1
|
Khowdiary MM, Al-Kuraishy HM, Al-Gareeb AI, Albuhadily AK, Elhenawy AA, Babalghith AO, Shokr MM, Alexiou A, Papadakis M, El-Saber Batiha G. Dysregulation of serotonergic neurotransmission in Parkinson disease: A key duet. Eur J Pharmacol 2025; 995:177419. [PMID: 39988096 DOI: 10.1016/j.ejphar.2025.177419] [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: 12/01/2024] [Revised: 02/04/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Parkinson's disease (PD) is the most common movement disorder, affecting approximately 1% of the general population over 65 years of age. PD is commonly associated with the development of motor and non-motor symptoms. Non-motor symptoms arise decades earlier than motor symptoms due to the degeneration of GABAergic, serotonergic, and other neurons involved in autonomic regulation. However, motor symptoms in PD are developed due to degeneration of the dopaminergic neurons in the substantia nigra pars compacta (SNpc) of midbrain. The PD neuropathology is related to the progressive loss of the dopaminergic neurons in the SNpc of midbrain. Particularly, dysfunction of serotonergic system is implicated in the development of non-motor symptoms such as sleep disorders, cognitive dysfunction, depression and anxiety. In addition, dysfunction of serotonergic neurons which affects the dopaminergic neurons in the SNpc leads to the development of motor symptoms. Moreover, dysfunction of serotonergic neurons is associated with the development of L-dopamine (L-DOPA)-induced dyskinesia. Consistently, administration of serotonin (5-HT) receptor agonist attenuates the development of L-DOPA-induced dyskinesia. These findings emphasized the possible role of serotonergic system in PD. However, the underlying mechanisms that mediate the latent effect of 5-HT in PD are not completely elucidated. Therefore, this mini-review aims to discuss the exact role of 5-HT in PD, and how the 5-HT modulators affect PD neuropathology.
Collapse
Affiliation(s)
- Manal M Khowdiary
- Department of Chemistry, Faculty of Applied Science, Lieth Collage, Umm Al-Qura University, Makkah, 24382, Saudi Arabia.
| | - Hayder M Al-Kuraishy
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine Jabir ibn Hayyan Medical University, Al-Ameer Qu., Najaf, Iraq.
| | - Ali K Albuhadily
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq.
| | - Ahmed A Elhenawy
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt; Chemistry Department, Faculty of Science, AlBaha University, Al Bahah, 65731, Saudi Arabia.
| | - Ahmad O Babalghith
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Saudi Arabia.
| | - Mustafa M Shokr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish, 45511, Egypt.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Mohali, India; Department of Research & Development, Funogen, Athens, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, Wuppertal, 42283, Germany.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
2
|
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
|
3
|
Beauchamp LC, Ellett LJ, Juan SMA, Liu XM, Hunt CPJ, Parish CL, Jacobson LH, Shepherd CE, Halliday GM, Bush AI, Vella LJ, Finkelstein DI, Barnham KJ. Evidence of COMT dysfunction in the olfactory bulb in Parkinson's disease. Acta Neuropathol 2025; 149:21. [PMID: 40024917 PMCID: PMC11872990 DOI: 10.1007/s00401-025-02861-y] [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: 10/11/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Hyposmia is one of the most prevalent non-motor symptoms of Parkinson's disease and antecedes motor dysfunction by up to a decade. However, the underlying pathophysiology remains poorly understood. In this study, we investigated the mechanisms of dopamine metabolism in post-mortem olfactory bulbs from ten Parkinson's disease and ten neurologic control subjects. In contrast to the loss of dopaminergic neurons in the midbrain, we observed an increase in tyrosine hydroxylase-positive neurons in the Parkinson's disease olfactory bulb, suggesting a potential role for dopamine in the hyposmia associated with the condition. Using immunohistochemistry, high-performance liquid chromatography, western blot, and enzyme-linked immunosorbent assays, we demonstrate a reduction in catechol-O-methyltransferase catabolism of dopamine to homovanillic acid, potentially due to a depletion of the methyl donor substrate S-adenosyl methionine. We hypothesized that reduction in catechol-O-methyltransferase activity would result in increased dopamine occupation of the D2 receptor, and consequent inhibition of olfactory processing. Next, we conducted pharmacological interventions to modify dopamine dynamics in hyposmic tau knockout mice, which exhibit altered dopamine metabolism. Our hypothesis was supported by the observation that the D2 receptor antagonist haloperidol temporarily alleviated olfactory deficits in these tau knockout mice. This study implicates a potential role of catechol-O-methyltransferase-mediated dopamine metabolism in the early olfactory impairments associated with Parkinson's disease.
