1
|
Berezhnoy G, Bae G, Wüst L, Schulte C, Cannet C, Wurster I, Zimmermann M, Jäck A, Spruth EJ, Hellmann-Regen J, Roeske S, Pürner D, Glanz W, Maass F, Hufschmidt F, Kilimann I, Dinter E, Kimmich O, Gamez A, Levin J, Priller J, Peters O, Wagner M, Storch A, Lingor P, Düzel E, van Riesen C, Wüllner U, Teipel S, Falkenburger B, Bähr M, Zerr I, Petzold GC, Spottke A, Rizzu P, Brosseron F, Schäfer H, Gasser T, Trautwein C. Application of IVDr NMR spectroscopy to stratify Parkinson's disease with absolute quantitation of blood serum metabolites and lipoproteins. Sci Rep 2025; 15:17738. [PMID: 40404791 PMCID: PMC12098827 DOI: 10.1038/s41598-025-01352-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 05/06/2025] [Indexed: 05/24/2025] Open
Abstract
The challenge of early detection and stratification in Parkinson's disease (PD) is urgent due to the current emergence of mechanism-based disease-modifying treatments. In here, metabolomic and lipidomic parameters obtained by a standardized and targeted in vitro diagnostic research (IVDr) platform have a significant potential to address therapy-related questions and generate improved biomarker panels. Our study aimed to use IVDr nuclear magnetic resonance (NMR) spectroscopy to quantify metabolites and lipoproteins in PD blood serum from different cohorts to stratify metabolically driven subtypes of idiopathic and genetic PD. Serum aliquots from three neurodegeneration biobank cohorts (287 samples in total, including 62 PD patient samples with GBA mutation, 98/43 PD patient samples of early/late stages of disease duration, 20 PD samples from patients with mutations in recessive PD genes and some smaller subgroups of mitochondrial and double mutation cases) were prepared and analyzed with IVDr NMR spectroscopy, covering 39 blood serum metabolites and 112 lipoprotein parameters. Uni- and multivariate statistics were used to identify metabolism-driven changes under consideration of typical confounders such as age, sex and disease duration and set into context with clinical biomarkers such as CSF concentrations of alpha-synuclein, neurofilament light chain, and tau protein. Based on the different PD subgroups we performed a total of eight different comparisons. Highlights from these comparisons include increased citrate and dimethylglycine with a decrease of creatinine and methionine in healthy controls and early PD group compared to GBA, PD late and recessive PD. We furthermore identified decreased HDL-3 free cholesterol in genetic PD cases compared to sporadic subject samples (sum of the PD early and PD late groups). Considering medication, we found that the levodopa equivalent daily dose (LEDD) is mostly positively correlated with tyrosine and citrate in sporadic PD compared to pyruvate and phenylalanine in genetic PD. Cerebrospinal fluid levels of alpha-synuclein were negatively correlated with alanine. Further metabolites and lipoproteins with discriminatory power for double mutation PD cases involved ornithine, 2-aminobutyrate and 2-hydroxybutyrate as well as for mitochondrial phenotypes via LDL phospholipid, apolipoprotein and cholesterol subfractions. Quantitative IVDr NMR serum spectroscopy is able to stratify PD patient samples of different etiology and can contribute to a wider understanding of the underlying metabolism-driven alterations e.g. in energy, amino acid, and lipoprotein metabolism. Though our overall cohort was large, major confounders such as age, sex and medication have a strong impact. That is why absolute quantification and detailed patient knowledge about metabolic confounders, is a premise for future translation of NMR serum spectroscopy to routine PD diagnostics.
Collapse
Affiliation(s)
- Georgy Berezhnoy
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Gyuntae Bae
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Leonie Wüst
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Claudia Schulte
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Claire Cannet
- Bruker BioSpin GmbH & Co. KG (AIC Division), Ettlingen, Germany
| | - Isabel Wurster
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Milan Zimmermann
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Alexander Jäck
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Neuropsychiatry and Laboratory of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julian Hellmann-Regen
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Neurosciences, Charité Universitätsmedizin Berlin, Berlin, Germany
- ECRC Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sandra Roeske
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Dominik Pürner
- Department of Neurology, School of Medicine, University Hospital München rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Clinic for Neurology, Medical Faculty, University Hospital Magdeburg, Magdeburg, Germany
| | - Fabian Maass
- Department of Neurology, University Medical Center, Georg August University, Göttingen, Germany
| | - Felix Hufschmidt
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn and University of Bonn, Bonn, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Elisabeth Dinter
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Okka Kimmich
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Vascular Neurology, University Hospital Bonn, Bonn, Germany
| | - Anna Gamez
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Neuropsychiatry and Laboratory of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University of Munich, German Center for Mental Health (DZPG), Munich, Germany
- University of Edinburgh and UK DRI, Edinburgh, UK
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin-Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn and University of Bonn, Bonn, Germany
| | - Alexander Storch
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Germany
- Department of Neurology, University Medical Centre, Rostock, Germany
| | - Paul Lingor
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, School of Medicine, University Hospital München rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Christoph van Riesen
- Department of Neurology, University Medical Center, Georg August University, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
| | - Ullrich Wüllner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn and University of Bonn, Bonn, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Björn Falkenburger
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center, Georg August University, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
| | - Inga Zerr
- Department of Neurology, University Medical Center, Georg August University, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Vascular Neurology, University Hospital Bonn, Bonn, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University of Bonn, Bonn, Germany
| | - Patricia Rizzu
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | | - Hartmut Schäfer
- Bruker BioSpin GmbH & Co. KG (AIC Division), Ettlingen, Germany
| | - Thomas Gasser
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
| | - Christoph Trautwein
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany.
- M3 Research Center for Malignome, Metabolome and Microbiome, Medical Faculty, University of Tübingen, Tübingen, Germany.
