaus
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Diana L Price
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| |
Collapse
|
42
|
Lin M, Mackie PM, Shaerzadeh F, Gamble-George J, Miller DR, Martyniuk CJ, Khoshbouei H. In Parkinson's patient-derived dopamine neurons, the triplication of α-synuclein locus induces distinctive firing pattern by impeding D2 receptor autoinhibition. Acta Neuropathol Commun 2021; 9:107. [PMID: 34099060 PMCID: PMC8185945 DOI: 10.1186/s40478-021-01203-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
Pathophysiological changes in dopamine neurons precede their demise and contribute to the early phases of Parkinson's disease (PD). Intracellular pathological inclusions of the protein α-synuclein within dopaminergic neurons are a cardinal feature of PD, but the mechanisms by which α-synuclein contributes to dopaminergic neuron vulnerability remain unknown. The inaccessibility to diseased tissue has been a limitation in studying progression of pathophysiology prior to degeneration of dopamine neurons. To address these issues, we differentiated induced pluripotent stem cells (iPSCs) from a PD patient carrying the α-synuclein triplication mutation (AST) and an unaffected first-degree relative (NAS) into dopaminergic neurons. In human-like dopamine neurons α-synuclein overexpression reduced the functional availability of D2 receptors, resulting in a stark dysregulation in firing activity, dopamine release, and neuronal morphology. We back-translated these findings into primary mouse neurons overexpressing α-synuclein and found a similar phenotype, supporting the causal role for α-synuclein. Importantly, application of D2 receptor agonist, quinpirole, restored the altered firing activity of AST-derived dopaminergic neurons to normal levels. These results provide novel insights into the pre-degenerative pathophysiological neuro-phenotype induced by α-synuclein overexpression and introduce a potential mechanism for the long-established clinical efficacy of D2 receptor agonists in the treatment of PD.
Collapse
Affiliation(s)
- Min Lin
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Phillip M Mackie
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Fatima Shaerzadeh
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | | | - Douglas R Miller
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Chris J Martyniuk
- Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
43
|
McGregor BA, Schommer J, Guo K, Raihan MO, Ghribi O, Hur J, Porter JE. Alpha-Synuclein-induced DNA Methylation and Gene Expression in Microglia. Neuroscience 2021; 468:186-198. [PMID: 34082066 DOI: 10.1016/j.neuroscience.2021.05.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 12/18/2022]
Abstract
Synucleinopathy disorders are characterized by aggregates of α-synuclein (α-syn), which engage microglia to elicit a neuroinflammatory response. Here, we determined the gene expression and DNA methylation changes in microglia induced by aggregate α-syn. Transgenic murine Thy-1 promoter (mThy1)-Asyn mice overexpressing human α-syn are a model of synucleinopathy. Microglia from 3 and 13-month-old mice were used to isolate nucleic acids for methylated DNA and RNA-sequencing. α-Syn-regulated changes in gene expression and genomic methylation were determined and examined for functional enrichment followed by network analysis to further elucidate possible connections within the data. Microglial DNA isolated from our 3-month cohort had 5315 differentially methylated gene (DMG) changes, while RNA levels demonstrated a change in 119 differentially expressed genes (DEGs) between mThy1-Asyn mice and wild-type littermate controls. The 3-month DEGs and DMGs were highly associated with adhesion and migration signaling, suggesting a phenotypic transition from resting to active microglia. We observed 3742 DMGs and 3766 DEGs in 13-month mThy1-Asyn mice. These genes were often related to adhesion, migration, cell cycle, cellular metabolism, and immune response. Network analysis also showed increased cell mobility and inflammatory functions at 3 months, shifting to cell cycle, immune response, and metabolism changes at 13 months. We observed significant α-syn-induced methylation and gene expression changes in microglia. Our data suggest that α-syn overexpression initiates microglial activation leading to neuroinflammation and cellular metabolic stresses, which is associated with disease progression.