Collapse
Affiliation(s)
- Leah C Beauchamp
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- The Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Laura J Ellett
- The Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Sydney M A Juan
- The Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Xiang M Liu
- The Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Cameron P J Hunt
- The Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Clare L Parish
- The Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Laura H Jacobson
- The Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, VIC, 3052, Australia
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC, 3052, Australia
| | | | - Glenda M Halliday
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney Brain and Mind Centre, Camperdown, NSW, Australia
| | - Ashley I Bush
- The Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Laura J Vella
- The Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, VIC, 3052, Australia
- Department of Surgery, University of Melbourne, Parkville, VIC, 3010, Australia
| | - David I Finkelstein
- The Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Kevin J Barnham
- The Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, VIC, 3052, Australia.
| |
Collapse
|
4
|
Leser FS, Júnyor FDS, Pagnoncelli IB, Delgado AB, Medeiros I, Nóbrega ACC, Andrade BDS, de Lima MN, da Silva NE, Jacob L, Boyé K, Geraldo LHM, de Souza AMT, Maron-Gutierrez T, Castro-Faria-Neto H, Follmer C, Braga C, Neves GA, Eichmann A, Romão LF, Lima FRS. CCL21-CCR7 blockade prevents neuroinflammation and degeneration in Parkinson's disease models. J Neuroinflammation 2025; 22:31. [PMID: 39894839 PMCID: PMC11789347 DOI: 10.1186/s12974-024-03318-x] [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: 08/13/2024] [Accepted: 12/02/2024] [Indexed: 02/04/2025] Open
Abstract
Parkinson's disease (PD) is a progressive degenerative disease of the central nervous system associated with neuroinflammation and microglial cell activation. Chemokine signaling regulates neuron-glia communication and triggers a microglial inflammatory profile. Herein, we identified the neuronal chemokine CCL21 as a major cause of microglial cell imbalance through the CCR7 receptor pathway with therapeutic implications for PD. In humans, we found that CCL21 transcript expression was increased in dopaminergic neurons (DANs) of the substantia nigra in PD patients. CCL21 and CCR7 expressions were spatially associated with brain regional vulnerability to synucleinopathies, as well as with the expression of microglial activation, neuroinflammation, and degeneration-related genes. Also, in mouse models of PD, we showed that CCL21 was overexpressed in DANs in vivo and in vitro. Mechanistically, neuronal CCL21 was shown to regulate microglial cell migration, proliferation, and activation in a CCR7-dependent manner through both canonical (PI3K/AKT) and non-canonical (ERK1/2/JNK) signaling pathways. Finally, we demonstrated that navarixin, a clinically relevant chemokine inhibitor with high affinity for the CCR7 receptor, could block CCL21 effects on microglia and prevent neurodegeneration and behavioral deficits in two mouse models of PD induced with either α-synuclein oligomers (αSynO) or 3,4-dihydroxyphenylacetaldehyde (DOPAL). Altogether, our data indicate that navarixin blocks CCL21/CCR7-mediated neuron-microglia communication and could be used as a therapeutic strategy against PD.
Collapse
Affiliation(s)
- Felipe Saceanu Leser
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France
| | - Flavio de Souza Júnyor
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Iohanna Bianca Pagnoncelli
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Anna Beatriz Delgado
- Laboratory of Neurobiology Applied to Biomedicine, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Isabelle Medeiros
- Laboratory of Neurobiology Applied to Biomedicine, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Ana Clara Campanelli Nóbrega
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Brenda da Silva Andrade
- Laboratory of Molecular Pharmacology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Maiara Nascimento de Lima
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Nícolas Emanoel da Silva
- Laboratory Molecular Modeling & QSAR, Pharmaceutical Sciences Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Laurent Jacob
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France
| | - Kevin Boyé
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France
| | - Luiz Henrique Medeiros Geraldo
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France
- Department of Internal Medicine, Department of Cellular and Molecular Physiology, Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, 06510-3221, USA
| | - Alessandra Mendonça Teles de Souza
- Laboratory Molecular Modeling & QSAR, Pharmaceutical Sciences Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Tatiana Maron-Gutierrez
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Hugo Castro-Faria-Neto
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Cristian Follmer
- Laboratory of Physical Chemistry of Proteins and Peptides (Lafipp), Chemistry Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-909, Brazil
| | - Carolina Braga
- Núcleo Multidisciplinar de Pesquisas em Biologia, NUMPEX-Bio, Universidade Federal do Rio de Janeiro, Duque de Caxias, RJ, 25240-005, Brasil
| | - Gilda Angela Neves
- Laboratory of Molecular Pharmacology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Anne Eichmann
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France.