- Core Facility Metabolomics, Medical Faculty, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
2
|
Schreiber CS, Ramil LN, Bieligk J, Meineke R, Käufer C, Richter F. Intravenous SARS-CoV-2 Spike protein induces neuroinflammation and alpha-synuclein accumulation in brain regions relevant to Parkinson's disease. Brain Behav Immun 2025:S0889-1591(25)00197-7. [PMID: 40404020 DOI: 10.1016/j.bbi.2025.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 04/28/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) frequently presents with neurological symptoms in human patients and leads to long-lasting brain pathology in a hamster model. There is no overt SARS-CoV-2 virus replication in central neurons. Whether viral proteins are sufficient to cause this pathology requires further investigations. The SARS-CoV-2 Spike-protein S1-subunit (S1-protein) has recently gained interest for causing neuroinflammation and accelerating aggregation of alpha-synuclein (aSyn) in vitro. Here, we show the impact of S1-protein in a broad spectrum of brain regions after injection via three different application routes in C57/BL6 mice. METHODS S1-protein was administered either intranasally, intravenously or intracerebrally. We quantified aSyn immunoreactivity and phosphorylated aSyn (pS129), microglia and astrocyte reactivity, ACE2/Neuropilin-1 receptor expression, and parvalbumin-positive interneurons in limbic system, basal ganglia, and cortical regions 14 days post-application. Plasma cytokine profiles were assessed 6 days post-injection. RESULTS While intracerebral injection resulted in decreased aSyn immunoreactivity with increased pS129 in males, intravenous injection led to increased levels of aSyn immunoreactivity and microglia cell density, predominantly in brain regions associated with Parkinson's disease pathology. Intranasal application of S1-protein induced microgliosis in some brain regions but resulted in sex-dependent alterations of aSyn levels, with increases in females and decreases in males. All routes showed sex-dependent alterations in astrocytic reactivity, receptor expression, and parvalbumin-positive interneurons. CONCLUSION Our results demonstrate that S1-protein itself leads to neuroinflammation, altered aSyn homeostasis, and disruption of inhibitory circuits in a route- and sex-dependent manner. These findings indicate the possibility of S1-protein being a crucial agent for both neuroinflammatory processes and altered protein regulation mechanisms. S1-protein trapped in tissue reservoirs could therefore explain symptoms occurring or persisting beyond viral clearance (Post COVID-19 condition).
Collapse
Affiliation(s)
- Cara Sophie Schreiber
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover, (ZSN), Germany
| | - Lucas Navarro Ramil
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Juliette Bieligk
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Robert Meineke
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Christopher Käufer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover, (ZSN), Germany.
| | - Franziska Richter
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover, (ZSN), Germany.
| |
Collapse
|
3
|
Drath I, Richter F, Feja M. Nose-to-brain drug delivery: from bench to bedside. Transl Neurodegener 2025; 14:23. [PMID: 40390100 PMCID: PMC12090632 DOI: 10.1186/s40035-025-00481-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/18/2025] [Indexed: 05/21/2025] Open
Abstract
There is increasing interest in nose-to-brain delivery as an innovative drug delivery strategy for neurodegenerative disorders such as Parkinson's or Alzheimer's disease. The unique anatomy of the nose-brain interface facilitates direct drug transport via the olfactory and trigeminal pathways to the brain, bypassing the blood-brain barrier. Different administration techniques as well as advanced drug formulations like targeted nanoparticles and thermoresponsive systems have been explored to improve the delivery efficiency and the therapeutic efficacy. This review provides an up-to-date perspective on this fast-developing field, and discusses different studies on safety and pharmacokinetic properties. A thorough evaluation of preclinical and clinical studies reveals both promises and challenges of this delivery method, highlighting approved drugs for the treatment of epilepsy and migraine that successfully utilize intranasal routes. The current landscape of research on nose-to-brain delivery is critically discussed, and a rationale is provided for ongoing research to optimize therapeutic strategies.
Collapse
Affiliation(s)
- Isabell Drath
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany.
- Center for Systems Neuroscience (ZSN), Hannover, Germany.
| | - Malte Feja
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany.
- Center for Systems Neuroscience (ZSN), Hannover, Germany.
| |
Collapse
|
4
|
Wichmann T, Nelson A, Torres ERS, Svenningsson P, Marongiu R. Leveraging animal models to understand non-motor symptoms of Parkinson's disease. Neurobiol Dis 2025; 208:106848. [PMID: 40023327 DOI: 10.1016/j.nbd.2025.106848] [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: 10/10/2024] [Revised: 01/01/2025] [Accepted: 01/02/2025] [Indexed: 03/04/2025] Open
Abstract
Parkinson's disease is diagnosed based on motor symptoms, but non-motor symptoms of the disease, such as cognitive impairment, autonomic dysfunction, hyposmia, sleep disorders, and psychiatric disorders heavily impact patient and caregiver quality of life. It has proven challenging to faithfully reproduce and quantify these non-motor phenotypes. Indeed, many non-motor signs in animals that may phenotypically resemble features in patients may be caused by different mechanisms or may not be consistent within the same or similar models. In this review, we survey the existing literature on the assessment of non-motor signs in parkinsonian rodents and non-human primates. We highlight the gaps in our understanding and suggest how researchers might improve experimental designs to produce more meaningful results with the hope of better understanding the disease and developing better therapies.