Collapse
Affiliation(s)
- Brett A McGregor
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Jared Schommer
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Kai Guo
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Md Obayed Raihan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Othman Ghribi
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA.
| | - James E Porter
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA.
| |
Collapse
|
44
|
Dynamic control of the dopamine transporter in neurotransmission and homeostasis. NPJ Parkinsons Dis 2021; 7:22. [PMID: 33674612 PMCID: PMC7935902 DOI: 10.1038/s41531-021-00161-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/08/2021] [Indexed: 01/31/2023] Open
Abstract
The dopamine transporter (DAT) transports extracellular dopamine into the intracellular space contributing to the regulation of dopamine neurotransmission. A reduction of DAT density is implicated in Parkinson's disease (PD) by neuroimaging; dopamine turnover is dopamine turnover is elevated in early symptomatic PD and in presymptomatic individuals with monogenic mutations causal for parkinsonism. As an integral plasma membrane protein, DAT surface expression is dynamically regulated through endocytic trafficking, enabling flexible control of dopamine signaling in time and space, which in turn critically modulates movement, motivation and learning behavior. Yet the cellular machinery and functional implications of DAT trafficking remain enigmatic. In this review we summarize mechanisms governing DAT trafficking under normal physiological conditions and discuss how PD-linked mutations may disturb DAT homeostasis. We highlight the complexity of DAT trafficking and reveal DAT dysregulation as a common theme in genetic models of parkinsonism.
Collapse
|
45
|
Roshanbin S, Aniszewska A, Gumucio A, Masliah E, Erlandsson A, Bergström J, Ingelsson M, Ekmark-Lewén S. Age-related increase of alpha-synuclein oligomers is associated with motor disturbances in L61 transgenic mice. Neurobiol Aging 2021; 101:207-220. [PMID: 33639338 PMCID: PMC9648497 DOI: 10.1016/j.neurobiolaging.2021.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 12/08/2020] [Accepted: 01/11/2021] [Indexed: 12/15/2022]
Abstract
The pathogenesis of Parkinson’s disease involves fibrillization and deposition of alpha-synuclein (α-syn) into Lewy bodies. Accumulating evidence suggests that α-syn oligomers are particularly neurotoxic. Transgenic (tg) mice overexpressing wild-type human α-syn under the Thy-1 promoter (L61) reproduce many Parkinson’s disease features, but the pathogenetic relevance of α-syn oligomers in this mouse model has not been studied in detail. Here, we report an age progressive increase of α-syn oligomers in the brain of L61 tg mice. Interestingly, more profound motor symptoms were observed in animals with higher levels of membrane-bound oligomers. As this tg model is X-linked, we also performed subset analyses, indicating that both sexes display a similar age-related increase in α-syn oligomers. However, compared with females, males featured increased brain levels of oligomers from an earlier age, in addition to a more severe behavioral phenotype with hyperactivity and thigmotaxis in the open field test. Taken together, our data indicate that α-syn oligomers are central to the development of brain pathology and behavioral deficits in the L61 tg α-syn mouse model.
Collapse
Affiliation(s)
- Sahar Roshanbin
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Agata Aniszewska
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Astrid Gumucio
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | | | - Anna Erlandsson
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Joakim Bergström
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Sara Ekmark-Lewén
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
46
|
Thomsen MB, Ferreira SA, Schacht AC, Jacobsen J, Simonsen M, Betzer C, Jensen PH, Brooks DJ, Landau AM, Romero-Ramos M. PET imaging reveals early and progressive dopaminergic deficits after intra-striatal injection of preformed alpha-synuclein fibrils in rats. Neurobiol Dis 2020; 149:105229. [PMID: 33352233 DOI: 10.1016/j.nbd.2020.105229] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/24/2022] Open
Abstract