- Department of Internal Medicine, Department of Cellular and Molecular Physiology, Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, 06510-3221, USA.
| | - Luciana Ferreira Romão
- Laboratory of Neurobiology Applied to Biomedicine, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Flavia Regina Souza Lima
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil.
| |
Collapse
|
5
|
O’Niel A, Pederson A, Saltontall E, Nguyen K, Pantoja M, Chaudhari M, Sandholm P, Yoon E, Harrison HF, Boutros S, Hirsch AJ, Raber J. Effects of COVID-19 virus-like particles on the behavioral and cognitive performance of human apolipoprotein E targeted replacement mice. Front Immunol 2024; 15:1473366. [PMID: 39660133 PMCID: PMC11628390 DOI: 10.3389/fimmu.2024.1473366] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction The effects of viral infections might be apolipoprotein E (apoE) isoform-dependent. In humans, there are three major apoE isoforms, E2, E3, and E4. E4 is associated with the enhanced entry of several viruses into the brain and their disease progression. A concern of infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the development of post-acute COVID-19 syndrome, also known as long COVID. Genetic risk factors for developing long COVID were reported. Methods In this study, we used virus-like particles (VLPs) that include expression of the SARS-CoV-2 nucleocapsid (N), membrane (M), and envelope (E) structural proteins together with S. In the current study, we used human E2, E3, and E4 targeted replacement mice to assess whether these VLPs affect body weight, behavioral and cognitive performance, and circadian body temperatures. Using VLPs allow working outside an ABSL-3 facility. Results The effects of VLPs on some behavioral measures were apoE isoform-dependent, with the E2 mice being more affected than E3 or E4 mice. The overall decreased activity in the open field containing objects in week 2 indicate that VLPs can also reduce activity levels in an apoE isoform-independent fashion. Discussion The results of the current study indicate that even in the absence of viral replication, detrimental effects of VLPs on behavioral measures and circadian body temperatures are seen.
Collapse
Affiliation(s)
- Abigail O’Niel
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Alexandra Pederson
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Elizabeth Saltontall
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Kayla Nguyen
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Monzerrat Pantoja
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Mitali Chaudhari
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Phoebe Sandholm
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Eric Yoon
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Henry F. Harrison
- Vaccine and Gene Therapy Center, ONPRC, Oregon Health & Science University, Portland, OR, United States
| | - Sydney Boutros
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Alec J. Hirsch
- Vaccine and Gene Therapy Center, ONPRC, Oregon Health & Science University, Portland, OR, United States
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR, United States
- Division of Neuroscience, Oregon National Primate Research Center (ONPRC), Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
6
|
Lal R, Singh A, Watts S, Chopra K. Experimental models of Parkinson's disease: Challenges and Opportunities. Eur J Pharmacol 2024; 980:176819. [PMID: 39029778 DOI: 10.1016/j.ejphar.2024.176819] [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: 09/07/2023] [Revised: 05/29/2024] [Accepted: 07/17/2024] [Indexed: 07/21/2024]
Abstract
Parkinson's disease (PD) is a widespread neurodegenerative disorder occurs due to the degradation of dopaminergic neurons present in the substantia nigra pars compacta (SNpc). Millions of people are affected by this devastating disorder globally, and the frequency of the condition increases with the increase in the elderly population. A significant amount of progress has been made in acquiring more knowledge about the etiology and the pathogenesis of PD over the past decades. Animal models have been regarded to be a vital tool for the exploration of complex molecular mechanisms involved in PD. Various animals used as models for disease monitoring include vertebrates (zebrafish, rats, mice, guinea pigs, rabbits and monkeys) and invertebrate models (Drosophila, Caenorhabditis elegans). The animal models most relevant for study of PD are neurotoxin induction-based models (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), 6-Hydroxydopamine (6-OHDA) and agricultural pesticides (rotenone, paraquat), pharmacological models (reserpine or haloperidol treated rats), genetic models (α-synuclein, Leucine-rich repeat kinase 2 (LRRK2), DJ-1, PINK-1 and Parkin). Several non-mammalian genetic models such as zebrafish, Drosophila and Caenorhabditis elegance have also gained popularity in recent years due to easy genetic manipulation, presence of genes homologous to human PD, and rapid screening of novel therapeutic molecules. In addition, in vitro models (SH-SY5Y, PC12, Lund human mesencephalic (LUHMES) cells, Human induced pluripotent stem cell (iPSC), Neural organoids, organ-on-chip) are also currently in trend providing edge in investigating molecular mechanisms involved in PD as they are derived from PD patients. In this review, we explain the current situation and merits and demerits of the various animal models.
Collapse
Affiliation(s)
- Roshan Lal
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India.
| | - Aditi Singh
- TR(i)P for Health Laboratory, Centre for Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City, Sector 81, SAS Nagar, Punjab, 140306, India.
| | - Shivam Watts
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India.
| | - Kanwaljit Chopra
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
7
|
He S, Ru Q, Chen L, Xu G, Wu Y. Advances in animal models of Parkinson's disease. Brain Res Bull 2024; 215:111024. [PMID: 38969066 DOI: 10.1016/j.brainresbull.2024.111024] [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: 05/12/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
Parkinson's disease is a complex neurodegenerative disease characterized by progressive movement impairments. Predominant symptoms encompass resting tremor, bradykinesia, limb rigidity, and postural instability. In addition, it also includes a series of non-motor symptoms such as sleep disorders, hyposmia, gastrointestinal dysfunction, autonomic dysfunction and cognitive impairment. Pathologically, the disease manifests through dopaminergic neuronal loss and the presence of Lewy bodies. At present, no significant breakthrough has been achieved in clinical Parkinson's disease treatment. Exploring treatment modalities necessitate the establishment of scientifically sound animal models. In recent years, researchers have focused on replicating the symptoms of human Parkinson's disease, resulting in the establishment of various experimental animal models primarily through drugs and transgenic methods to mimic relevant pathologies and identify more effective treatments. This review examines traditional neurotoxin and transgenic animal models as well as α-synuclein pre-formed fibrils models, non-human primate models and non-mammalian specie models. Additionally, it introduces emerging models, including models based on optogenetics, induced pluripotent stem cells, and gene editing, aiming to provide a reference for the utilization of experimental animal models and clinical research for researchers in this field.