Collapse
Affiliation(s)
- Thomas Wichmann
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA 30329, USA; Udall Center of Excellence in Parkinson's Disease Research, Emory University, Atlanta, GA 30329, USA; Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Alexandra Nelson
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA; Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA; Department of Neurology, UCSF, San Francisco, CA 94158, USA
| | - Eileen Ruth S Torres
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Per Svenningsson
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Basic and Clinical Neuroscience, King's College London, London, United Kingdom
| | - Roberta Marongiu
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA; Department of Genetic Medicine, New-York Hospital-Cornell Medical College, New York, NY, USA; Feil Family Brain and Mind Institute, New-York Hospital-Cornell Medical College, New York, NY, USA.
| |
Collapse
|
5
|
Lai TT, Xiang W, Stanojlovic M, Käufer C, Feja M, Lau K, Zunke F, Richter F. The basolateral amygdala and striatum propagate alpha-synuclein pathology causing increased fear response in a Parkinson's disease model. Brain Behav Immun 2025; 128:469-486. [PMID: 40274000 DOI: 10.1016/j.bbi.2025.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/30/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025] Open
Abstract
Alpha-synuclein (aSyn)-related pathology crucially contributes to the pathogenesis of Parkinson's disease, a frequent and incurable neurodegenerative disease characterized by progressive motor and non-motor symptoms. Anxiety and fear- related neuropsychiatric symptoms develop frequently and early in the disease, but a lack of understanding of pathogenesis hampers rational therapy. This study aimed to decipher whether aSyn pathology in the basolateral amygdala (BLA) is causative of fear and anxiety. Bilateral stereotaxic injections of human aSyn-preformed amyloid fibrils (PFF) in BLA, striatum, or substantia nigra were conducted in female mice overexpressing human aSyn (Thy1-aSyn) and in wildtype littermates (WT). We characterized the propagation of aSyn pathology and related neuropathological changes across brain regions and examined the behavioral and fear responses in mice up to 2 months post-injection of PFF. While PFF injections induced local aSyn fibril pathology close to all respective injection sites in transgenic mice, we observed differences in propagation, downstream pathology and behavioral alterations. The BLA and the striatum, but not the substantia nigra, effectively propagated aSyn pathology to connected brain regions at 2.5 months post injection. This involved enhanced microgliosis and astrogliosis in the nigrostriatal system and loss of GABAergic parvalbuminergic interneurons in the striatum and corticolimbic brain regions. Intra-BLA PFF injections resulted in increased cued fear response in both transgenic mice and WT mice at 1 month post injection. The effect was more pronounced in the transgenic mice. Conversely, intra-striatal PFF injections enhanced contextual fear in WT at 2 months post injection. These findings imply that increased fear is inducible by aSyn pathology, especially if originating in the BLA or striatum. Furthermore, both regions are hub regions of aSyn pathology propagation, thereby contributing to disease progression. These insights provide mechanisms that can guide rational therapeutic development.
Collapse
Affiliation(s)
- Thuy Thi Lai
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience Hannover, Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Germany
| | - Milos Stanojlovic
- Department of Neurobiology, Institute for Biological Research Siniša Stanković - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Christopher Käufer
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience Hannover, Germany
| | - Malte Feja
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience Hannover, Germany
| | - Kristina Lau
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience Hannover, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience Hannover, Germany.
| |
Collapse
|
6
|
Kim Y, Vaidya B, McInnes J, Zoghbi HY. Alpha-Synuclein Phosphomimetic Y39E and S129D Knock-In Mice Show Cytosolic Alpha-Synuclein Localization without Developing Neurodegeneration or Motor Deficits. eNeuro 2025; 12:ENEURO.0357-24.2025. [PMID: 40164505 PMCID: PMC11998964 DOI: 10.1523/eneuro.0357-24.2025] [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: 08/15/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 04/02/2025] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by motor and nonmotor symptoms. Its pathological hallmarks include the accumulation of misfolded alpha-synuclein (α-Syn) in Lewy bodies and Lewy neurites. Phosphorylation of α-Syn is a prominent feature of these inclusions, but its role in disease pathogenesis remains unclear. To identify the role of α-Syn phosphorylation in synucleinopathy, we generated two Snca knock-in (KI) mouse models carrying phosphomimetic mutations at SncaY39 or SncaS129 (SncaY39E or SncaS129D ) which manipulated epitopes phosphorylated in the PD brain. Both SncaY39E and SncaS129D KI mice displayed increased α-Syn phosphorylation, enhanced oligomer formation, and a shift of α-Syn localization from membrane-bound to cytoplasm. However, neurodegeneration in the substantia nigra was not observed up to 24 months of age. These findings demonstrate that mimicking the phosphorylation of Y39 or S129 can induce endogenous α-Syn phosphorylation. Still, a single phosphomimetic mutation alone is insufficient to induce PD-like behavior and pathology in the mouse's lifespan. Overall, our study provides a mouse model for investigating the role of phosphorylation at Y39 and S129 α-Syn epitopes in vivo.
Collapse
Affiliation(s)
- YoungDoo Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Bhupesh Vaidya
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Joseph McInnes
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Huda Y Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
- Department of Neuroscience, BCM, Houston, Texas 77030
- Department of Pediatrics, BCM, Houston, Texas 77030
- Department of Neurology, BCM, Houston, Texas 77030
- Howard Hughes Medical Institute, Houston, Texas 77030
| |
Collapse
|
7
|
Käufer C, Stanojlović M, Schidlitzki A, Bonsberger J, Storch A, Richter F. Alterations in non-REM sleep and EEG spectra precede REM-sleep deficits in a model of synucleinopathy. JOURNAL OF PARKINSON'S DISEASE 2025; 15:311-328. [PMID: 39973511 DOI: 10.1177/1877718x241310723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
BackgroundSleep disturbances often precede motor symptoms in neurodegenerative diseases like Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Neuroinflammation is implicated in PD pathophysiology and may contribute to non-motor symptoms such as sleep disturbances. The Thy1-αSyn mouse model, overexpressing human alpha-synuclein (αSyn), mimics key aspects of PD and DLB, making it valuable for studying related sleep disturbances and neuroinflammatory changes.ObjectiveTo investigate early-stage alterations in sleep architecture, electroencephalographic (EEG) patterns, and neuroinflammation in Thy1-αSyn mice.MethodsWe used telemetric EEG/electromyography (EMG) with video surveillance to compare sleep patterns and EEG spectral power between 2.5- and 4.5-month-old male Thy1-αSyn transgenic mice and wild-type littermates. Neuroinflammation was assessed by examining microglial (Iba1) and astrocytic (GFAP) activation in key sleep-regulating brain regions.ResultsThy1-αSyn mice showed decreased resting wake time and increased non-REM sleep, with altered sleep bout frequency and length, indicating significant sleep architecture changes. Spectral analysis revealed a shift from higher to lower frequency bands, suggesting early neural circuitry disruptions due to αSyn overexpression. Significant microglial activation was observed at 3 months, with astrogliosis progressing by 5 months in key sleep-regulating regions, indicating that neuroinflammation may contribute to the observed sleep disturbances.ConclusionsEarly-stage Thy1-αSyn mice exhibit significant sleep architecture changes, EEG spectral shifts, and neuroinflammatory alterations. These findings suggest that neuroinflammation may play a role in the initial pathophysiological changes in PD and related synucleinopathies. Sleep, EEG, and neuroinflammatory changes could serve as early biomarkers for these diseases.