Alpha-synuclein (a-syn) can aggregate and form toxic oligomers and insoluble fibrils which are the main component of Lewy bodies. Intra-neuronal Lewy bodies are a major pathological characteristic of Parkinson's disease (PD). These fibrillar structures can act as seeds and accelerate the aggregation of monomeric a-syn. Indeed, recent studies show that injection of preformed a-syn fibrils (PFF) into the rodent brain can induce aggregation of the endogenous monomeric a-syn resulting in neuronal dysfunction and eventual cell death. We injected 8 μg of murine a-syn PFF, or soluble monomeric a-syn into the right striatum of rats. The animals were monitored behaviourally using the cylinder test, which measures paw asymmetry, and the corridor task that measures lateralized sensorimotor response to sugar treats. In vivo PET imaging was performed after 6, 13 and 22 weeks using [11C]DTBZ, a marker of the vesicular monoamine 2 transporter (VMAT2), and after 15 and 22 weeks using [11C]UCB-J, a marker of synaptic SV2A protein in nerve terminals. Histology was performed at the three time points using antibodies against dopaminergic markers, aggregated a-syn, and MHCII to evaluate the immune response. While the a-syn PFF injection caused only mild behavioural changes, [11C]DTBZ PET showed a significant and progressive decrease of VMAT2 binding in the ipsilateral striatum. This was accompanied by a small progressive decrease in [11C]UCB-J binding in the same area. In addition, our histological analysis revealed a gradual spread of misfolded a-syn pathology in areas anatomically connected to striatum that became bilateral with time. The striatal a-syn PFF injection resulted in a progressive unilateral degeneration of dopamine terminals, and an early and sustained presence of MHCII positive ramified microglia in the ipsilateral striatum and substantia nigra. Our study shows that striatal injections of a-syn fibrils induce progressive pathological synaptic dysfunction prior to cell death that can be detected in vivo with PET. We confirm that intrastriatal injection of a-syn PFFs provides a model of progressive a-syn pathology with loss of dopaminergic and synaptic function accompanied by neuroinflammation, as found in human PD.
Collapse
Affiliation(s)
- Majken B Thomsen
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Sara A Ferreira
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Anna C Schacht
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Jan Jacobsen
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Mette Simonsen
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Cristine Betzer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Poul H Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - David J Brooks
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark; Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Anne M Landau
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark; Translational Neuropsychiatry Unit, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
47
|
Polissidis A, Koronaiou M, Kollia V, Koronaiou E, Nakos-Bimpos M, Bogiongko M, Vrettou S, Karali K, Casadei N, Riess O, Sardi SP, Xilouri M, Stefanis L. Psychosis-Like Behavior and Hyperdopaminergic Dysregulation in Human α-Synuclein BAC Transgenic Rats. Mov Disord 2020; 36:716-728. [PMID: 33200461 DOI: 10.1002/mds.28383] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/28/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Parkinson's disease psychosis is a prevalent yet underreported and understudied nonmotor manifestation of Parkinson's disease and, arguably, the most debilitating. It is unknown if α-synuclein plays a role in psychosis, and if so, this endophenotype may be crucial for elucidating the neurodegenerative process. OBJECTIVES We sought to dissect the underlying neurobiology of novelty-induced hyperactivity, reminiscent of psychosis-like behavior, in human α-synuclein BAC rats. RESULTS Herein, we demonstrate a prodromal psychosis-like phenotype, including late-onset sensorimotor gating disruption, striatal hyperdopaminergic signaling, and persistent novelty-induced hyperactivity (up to 18 months), albeit reduced baseline locomotor activity, that is augmented by d-amphetamine and reversed by classical and atypical antipsychotics. MicroRNA-mediated α-synuclein downregulation in the ventral midbrain rescues the hyperactive phenotype and restores striatal dopamine levels. This phenotype is accompanied by an abundance of age-, brain region- and gene dose-dependent aberrant α-synuclein, including hyperphosphorylation, C-terminal truncation, aggregation pathology, and mild nigral neurodegeneration (27%). CONCLUSIONS Our findings demonstrate a potential role of α-synuclein in Parkinson's disease psychosis and provide evidence of region-specific perturbations prior to neurodegeneration phenoconversion. The reported phenotype coincides with the latest clinical findings that suggest a premotor hyperdopaminergic state may occur, while at the same time, premotor psychotic symptoms are increasingly being recognized. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Alexia Polissidis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Maria Koronaiou
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Vasia Kollia
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Effrosyni Koronaiou
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Modestos Nakos-Bimpos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Marios Bogiongko