Collapse
Affiliation(s)
- Sui He
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Guodong Xu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
8
|
Guimarães RP, de Resende MCS, Tavares MM, Belardinelli de Azevedo C, Ruiz MCM, Mortari MR. Construct, Face, and Predictive Validity of Parkinson's Disease Rodent Models. Int J Mol Sci 2024; 25:8971. [PMID: 39201659 PMCID: PMC11354451 DOI: 10.3390/ijms25168971] [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: 07/11/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease globally. Current drugs only alleviate symptoms without halting disease progression, making rodent models essential for researching new therapies and understanding the disease better. However, selecting the right model is challenging due to the numerous models and protocols available. Key factors in model selection include construct, face, and predictive validity. Construct validity ensures the model replicates pathological changes seen in human PD, focusing on dopaminergic neurodegeneration and a-synuclein aggregation. Face validity ensures the model's symptoms mirror those in humans, primarily reproducing motor and non-motor symptoms. Predictive validity assesses if treatment responses in animals will reflect those in humans, typically involving classical pharmacotherapies and surgical procedures. This review highlights the primary characteristics of PD and how these characteristics are validated experimentally according to the three criteria. Additionally, it serves as a valuable tool for researchers in selecting the most appropriate animal model based on established validation criteria.
Collapse
Affiliation(s)
- Rayanne Poletti Guimarães
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Maria Clara Souza de Resende
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Miguel Mesquita Tavares
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Caio Belardinelli de Azevedo
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Miguel Cesar Merino Ruiz
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
- Neurological Rehabilitation Unit, Sarah Network of Rehabilitation Hospitals, Brasília 70335-901, Brazil
| | - Márcia Renata Mortari
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| |
Collapse
|
9
|
Bucher ML, Dicent J, Duarte Hospital C, Miller GW. Neurotoxicology of dopamine: Victim or assailant? Neurotoxicology 2024; 103:175-188. [PMID: 38857676 PMCID: PMC11694735 DOI: 10.1016/j.neuro.2024.06.001] [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: 04/13/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
Since the identification of dopamine as a neurotransmitter in the mid-20th century, investigators have examined the regulation of dopamine homeostasis at a basic biological level and in human disorders. Genetic animal models that manipulate the expression of proteins involved in dopamine homeostasis have provided key insight into the consequences of dysregulated dopamine. As a result, we have come to understand the potential of dopamine to act as an endogenous neurotoxin through the generation of reactive oxygen species and reactive metabolites that can damage cellular macromolecules. Endogenous factors, such as genetic variation and subcellular processes, and exogenous factors, such as environmental exposures, have been identified as contributors to the dysregulation of dopamine homeostasis. Given the variety of dysregulating factors that impact dopamine homeostasis and the potential for dopamine itself to contribute to further cellular dysfunction, dopamine can be viewed as both the victim and an assailant of neurotoxicity. Parkinson's disease has emerged as the exemplar case study of dopamine dysregulation due to the genetic and environmental factors known to contribute to disease risk, and due to the evidence of dysregulated dopamine as a pathologic and pathogenic feature of the disease. This review, inspired by the talk, "Dopamine in Durham: location, location, location" presented by Dr. Miller for the Jacob Hooisma Memorial Lecture at the International Neurotoxicology Association meeting in 2023, offers a primer on dopamine toxicity covering endogenous and exogenous factors that disrupt dopamine homeostasis and the actions of dopamine as an endogenous neurotoxin.
Collapse
Affiliation(s)
- Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Jocelyn Dicent
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Carolina Duarte Hospital
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA; Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
10
|
Boi L, Johansson Y, Tonini R, Moratalla R, Fisone G, Silberberg G. Serotonergic and dopaminergic neurons in the dorsal raphe are differentially altered in a mouse model for parkinsonism. eLife 2024; 12:RP90278. [PMID: 38940422 PMCID: PMC11213571 DOI: 10.7554/elife.90278] [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] [Indexed: 06/29/2024] Open
Abstract
Parkinson's disease (PD) is characterized by motor impairments caused by degeneration of dopamine neurons in the substantia nigra pars compacta. In addition to these symptoms, PD patients often suffer from non-motor comorbidities including sleep and psychiatric disturbances, which are thought to depend on concomitant alterations of serotonergic and noradrenergic transmission. A primary locus of serotonergic neurons is the dorsal raphe nucleus (DRN), providing brain-wide serotonergic input. Here, we identified electrophysiological and morphological parameters to classify serotonergic and dopaminergic neurons in the murine DRN under control conditions and in a PD model, following striatal injection of the catecholamine toxin, 6-hydroxydopamine (6-OHDA). Electrical and morphological properties of both neuronal populations were altered by 6-OHDA. In serotonergic neurons, most changes were reversed when 6-OHDA was injected in combination with desipramine, a noradrenaline (NA) reuptake inhibitor, protecting the noradrenergic terminals. Our results show that the depletion of both NA and dopamine in the 6-OHDA mouse model causes changes in the DRN neural circuitry.