Collapse
Affiliation(s)
- Christopher Käufer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Miloš Stanojlović
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
- Department of Neurobiology, Institute for Biological Research Siniša Stanković - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Alina Schidlitzki
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Jana Bonsberger
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Alexander Storch
- Department of Neurology, University of Rostock, Rostock, Germany
- German Centre for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience Hannover, Hannover, Germany
| |
Collapse
|
8
|
McGregor BA, Raihan MO, Brishti A, Hur J, Porter JE. Deciphering motor dysfunction and microglial activation in mThy1- α-synuclein mice: a comprehensive study of behavioral, gene expression, and methylation changes. Front Mol Neurosci 2025; 18:1544971. [PMID: 40018011 PMCID: PMC11865073 DOI: 10.3389/fnmol.2025.1544971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/24/2025] [Indexed: 03/01/2025] Open
Abstract
Introduction Growing recognition of microglia's role in neurodegenerative disorders has accentuated the need to characterize microglia profiles and their influence on pathogenesis. To understand changes observed in the microglial profile during the progression of synucleinopathies, microglial gene expression and DNA methylation were examined in the mThy1-α-synuclein mouse model. Methods Disease progression was determined using behavioral tests evaluating locomotor deficits before DNA and RNA extraction at 7 and 10 months from isolated microglia for enzymatic methyl-sequencing and RNA-sequencing. Results Pathway analysis of these changes at 7 months indicates a pro-inflammatory profile and changes in terms related to synaptic maintenance. Expression and methylation at both 7 and 10 months included terms regarding mitochondrial and metabolic stress. While behavior symptoms progressed at 10 months, we see many previously activated pathways being inhibited in microglia at a later stage, with only 8 of 53 shared pathways predicted to be directionally concordant. Despite the difference in pathway directionality, 21 of the 22 genes that were differentially expressed and annotated to differentially methylated regions at both 7 and 10 months had conserved directionality changes. Discussion These results highlight a critical period in disease progression, during which the microglia respond to α-synuclein, suggesting a transition in the role of microglia from the early to late stages of the disease.
Collapse
Affiliation(s)
- Brett A. McGregor
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - Md. Obayed Raihan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
- Department of Pharmaceutical Sciences, Chicago State University School of Pharmacy, Chicago, IL, United States
| | - Afrina Brishti
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - James E. Porter
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| |
Collapse
|
9
|
Sampson TR, Wallen ZD, Won WJ, Standaert DG, Payami H, Harms AS. Alpha synuclein overexpression can drive microbiome dysbiosis in mice. Sci Rep 2025; 15:4014. [PMID: 39893159 PMCID: PMC11787330 DOI: 10.1038/s41598-024-82139-7] [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: 04/28/2024] [Accepted: 12/03/2024] [Indexed: 02/04/2025] Open
Abstract
Growing evidence indicates that persons with Parkinson disease (PD), have a unique composition of indigenous gut microbes. Given the long prodromal or pre-diagnosed period, longitudinal studies of the human and rodent gut microbiome before symptomatic onset and for the duration of the disease are currently lacking. PD is partially characterized by the accumulation of the protein α-synuclein (α-syn) into insoluble aggregates, in both the central and enteric nervous systems. As such, several experimental rodent and non-human primate models of α-syn overexpression recapitulate some of the hallmark pathophysiologies of PD. These animal models provide an opportunity to assess how the gut microbiome changes with age under disease-relevant conditions. Here, we used a transgenic mouse strain, which overexpress wild-type human α-syn to test how the gut microbiome composition responds in this model of PD pathology during aging. Using shotgun metagenomics, we find significant, age and genotype-dependent bacterial taxa whose abundance becomes altered with age. We reveal that α-syn overexpression can drive alterations to the gut microbiome composition and suggest that it limits diversity through age. Taxa that were most affected by genotype-age interaction were Lactobacillus and Bifidobacteria. In a mouse model, we showed direct link between alpha synuclein geneotype (hallmark of PD), a dysbiotic and low-diversity gut microbiome, and dysbiotic levels of Bifidobacteria and Lactobacillus (most robust features of PD microbiome). Given emerging data on the potential contributions of the gut microbiome to PD pathologies, our data provide an experimental foundation to understand how the PD-associated microbiome may arise as a trigger or co-pathology to disease.