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Sofia Vrettou
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Katerina Karali
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Sergio P Sardi
- Rare and Neurologic Diseases Research Therapeutic Area, Framingham, Massachusetts, USA
| | - Maria Xilouri
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Leonidas Stefanis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.,1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
48
|
Helmschrodt C, Becker S, Perl S, Schulz A, Richter A. Development of a fast liquid chromatography-tandem mass spectrometry method for simultaneous quantification of neurotransmitters in murine microdialysate. Anal Bioanal Chem 2020; 412:7777-7787. [PMID: 32939566 PMCID: PMC7550289 DOI: 10.1007/s00216-020-02906-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/09/2020] [Accepted: 08/20/2020] [Indexed: 12/23/2022]
Abstract
The continuous measurement of multiple neurotransmitters in microdialysate of freely moving mice to study neurochemical changes in specific brain regions requires a rapid and very sensitive quantitative analytical method. The quantitative analysis of 11 neurotransmitters and metabolites, including serotonin (5-HT), 5-hydroxyindoleacetic acid (5-HIAA), melatonin (ME), dopamine (DA), levodopa (l-DOPA), 3-methoxytyramine (3-MT), norepinephrine (NE), epinephrine (EP), acetylcholine (ACh), choline (Ch), and γ-aminobutyric acid (GABA), was performed using a biphenyl column coupled to an API-QTrap 3200 (AB SCIEX) mass spectrometer in positive electrospray ionization mode. To the microdialysate samples, 0.5 ng of isotopically labeled standard was added for analyte quantification. A rapid liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed and validated for the simultaneous analysis of monoamines, their precursor, and metabolites, as well as ACh, Ch, and GABA in murine microdialysate within 7.0 min. The limit of detection in artificial CSF ranged from 0.005 ng/mL (ME) to 0.75 ng/mL (NE and GABA). A comprehensive pre-analytical protocol was validated. Recovery was between 87 and 117% for neurotransmitter concentrations from 0.6 to 45 ng/mL with an inter-day accuracy of below 20%. Basal neurotransmitter values were determined in the striatum of mice over a time period of 3 h. This LC-MS/MS method, including a short and gentle sample preparation, is suitable for simultaneous measurements of neurotransmitters in murine cerebral microdialysate and enables the determination of basal neurotransmitter levels in specific brain regions to detect disease-related and drug-induced neurochemical changes. Graphical abstract![]()
Collapse
Affiliation(s)
- Christin Helmschrodt
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, An den Tierkliniken 15, 04103, Leipzig, Germany.
| | - Susen Becker
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, An den Tierkliniken 15, 04103, Leipzig, Germany
| | - Stefanie Perl
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, An den Tierkliniken 15, 04103, Leipzig, Germany
| | - Anja Schulz
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, An den Tierkliniken 15, 04103, Leipzig, Germany
| | - Angelika Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, An den Tierkliniken 15, 04103, Leipzig, Germany
| |
Collapse
|
49
|
Bellucci A, Bubacco L, Longhena F, Parrella E, Faustini G, Porrini V, Bono F, Missale C, Pizzi M. Nuclear Factor-κB Dysregulation and α-Synuclein Pathology: Critical Interplay in the Pathogenesis of Parkinson's Disease. Front Aging Neurosci 2020; 12:68. [PMID: 32265684 PMCID: PMC7105602 DOI: 10.3389/fnagi.2020.00068] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/25/2020] [Indexed: 12/13/2022] Open
Abstract
The loss of dopaminergic neurons of the nigrostriatal system underlies the onset of the typical motor symptoms of Parkinson's disease (PD). Lewy bodies (LB) and Lewy neurites (LN), proteinaceous inclusions mainly composed of insoluble α-synuclein (α-syn) fibrils are key neuropathological hallmarks of the brain of affected patients. Compelling evidence supports that in the early prodromal phases of PD, synaptic terminal and axonal alterations initiate and drive a retrograde degeneration process culminating with the loss of nigral dopaminergic neurons. This notwithstanding, the molecular triggers remain to be fully elucidated. Although it has been shown that α-syn fibrillary aggregation can induce early synaptic and axonal impairment and cause nigrostriatal degeneration, we still ignore how and why α-syn fibrillation begins. Nuclear factor-κB (NF-κB) transcription factors, key regulators of inflammation and apoptosis, are involved in the brain programming of systemic aging as well as in the pathogenesis of several neurodegenerative diseases. The NF-κB family of factors consists of five different subunits (c-Rel, p65/RelA, p50, RelB, and