Collapse
Affiliation(s)
- Laura Boi
- Department of Neuroscience, Karolinska InstituteStockholmSweden
| | - Yvonne Johansson
- Department of Neuroscience, Karolinska InstituteStockholmSweden
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College LondonLondonUnited Kingdom
| | - Raffaella Tonini
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Istituto Italiano di TecnologiaGenovaItaly
| | - Rosario Moratalla
- Cajal Institute, Spanish National Research Council (CSIC)MadridSpain
- CIBERNED, Instituto de Salud Carlos IIIMadridSpain
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska InstituteStockholmSweden
| | | |
Collapse
|
11
|
Bucher ML, Dunn AR, Bradner JM, Stout Egerton K, Burkett JP, Johnson MA, Miller GW. Synaptic vesicle glycoprotein 2C enhances vesicular storage of dopamine and counters dopaminergic toxicity. Eur J Neurosci 2024; 59:2483-2501. [PMID: 38532289 PMCID: PMC11647951 DOI: 10.1111/ejn.16311] [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: 06/20/2023] [Revised: 02/14/2024] [Accepted: 02/22/2024] [Indexed: 03/28/2024]
Abstract
Dopaminergic neurons of the substantia nigra exist in a persistent state of vulnerability resulting from high baseline oxidative stress, high-energy demand, and broad unmyelinated axonal arborisations. Impairments in the storage of dopamine compound this stress because of cytosolic reactions that transform the vital neurotransmitter into an endogenous neurotoxicant, and this toxicity is thought to contribute to the dopamine neuron degeneration that occurs Parkinson's disease. We have previously identified synaptic vesicle glycoprotein 2C (SV2C) as a modifier of vesicular dopamine function, demonstrating that genetic ablation of SV2C in mice results in decreased dopamine content and evoked dopamine release in the striatum. Here, we adapted a previously published in vitro assay utilising false fluorescent neurotransmitter 206 (FFN206) to visualise how SV2C regulates vesicular dopamine dynamics and determined that SV2C promotes the uptake and retention of FFN206 within vesicles. In addition, we present data indicating that SV2C enhances the retention of dopamine in the vesicular compartment with radiolabelled dopamine in vesicles isolated from immortalised cells and from mouse brain. Further, we demonstrate that SV2C enhances the ability of vesicles to store the neurotoxicant 1-methyl-4-phenylpyridinium (MPP+) and that genetic ablation of SV2C results in enhanced 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced vulnerability in mice. Together, these findings suggest that SV2C functions to enhance vesicular storage of dopamine and neurotoxicants and helps maintain the integrity of dopaminergic neurons.
Collapse
Affiliation(s)
- Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Amy R Dunn
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Joshua M Bradner
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Kristen Stout Egerton
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - James P Burkett
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Michelle A Johnson
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York NY 10031, USA
| |
Collapse
|
12
|
Laurencin C, Lancelot S, Brosse S, Mérida I, Redouté J, Greusard E, Lamberet L, Liotier V, Le Bars D, Costes N, Thobois S, Boulinguez P, Ballanger B. Noradrenergic alterations in Parkinson's disease: a combined 11C-yohimbine PET/neuromelanin MRI study. Brain 2024; 147:1377-1388. [PMID: 37787503 PMCID: PMC10994534 DOI: 10.1093/brain/awad338] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 10/04/2023] Open
Abstract
Degeneration of the noradrenergic system is now considered a pathological hallmark of Parkinson's disease, but little is known about its consequences in terms of parkinsonian manifestations. Here, we evaluated two aspects of the noradrenergic system using multimodal in vivo imaging in patients with Parkinson's disease and healthy controls: the pigmented cell bodies of the locus coeruleus with neuromelanin sensitive MRI; and the density of α2-adrenergic receptors (ARs) with PET using 11C-yohimbine. Thirty patients with Parkinson's disease and 30 age- and sex-matched healthy control subjects were included. The characteristics of the patients' symptoms were assessed using the Movement Disorder Society Unified Parkinson's Disease Rating Scale (MDS-UPDRS). Patients showed reduced neuromelanin signal intensity in the locus coeruleus compared with controls and diminished 11C-yohimbine binding in widespread cortical regions, including the motor cortex, as well as in the insula, thalamus and putamen. Clinically, locus coeruleus neuronal loss was correlated with motor (bradykinesia, motor fluctuations, tremor) and non-motor (fatigue, apathy, constipation) symptoms. A reduction of α2-AR availability in the thalamus was associated with tremor, while a reduction in the putamen, the insula and the superior temporal gyrus was associated with anxiety. These results highlight a multifaceted alteration of the noradrenergic system in Parkinson's disease since locus coeruleus and α2-AR degeneration were found to be partly uncoupled. These findings raise important issues about noradrenergic dysfunction that may encourage the search for new drugs targeting this system, including α2-ARs, for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Chloé Laurencin