Collapse
Affiliation(s)
- Timothy R Sampson
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zachary D Wallen
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Woong-Jai Won
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - David G Standaert
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Haydeh Payami
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| | - Ashley S Harms
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
10
|
Hartke AS, Schreiber CS, Lau K, Wiesweg I, Waltl I, Kalinke U, Richter F, Käufer C. Alpha-synuclein pathology enhances peripheral and CNS immune responses to bacterial endotoxins. Neurobiol Dis 2025; 205:106773. [PMID: 39710067 DOI: 10.1016/j.nbd.2024.106773] [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: 09/04/2024] [Revised: 11/18/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024] Open
Abstract
Increasing evidence points to infectious diseases as contributor to the pathogenesis of neurodegeneration in Parkinson's disease (PD), probably driven by a peripheral and CNS inflammatory response together with alpha-synuclein (aSyn) pathology. Pro-inflammatory lipopolysaccharide (LPS) endotoxin is suggested as a risk factor, and LPS shedding gram-negative bacteria are more prevalent in the gut-microbiome of PD patients. Here, we investigated whether LPS could contribute to the neurodegenerative disease progression via neuroinflammation, especially under conditions of aSyn pathology. To investigate this, we created a double-hit model based on the Thy1-aSyn mouse line (line 61), an established aSyn-overexpression model of PD, exposed to a single intraperitoneal injection of LPS at a dose of 0.8 mg/kg (equivalent to approximately 1,200,000 EU/kg). Clinical parameters, flow cytometry of blood and immune cells in the brain, brain immunohistology and motor behavior were evaluated over time. As expected, the LPS dosage induced transient acute symptoms and mild weight loss in mice, with full recovery after 7 days. In aSyn over-expressing mice, this single low dose of LPS was sufficient to alter the expression of specific markers on blood and brain immune cells and induced brain region-specific microgliosis that were present at 7 days post LPS injection. At 14 days post injection of LPS, aSyn expression was reduced in wild-type mice, indicating a specific response of the endogenous protein to the endotoxin. At this early time point, motor behavior is not yet robustly impacted by the observed pathological alterations. In conclusion, aSyn pathology renders the peripheral and central immune response more sensitive to a single low dose of bacterial endotoxin, which mimics a transient dysbiosis or gut infection. Thus, this data suggests that such peripheral triggers should be monitored in PD patients for instance by blood immune cell response as biomarkers. Furthermore, results from this study lend further support to the development of treatments aiming to reduce the impact of bacterial dysbiosis as a promising strategy to mitigate PD progression.
Collapse
Affiliation(s)
- Anna-Sophia Hartke
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany; Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Cara S Schreiber
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany; Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Kristina Lau
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany; Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Ivo Wiesweg
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany
| | - Inken Waltl
- Institute for Experimental Infection Research, TWINCORE, Center for Experimental and Clinical Infection Research, Feodor-Lynen-Str. 7, 30625 Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Center for Experimental and Clinical Infection Research, Feodor-Lynen-Str. 7, 30625 Hannover, Germany; Center for Systems Neuroscience (ZSN), Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany; Center for Systems Neuroscience (ZSN), Hannover, Germany.
| | - Christopher Käufer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany.
| |
Collapse
|
11
|
Pintado-Grima C, Ventura S. The role of amphipathic and cationic helical peptides in Parkinson's disease. Protein Sci 2025; 34:e70020. [PMID: 39720890 DOI: 10.1002/pro.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/29/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024]
Abstract
Peptides are attracting a growing interest for therapeutic applications in biomedicine. In Parkinson's disease (PD), different human endogenous peptides have been associated with beneficial effects, including protein aggregation inhibition, reduced inflammation, or the protection of dopaminergic neurons. Such effects seem to be connected to the spatial arrangement of peptide side chains, and many of these human molecules share common conformational traits, displaying a distinctive amphipathic and cationic helical structure, which is believed to be crucial for their activities. This review delves into the relationship between these structural properties and the current evidence connecting biogenic peptides to the amelioration of PD symptoms. We discuss their implications in the disease, the different mechanisms of action, their state of validation, and their therapeutic potential.
Collapse
Affiliation(s)
- Carlos Pintado-Grima
- Institut de Biotecnologia i de Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Salvador Ventura
- Institut de Biotecnologia i de Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
- Hospital Universitari Parc Taulí, Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, Sabadell, Spain
| |
Collapse
|
12
|
Horan-Portelance L, Iba M, Acri DJ, Gibbs JR, Cookson MR. Imaging spatial transcriptomics reveals molecular patterns of vulnerability to pathology in a transgenic α-synucleinopathy model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606032. [PMID: 39372781 PMCID: PMC11451628 DOI: 10.1101/2024.07.31.606032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
In Parkinson's disease and dementia with Lewy bodies, aggregated and phosphorylated α-synuclein pathology appears in select neurons throughout cortical and subcortical regions, but little is currently known about why certain populations are selectively vulnerable. Here, using imaging spatial transcriptomics (IST) coupled with downstream immunofluorescence for α-synuclein phosphorylated at Ser129 (pSyn) in the same tissue sections, we identified neuronal subtypes in the cortex and hippocampus of transgenic human α-synuclein-overexpressing mice that preferentially developed pSyn pathology. Additionally, we investigated the transcriptional underpinnings of this vulnerability, pointing to expression of Plk2, which phosphorylates α-synuclein at Ser129, and human SNCA (hSNCA), as key to pSyn pathology development. Finally, we performed differential expression analysis, revealing gene expression changes broadly downstream of hSNCA overexpression, as well as pSyn-dependent alterations in mitochondrial and endolysosomal genes. Overall, this study yields new insights into the formation of α-synuclein pathology and its downstream effects in a synucleinopathy mouse model.