p52), which combine to form transcriptionally active dimers. Different findings point out a role of RelA in PD. Interestingly, the nuclear content of RelA is abnormally increased in nigral dopamine (DA) neurons and glial cells of PD patients. Inhibition of RelA exert neuroprotection against (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) MPTP and 1-methyl-4-phenylpyridinium (MPP+) toxicity, suggesting that this factor decreases neuronal resilience. Conversely, the c-Rel subunit can exert neuroprotective actions. We recently described that mice deficient for c-Rel develop a PD-like motor and non-motor phenotype characterized by progressive brain α-syn accumulation and early synaptic changes preceding the frank loss of nigrostriatal neurons. This evidence supports that dysregulations in this transcription factors may be involved in the onset of PD. This review highlights observations supporting a possible interplay between NF-κB dysregulation and α-syn pathology in PD, with the aim to disclose novel potential mechanisms involved in the pathogenesis of this disorder.
Collapse
Affiliation(s)
- Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luigi Bubacco
- Department of Biology, University of Padua, Padua, Italy
| | - Francesca Longhena
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Edoardo Parrella
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gaia Faustini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Vanessa Porrini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Federica Bono
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Cristina Missale
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marina Pizzi
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
50
|
Sampson TR, Challis C, Jain N, Moiseyenko A, Ladinsky MS, Shastri GG, Thron T, Needham BD, Horvath I, Debelius JW, Janssen S, Knight R, Wittung-Stafshede P, Gradinaru V, Chapman M, Mazmanian SK. A gut bacterial amyloid promotes α-synuclein aggregation and motor impairment in mice. eLife 2020; 9:53111. [PMID: 32043464 PMCID: PMC7012599 DOI: 10.7554/elife.53111] [Citation(s) in RCA: 224] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/23/2020] [Indexed: 12/14/2022] Open
Abstract
Amyloids are a class of protein with unique self-aggregation properties, and their aberrant accumulation can lead to cellular dysfunctions associated with neurodegenerative diseases. While genetic and environmental factors can influence amyloid formation, molecular triggers and/or facilitators are not well defined. Growing evidence suggests that non-identical amyloid proteins may accelerate reciprocal amyloid aggregation in a prion-like fashion. While humans encode ~30 amyloidogenic proteins, the gut microbiome also produces functional amyloids. For example, curli are cell surface amyloid proteins abundantly expressed by certain gut bacteria. In mice overexpressing the human amyloid α-synuclein (αSyn), we reveal that colonization with curli-producing Escherichia coli promotes αSyn pathology in the gut and the brain. Curli expression is required for E. coli to exacerbate αSyn-induced behavioral deficits, including intestinal and motor impairments. Purified curli subunits accelerate αSyn aggregation in biochemical assays, while oral treatment of mice with a gut-restricted amyloid inhibitor prevents curli-mediated acceleration of pathology and behavioral abnormalities. We propose that exposure to microbial amyloids in the gastrointestinal tract can accelerate αSyn aggregation and disease in the gut and the brain.
Collapse
Affiliation(s)
- Timothy R Sampson
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Collin Challis
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Neha Jain
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Anastasiya Moiseyenko
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Mark S Ladinsky
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Gauri G Shastri
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Taren Thron
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Brittany D Needham
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Istvan Horvath
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Justine W Debelius
- Department of Pediatrics, University of California, San Diego, San Diego, United States
| | - Stefan Janssen
- Department of Pediatrics, University of California, San Diego, San Diego, United States
| | - Rob Knight
- Department of Pediatrics, University of California, San Diego, San Diego, United States.,Department of Computer Science and Engineering, University of California, San Diego, San Diego, United States
| | | | - Viviana Gradinaru
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Matthew Chapman
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Sarkis K Mazmanian
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, United States
| |
Collapse
|