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
- Department of Neurology C, Expert Parkinson Centre, Hospices Civils de Lyon, Pierre Wertheimer Neurological Hospital, NS-Park/F-CRIN, 69500 Bron, France
| | - Sophie Lancelot
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Sarah Brosse
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
| | - Inés Mérida
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Jérôme Redouté
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Elise Greusard
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Ludovic Lamberet
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | | | - Didier Le Bars
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Nicolas Costes
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Stéphane Thobois
- Department of Neurology C, Expert Parkinson Centre, Hospices Civils de Lyon, Pierre Wertheimer Neurological Hospital, NS-Park/F-CRIN, 69500 Bron, France
- Institut des Sciences Cognitives Marc Jeannerod, UMR 5229, CNRS, 69500 Bron, France
| | - Philippe Boulinguez
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
| | - Bénédicte Ballanger
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
| |
Collapse
|
13
|
Chaklai A, O’Neil A, Goel S, Margolies N, Krenik D, Perez R, Kessler K, Staltontall E, Yoon HK(E, Pantoja M, Stagaman K, Kasschau K, Unni V, Duvoisin R, Sharpton T, Raber J. Effects of Paraquat, Dextran Sulfate Sodium, and Irradiation on Behavioral and Cognitive Performance and the Gut Microbiome in A53T and A53T-L444P Mice. Genes (Basel) 2024; 15:282. [PMID: 38540341 PMCID: PMC11154584 DOI: 10.3390/genes15030282] [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: 01/13/2024] [Revised: 01/31/2024] [Accepted: 02/21/2024] [Indexed: 06/09/2024] Open
Abstract
Heterozygous carriers of the glucocerebrosidase 1 (GBA) L444P Gaucher mutation have an increased risk of developing Parkinson's disease (PD). The GBA mutations result in elevated alpha synuclein (aSyn) levels. Heterozygous mice carrying one allele with the L444P mutation knocked-into the mouse gene show increased aSyn levels and are more sensitive to motor deficits following exposure to the neurotoxin (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) MPTP than wild-type mice. Paraquat (PQ), a herbicide, increases PD risk in most studies. Its effects on the brain involve alterations in the gut microbiome. Exposure to dextran sulfate sodium (DSS), a mouse model of colitis, can be used to determine whether gut microbiome alterations are sufficient to induce PD-relevant phenotypes. We rederived the A53T-L444P and A53T mouse lines to assess whether PQ, PQ in combination with radiation exposure (IR), and DSS have differential effects in A53T and A53T-L444P mice and whether these effects are associated with alterations in the gut microbiome. PQ and PQ + IR have differential effects in A53T and A53T-L444P mice. In contrast, effects of DSS are only seen in A53T-L444P mice. Exposure and genotype modulate the relationship between the gut microbiome and behavioral performance. The gut microbiome may be an important mediator of how environmental exposures or genetic mutations yield behavioral and cognitive impacts.
Collapse
Affiliation(s)
- Ariel Chaklai
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (A.C.); (A.O.); (S.G.); (N.M.); (D.K.); (R.P.); (K.K.); (E.S.); (M.P.)
| | - Abigail O’Neil
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (A.C.); (A.O.); (S.G.); (N.M.); (D.K.); (R.P.); (K.K.); (E.S.); (M.P.)
| | - Shrey Goel
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (A.C.); (A.O.); (S.G.); (N.M.); (D.K.); (R.P.); (K.K.); (E.S.); (M.P.)
| | - Nick Margolies
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (A.C.); (A.O.); (S.G.); (N.M.); (D.K.); (R.P.); (K.K.); (E.S.); (M.P.)
| | - Destine Krenik
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (A.C.); (A.O.); (S.G.); (N.M.); (D.K.); (R.P.); (K.K.); (E.S.); (M.P.)
| | - Ruby Perez
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (A.C.); (A.O.); (S.G.); (N.M.); (D.K.); (R.P.); (K.K.); (E.S.); (M.P.)
| | - Kat Kessler
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (A.C.); (A.O.); (S.G.); (N.M.); (D.K.); (R.P.); (K.K.); (E.S.); (M.P.)
| | - Elizabeth Staltontall
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (A.C.); (A.O.); (S.G.); (N.M.); (D.K.); (R.P.); (K.K.); (E.S.); (M.P.)