Collapse
Affiliation(s)
- Liam Horan-Portelance
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michiyo Iba
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dominic J. Acri
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - J. Raphael Gibbs
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
13
|
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 PMCID: PMC11525669 DOI: 10.1038/s41531-024-00816-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [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
|
14
|
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, TX77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX77030
| | - Joseph McInnes
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX77030
| | - Jiyoen Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX77030
| | - Yan Hong Wei Liang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX77030
| | - Surabi Veeraragavan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX77030
| | - Alexandra Rae Garza
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX77030
| | - Benjamin David Webst Belfort
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX77030
| | - Benjamin Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX77030
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
| | - Rodney Samaco
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX77030
| | - Huda Yahya Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX77030
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
- Department of Pediatrics, Baylor College of Medicine, Houston, TX77030
- Department of Neurology, Baylor College of Medicine, Houston, TX77030
- HHMI, Baylor College of Medicine, Houston, TX77030
| |
Collapse
|
15
|
Hong H, Wang Y, Menard M, Buckley JA, Zhou L, Volpicelli-Daley L, Standaert DG, Qin H, Benveniste EN. Suppression of the JAK/STAT pathway inhibits neuroinflammation in the line 61-PFF mouse model of Parkinson's disease. J Neuroinflammation 2024; 21:216. [PMID: 39218899 PMCID: PMC11368013 DOI: 10.1186/s12974-024-03210-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Parkinson's disease (PD) is characterized by neuroinflammation, progressive loss of dopaminergic neurons, and accumulation of α-synuclein (α-Syn) into insoluble aggregates called Lewy pathology. The Line 61 α-Syn mouse is an established preclinical model of PD; Thy-1 is used to promote human α-Syn expression, and features of sporadic PD develop at 9-18 months of age. To accelerate the PD phenotypes, we injected sonicated human α-Syn preformed fibrils (PFFs) into the striatum, which produced phospho-Syn (p-α-Syn) inclusions in the substantia nigra pars compacta and significantly increased MHC Class II-positive immune cells. Additionally, there was enhanced infiltration and activation of innate and adaptive immune cells in the midbrain. We then used this new model, Line 61-PFF, to investigate the effect of inhibiting the JAK/STAT signaling pathway, which is critical for regulation of innate and adaptive immune responses. After administration of the JAK1/2 inhibitor AZD1480, immunofluorescence staining showed a significant decrease in p-α-Syn inclusions and MHC Class II expression. Flow cytometry showed reduced infiltration of CD4+ T-cells, CD8+ T-cells, CD19+ B-cells, dendritic cells, macrophages, and endogenous microglia into the midbrain. Importantly, single-cell RNA-Sequencing analysis of CD45+ cells from the midbrain identified 9 microglia clusters, 5 monocyte/macrophage (MM) clusters, and 5 T-cell (T) clusters, in which potentially pathogenic MM4 and T3 clusters were associated with neuroinflammatory responses in Line 61-PFF mice. AZD1480 treatment reduced cell numbers and cluster-specific expression of the antigen-presentation genes H2-Eb1, H2-Aa, H2-Ab1, and Cd74 in the MM4 cluster and proinflammatory genes such as Tnf, Il1b, C1qa, and C1qc in the T3 cluster. Together, these results indicate that inhibiting the JAK/STAT pathway suppresses the activation and infiltration of innate and adaptive cells, reducing neuroinflammation in the Line 61-PFF mouse model.
Collapse
Affiliation(s)
- Huixian Hong
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Boulevard, MCLM 907, Birmingham, AL, 35294, USA
| | - Yong Wang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Boulevard, MCLM 907, Birmingham, AL, 35294, USA
| | - Marissa Menard
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jessica A Buckley
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Boulevard, MCLM 907, Birmingham, AL, 35294, USA
| | - Lianna Zhou
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Boulevard, MCLM 907, Birmingham, AL, 35294, USA
| | - Laura Volpicelli-Daley
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - David G Standaert
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Hongwei Qin
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Boulevard, MCLM 907, Birmingham, AL, 35294, USA.
| | - Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Boulevard, MCLM 907, Birmingham, AL, 35294, USA.
| |
Collapse
|
16
|
Lau K, Kotzur R, Richter F. Blood-brain barrier alterations and their impact on Parkinson's disease pathogenesis and therapy. Transl Neurodegener 2024; 13:37. [PMID: 39075566 PMCID: PMC11285262 DOI: 10.1186/s40035-024-00430-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024] Open
Abstract
There is increasing evidence for blood-brain barrier (BBB) alterations in Parkinson's disease (PD), the second most common neurodegenerative disorder with rapidly rising prevalence. Altered tight junction and transporter protein levels, accumulation of α-synuclein and increase in inflammatory processes lead to extravasation of blood molecules and vessel degeneration. This could result in a self-perpetuating pathophysiology of inflammation and BBB alteration, which contribute to neurodegeneration. Toxin exposure or α-synuclein over-expression in animal models has been shown to initiate similar pathologies, providing a platform to study underlying mechanisms and therapeutic interventions. Here we provide a comprehensive review of the current knowledge on BBB alterations in PD patients and how rodent models that replicate some of these changes can be used to study disease mechanisms. Specific challenges in assessing the BBB in patients and in healthy controls are discussed. Finally, a potential role of BBB alterations in disease pathogenesis and possible implications for therapy are explored. The interference of BBB alterations with current and novel therapeutic strategies requires more attention. Brain region-specific BBB alterations could also open up novel opportunities to target specifically vulnerable neuronal subpopulations.
Collapse
Affiliation(s)
- Kristina Lau
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Rebecca Kotzur
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
17
|
Schreiber CS, Wiesweg I, Stanelle-Bertram S, Beck S, Kouassi NM, Schaumburg B, Gabriel G, Richter F, Käufer C. Sex-specific biphasic alpha-synuclein response and alterations of interneurons in a COVID-19 hamster model. EBioMedicine 2024; 105:105191. [PMID: 38865747 PMCID: PMC11293593 DOI: 10.1016/j.ebiom.2024.105191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 05/02/2024] [Accepted: 05/25/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) frequently leads to neurological complications after recovery from acute infection, with higher prevalence in women. However, mechanisms by which SARS-CoV-2 disrupts brain function remain unclear and treatment strategies are lacking. We previously demonstrated neuroinflammation in the olfactory bulb of intranasally infected hamsters, followed by alpha-synuclein and tau accumulation in cortex, thus mirroring pathogenesis of neurodegenerative diseases such as Parkinson's or Alzheimer's disease. METHODS To uncover the sex-specific spatiotemporal profiles of neuroinflammation and neuronal dysfunction following intranasal SARS-CoV-2 infection, we quantified microglia cell density, alpha-synuclein immunoreactivity and inhibitory interneurons in cortical regions, limbic system and basal ganglia at acute and late post-recovery time points. FINDINGS Unexpectedly, microglia cell density and alpha-synuclein immunoreactivity decreased at 6 days post-infection, then rebounded to overt accumulation at 21 days post-infection. This biphasic response was most pronounced in amygdala and striatum, regions affected early in Parkinson's disease. Several brain regions showed altered densities of parvalbumin and calretinin interneurons which are involved in cognition and motor control. Of note, females appeared more affected. INTERPRETATION Our results demonstrate that SARS-CoV-2 profoundly disrupts brain homeostasis without neuroinvasion, via neuroinflammatory and protein regulation mechanisms that persist beyond viral clearance. The regional patterns and sex differences are in line with neurological deficits observed after SARS-CoV-2 infection. FUNDING Federal Ministry of Health, Germany (BMG; ZMV I 1-2520COR501 to G.G.), Federal Ministry of Education and Research, Germany (BMBF; 03COV06B to G.G.), Ministry of Science and Culture of Lower Saxony in Germany (14-76403-184, to G.G. and F.R.).
Collapse
Affiliation(s)
- Cara Sophie Schreiber
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover (ZSN), Germany
| | - Ivo Wiesweg
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Sebastian Beck
- Department for Viral Zoonoses-One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Nancy Mounogou Kouassi
- Department for Viral Zoonoses-One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Berfin Schaumburg
- Department for Viral Zoonoses-One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Gülsah Gabriel
- Department for Viral Zoonoses-One Health, Leibniz Institute of Virology, Hamburg, Germany; Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover (ZSN), Germany.
| | - Christopher Käufer
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover (ZSN), Germany.
| |
Collapse
|
18
|
Selvaraj DB, Panneerselvam A, Vergil Andrews JF, Kandasamy M. Cysteamine HCl Administration Impedes Motor and Olfactory Functions, Accompanied by a Reduced Number of Dopaminergic Neurons, in Experimental Mice: A Preclinical Mimetic Relevant to Parkinson's Disease. Brain Sci 2024; 14:632. [PMID: 39061373 PMCID: PMC11275195 DOI: 10.3390/brainsci14070632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Cysteamine hydrochloride (Cys-HCl) has been established as a potent ulcerogenic agent of the gastrointestinal (GI) system. GI dysfunction and olfactory deficits are the most common clinical symptoms of many movement disorders, including Parkinson's disease (PD). Cys-HCl has been shown to interfere with dopamine, a neurotransmitter crucial for motor, olfactory, and cognitive functions. However, the reports on the effect of Cys-HCl treatment on the behavioral aspects and functions of the dopamine system appear to be inconsistent. Therefore, we revisited the impact of Cys-HCl on the motor function in experimental mice using a battery of behavioral tests, such as the pole test (PT), beam-walking test (BWT), and rotarod test (RDT), while the olfactory ability and cognitive functions were examined through the buried-food test (BFT) and Y-maze test. Furthermore, we investigated the effect of Cys-HCl on the number of dopaminergic tyrosine hydroxylase (TH)-positive cells in the substantia nigra (SN) and olfactory bulb (OB) of the experimental mice using immunohistochemistry. The results revealed that Cys-HCl administration in the mice induced significant impairments in their motor balance and coordination, as their movement-related performances were markedly reduced in terms of the behavioral tasks. Mice exposed to Cys-HCl showed pronounced reductions in their odor discrimination abilities as well as cognitive impairments. Strikingly, the number of TH-positive neurons was found to be reduced in the SN and OB of the Cys-HCl-treated group, which is a bonafide neuropathogenic hallmark of PD. This study highlights the potential neurotoxic effects of Cys-HCl in experimental brains and suggests further investigation into its role in the pathogenesis of Parkinsonism.
Collapse
Affiliation(s)
- Divya Bharathi Selvaraj
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India; (D.B.S.); (J.F.V.A.)
| | - Anusiya Panneerselvam
- Department of Biotechnology, Bharathidasan University, Tiruchirappalli 620024, India;
| | - Jemi Feiona Vergil Andrews
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India; (D.B.S.); (J.F.V.A.)
| | - Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India; (D.B.S.); (J.F.V.A.)
- University Grants Commission-Faculty Recharge Programme (UGC-FRP), New Delhi 110002, India
| |
Collapse
|
19
|
Hong H, Wang Y, Menard M, Buckley J, Zhou L, Volpicelli-Daley L, Standaert D, Qin H, Benveniste E. Suppression of the JAK/STAT Pathway Inhibits Neuroinflammation in the Line 61-PFF Mouse Model of Parkinson's Disease. RESEARCH SQUARE 2024:rs.3.rs-4307273. [PMID: 38766241 PMCID: PMC11100885 DOI: 10.21203/rs.3.rs-4307273/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Parkinson's disease (PD) is characterized by neuroinflammation, progressive loss of dopaminergic neurons, and accumulation of a-synuclein (a-Syn) into insoluble aggregates called Lewy pathology. The Line 61 a-Syn mouse is an established preclinical model of PD; Thy-1 is used to promote human a-Syn expression, and features of sporadic PD develop at 9-18 months of age. To accelerate the PD phenotypes, we injected sonicated human a-Syn preformed fibrils (PFFs) into the striatum, which produced phospho-Syn (p-a-Syn) inclusions in the substantia nigra pars compacta and significantly increased MHC Class II-positive immune cells. Additionally, there was enhanced infiltration and activation of innate and adaptive immune cells in the midbrain. We then used this new model, Line 61-PFF, to investigate the effect of inhibiting the JAK/STAT signaling pathway, which is critical for regulation of innate and adaptive immune responses. After administration of the JAK1/2 inhibitor AZD1480, immunofluorescence staining showed a significant decrease in p-a-Syn inclusions and MHC Class II expression. Flow cytometry showed reduced infiltration of CD4+ T-cells, CD8+ T-cells, CD19+ B-cells, dendritic cells, macrophages, and endogenous microglia into the midbrain. Importantly, single-cell RNA-Sequencing analysis of CD45+ cells from the midbrain identified 9 microglia clusters, 5 monocyte/macrophage (MM) clusters, and 5 T-cell (T) clusters, in which potentially pathogenic MM4 and T3 clusters were associated with neuroinflammatory responses in Line 61-PFF mice. AZD1480 treatment reduced cell numbers and cluster-specific expression of the antigen-presentation genes H2-Eb1, H2-Aa, H2-Ab1, and Cd74 in the MM4 cluster and proinflammatory genes such as Tnf, Il1b, C1qa, and C1qc in the T3 cluster. Together, these results indicate that inhibiting the JAK/STAT pathway suppresses the activation and infiltration of innate and adaptive cells, reducing neuroinflammation in the Line 61-PFF mouse model.