| | - Hong Ki (Eric) Yoon
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (A.C.); (A.O.); (S.G.); (N.M.); (D.K.); (R.P.); (K.K.); (E.S.); (M.P.)
| | - Montzerrat Pantoja
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (A.C.); (A.O.); (S.G.); (N.M.); (D.K.); (R.P.); (K.K.); (E.S.); (M.P.)
| | - Keaton Stagaman
- Department of Microbiology, Oregon State University, Corvallis, OR 97331, USA; (K.S.); (K.K.); (T.S.)
| | - Kristin Kasschau
- Department of Microbiology, Oregon State University, Corvallis, OR 97331, USA; (K.S.); (K.K.); (T.S.)
| | - Vivek Unni
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA;
- Jungers Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Robert Duvoisin
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA;
| | - Thomas Sharpton
- Department of Microbiology, Oregon State University, Corvallis, OR 97331, USA; (K.S.); (K.K.); (T.S.)
- Department of Statistics, Oregon State University, Corvallis, OR 97331, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; (A.C.); (A.O.); (S.G.); (N.M.); (D.K.); (R.P.); (K.K.); (E.S.); (M.P.)
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA;
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
14
|
Chen YC, Wang WS, Lewis SJG, Wu SL. Fighting Against the Clock: Circadian Disruption and Parkinson's Disease. J Mov Disord 2024; 17:1-14. [PMID: 37989149 PMCID: PMC10846969 DOI: 10.14802/jmd.23216] [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: 10/19/2023] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 11/23/2023] Open
Abstract
Circadian disruption is being increasingly recognized as a critical factor in the development and progression of Parkinson's disease (PD). This review aims to provide an in-depth overview of the relationship between circadian disruption and PD by exploring the molecular, cellular, and behavioral aspects of this interaction. This review will include a comprehensive understanding of how the clock gene system and transcription-translation feedback loops function and how they are diminished in PD. The article also discusses the role of clock genes in the regulation of circadian rhythms, as well as the impact of clock gene dysregulation on mitochondrial function, oxidative stress, and neuroinflammation, including the microbiota-gut-brain axis, which have all been proposed as being crucial mechanisms in the pathophysiology of PD. Finally, this review highlights potential therapeutic strategies targeting the clock gene system and circadian rhythm for the treatment of PD.
Collapse
Affiliation(s)
- Yen-Chung Chen
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan
- Department of Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Wei-Sheng Wang
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan
| | - Simon J G Lewis
- Brain and Mind Centre, School of Medical Sciences, The University of Sydney, Camperdown, New South Wales, Australia
| | - Shey-Lin Wu
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan
- Department of Electrical Engineering, National Changhua University of Education, Changhua, Taiwan
| |
Collapse
|
15
|
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
|
16
|
Boyd SL, Kuhn NC, Patterson JR, Stoll AC, Zimmerman SA, Kolanowski MR, Neubecker JJ, Luk KC, Ramsson ES, Sortwell CE, Bernstein AI. Developmental exposure to the Parkinson's disease-associated organochlorine pesticide dieldrin alters dopamine neurotransmission in α-synuclein pre-formed fibril (PFF)-injected mice. Toxicol Sci 2023; 196:99-111. [PMID: 37607008 PMCID: PMC10613968 DOI: 10.1093/toxsci/kfad086] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023] Open
Abstract
Parkinson's disease (PD) is the fastest-growing neurological disease worldwide, with increases outpacing aging and occurring most rapidly in recently industrialized areas, suggesting a role of environmental factors. Epidemiological, post-mortem, and mechanistic studies suggest that persistent organic pollutants, including the organochlorine pesticide dieldrin, increase PD risk. In mice, developmental dieldrin exposure causes male-specific exacerbation of neuronal susceptibility to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and synucleinopathy. Specifically, in the α-synuclein (α-syn) pre-formed fibril (PFF) model, exposure leads to increased deficits in striatal dopamine (DA) turnover and motor deficits on the challenging beam. Here, we hypothesized that alterations in DA handling contribute to the observed changes and assessed vesicular monoamine transporter 2 (VMAT2) function and DA release in this dieldrin/PFF 2-hit model. Female C57BL/6 mice were exposed to 0.3 mg/kg dieldrin or vehicle every 3 days by feeding, starting at 8 weeks of age and continuing throughout breeding, gestation, and lactation. Male offspring from independent litters underwent unilateral, intrastriatal injections of α-syn PFFs at 12 weeks of age, and vesicular 3H-DA uptake assays and fast-scan cyclic voltammetry were performed 4 months post-PFF injection. Dieldrin-induced an increase in DA release in striatal slices in PFF-injected animals, but no change in VMAT2 activity. These results suggest that developmental dieldrin exposure increases a compensatory response to synucleinopathy-triggered striatal DA loss. These findings are consistent with silent neurotoxicity, where developmental exposure to dieldrin primes the nigrostriatal striatal system to have an exacerbated response to synucleinopathy in the absence of observable changes in typical markers of nigrostriatal dysfunction and degeneration.