Collapse
|
20
|
Mercado G, Kaeufer C, Richter F, Peelaerts W. Infections in the Etiology of Parkinson's Disease and Synucleinopathies: A Renewed Perspective, Mechanistic Insights, and Therapeutic Implications. JOURNAL OF PARKINSON'S DISEASE 2024; 14:1301-1329. [PMID: 39331109 PMCID: PMC11492057 DOI: 10.3233/jpd-240195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/06/2024] [Indexed: 09/28/2024]
Abstract
Increasing evidence suggests a potential role for infectious pathogens in the etiology of synucleinopathies, a group of age-related neurodegenerative disorders including Parkinson's disease (PD), multiple system atrophy and dementia with Lewy bodies. In this review, we discuss the link between infections and synucleinopathies from a historical perspective, present emerging evidence that supports this link, and address current research challenges with a focus on neuroinflammation. Infectious pathogens can elicit a neuroinflammatory response and modulate genetic risk in PD and related synucleinopathies. The mechanisms of how infections might be linked with synucleinopathies as well as the overlap between the immune cellular pathways affected by virulent pathogens and disease-related genetic risk factors are discussed. Here, an important role for α-synuclein in the immune response against infections is emerging. Critical methodological and knowledge gaps are addressed, and we provide new future perspectives on how to address these gaps. Understanding how infections and neuroinflammation influence synucleinopathies will be essential for the development of early diagnostic tools and novel therapies.
Collapse
Affiliation(s)
- Gabriela Mercado
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher Kaeufer
- Center for Systems Neuroscience, Hannover, Germany
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Wouter Peelaerts
- Laboratory for Virology and Gene Therapy, Department of Pharmacy and Pharmaceutical Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
21
|
Yamakado H, Takahashi R. Experimental Animal Models of Prodromal Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2024; 14:S369-S379. [PMID: 38427504 PMCID: PMC11492006 DOI: 10.3233/jpd-230393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 03/03/2024]
Abstract
There is an estimated 35-45% loss of striatal dopamine at the time of diagnosis of Parkinson's disease (PD), and cases clinically diagnosed in the early stages may already be pathologically in advanced stages. Recent large-scale clinical trials of disease-modifying therapies (DMT) also suggest the necessity of targeting patients at earlier stages of the disease. From this perspective, the prodromal phase of PD is currently the focus of attention, emphasizing the need for a prodromal mouse model that accurately reflects the pathophysiology, along with early biomarkers. To establish prodromal animal model of PD with high face validity that reflects the disease state, the model must possess high construct validity that accurately incorporates clinical and pathological features in the prodromal phase. Furthermore, as a preclinical model of DMT, the model must possess high predictive validity to accurately evaluate the response to intervention. This review provides an overview of animal models which reflect the characteristics of prodromal PD, including alpha-synuclein (aS) accumulation and associated early non-motor symptoms, with a focus on the aS propagation model and genetic model. In addition, we discuss the challenges associated with these models. The genetic model often fails to induce motor symptoms, while aS propagation models skip the crucial step of initial aS aggregate formation, thereby not fully replicating the entire natural course of the disease. Identifying factors that induce the transition from prodromal to symptomatic phase is important as a preclinical model for DMT to prevent or delay the onset of the disease.
Collapse
Affiliation(s)
- Hodaka Yamakado
- Department of Therapeutics for Multiple System Atrophy, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
22
|
Lai TT, Gericke B, Feja M, Conoscenti M, Zelikowsky M, Richter F. Anxiety in synucleinopathies: neuronal circuitry, underlying pathomechanisms and current therapeutic strategies. NPJ Parkinsons Dis 2023; 9:97. [PMID: 37349373 DOI: 10.1038/s41531-023-00547-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023] Open
Abstract
Synucleinopathies are neurodegenerative disorders characterized by alpha-synuclein (αSyn) accumulation in neurons or glial cells, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). αSyn-related pathology plays a critical role in the pathogenesis of synucleinopathies leading to the progressive loss of neuronal populations in specific brain regions and the development of motor and non-motor symptoms. Anxiety is among the most frequent non-motor symptoms in patients with PD, but it remains underrecognized and undertreated, which significantly reduces the quality of life for patients. Anxiety is defined as a neuropsychiatric complication with characteristics such as nervousness, loss of concentration, and sweating due to the anticipation of impending danger. In patients with PD, neuropathology in the amygdala, a central region in the anxiety and fear circuitry, may contribute to the high prevalence of anxiety. Studies in animal models reported αSyn pathology in the amygdala together with alteration of anxiety or fear learning response. Therefore, understanding the progression, extent, and specifics of pathology in the anxiety and fear circuitry in synucleinopathies will suggest novel approaches to the diagnosis and treatment of neuropsychiatric symptoms. Here, we provide an overview of studies that address neuropsychiatric symptoms in synucleinopathies. We offer insights into anxiety and fear circuitry in animal models and the current implications for therapeutic intervention. In summary, it is apparent that anxiety is not a bystander symptom in these disorders but reflects early pathogenic mechanisms in the cortico-limbic system which may even contribute as a driver to disease progression.
Collapse
Affiliation(s)
- Thuy Thi Lai
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Birthe Gericke
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Malte Feja
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | | | | | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
23
|
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
|