Collapse
Affiliation(s)
- Sierra L Boyd
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Nathan C Kuhn
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Joseph R Patterson
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Anna C Stoll
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Sydney A Zimmerman
- Biomedical Sciences Department, Grand Valley State University, Allendale, MI, USA
| | - Mason R Kolanowski
- Biomedical Sciences Department, Grand Valley State University, Allendale, MI, USA
| | - Joseph J Neubecker
- Biomedical Sciences Department, Grand Valley State University, Allendale, MI, USA
| | - Kelvin C Luk
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric S Ramsson
- Biomedical Sciences Department, Grand Valley State University, Allendale, MI, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Alison I Bernstein
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
- Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, NJ, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
17
|
Ren Z, Sun H, Xiu S, Yang N, Liu Y, Chan P. Investigation of rhodamine derivative on behavioral impairment in a double neurotoxin lesion of substantia nigra and locus coeruleus dysfunctional mice. Eur J Pharmacol 2023; 956:175944. [PMID: 37536627 DOI: 10.1016/j.ejphar.2023.175944] [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: 05/03/2023] [Revised: 07/11/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Although multiple mechanisms have been studied, there is still a lack of effective treatment on non-motor symptoms in Parkinson's disease (PD) patients. Therapeutic effects of 5-(4-hydroxy-3-dimethoxybenzylidene)-thiazolidinone (RD-1), one of rhodamine derivatives, on motor recovery have been previously demonstrated, but its effects on non-motor symptoms remain unclear. Herein, we explored the beneficial effects of RD-1 on PD-related non-motor symptoms and changes in synaptic plasticity in the mesencephalon. To investigate its therapeutic effects in the non-motor symptoms of Parkinsonian model, we employed male C57BL/6N mice and double injection with noradrenergic specific neurotoxin N-2-Chloroethyl-N-ethyl-2-bromobenzylamine hydrochloride, followed 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Next, we performed behavioral tests, histological analyses and immunoblotting. Our findings showed that RD-1 significantly alleviated locomotor abnormality, motor disturbance, anxiety/depression-like behavior and memory deficit. It rescued the levels of tyrosine hydroxylase in substantia nigra, and striatum. Moreover, RD-1 upregulated expression levels of α-synuclein, synapsin II, postsynaptic density 95 and vesicle-associated membrane protein 2. The restoration of synaptic function may underlie the neuroprotective effects of RD-1 in double lesioned mice, confirming its protective effect for dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Zhili Ren
- Department of Neurobiology, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing Institute of Geriatrics, Beijing, China.
| | - Hong Sun
- Department of Neurobiology, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing Institute of Geriatrics, Beijing, China
| | - Shuangling Xiu
- Department of Endocrinology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Nan Yang
- Department of Pharmacology, Institute of Basic Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Yanyong Liu
- Department of Pharmacology, Institute of Basic Medical Sciences, Peking Union Medical College, Beijing, 100005, China.
| | - Piu Chan
- Department of Neurobiology, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing Institute of Geriatrics, Beijing, China; Clinical Center for Parkinson's Disease, Capital Medical University, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Beijing, China; Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Boi L, Fisone G. Investigating affective neuropsychiatric symptoms in rodent models of Parkinson's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 174:119-186. [PMID: 38341228 DOI: 10.1016/bs.irn.2023.08.006] [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: 02/12/2024]
Abstract
Affective neuropsychiatric disorders such as depression, anxiety and apathy are among the most frequent non-motor symptoms observed in people with Parkinson's disease (PD). These conditions often emerge during the prodromal phase of the disease and are generally considered to result from neurodegenerative processes in meso-corticolimbic structures, occurring in parallel to the loss of nigrostriatal dopaminergic neurons. Depression, anxiety, and apathy are often treated with conventional medications, including selective serotonin reuptake inhibitors, tricyclic antidepressants, and dopaminergic agonists. The ability of these pharmacological interventions to consistently counteract such neuropsychiatric symptoms in PD is still relatively limited and the development of reliable experimental models represents an important tool to identify more effective treatments. This chapter provides information on rodent models of PD utilized to study these affective neuropsychiatric symptoms. Neurotoxin-based and genetic models are discussed, together with the main behavioral tests utilized to identify depression- and anxiety-like behaviors, anhedonia, and apathy. The ability of various therapeutic approaches to counteract the symptoms observed in the various models is also reviewed.
Collapse
Affiliation(s)
- Laura Boi
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
19
|
Bucher ML, Dunn AR, Bradner JM, Egerton KS, Burkett JP, Johnson MA, Miller GW. Synaptic vesicle glycoprotein 2C enhances vesicular storage of dopamine and counters dopaminergic toxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.26.546143. [PMID: 37425736 PMCID: PMC10326994 DOI: 10.1101/2023.06.26.546143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Dopaminergic neurons of the substantia nigra exist in a persistent state of vulnerability resulting from high baseline oxidative stress, high energy demand, and broad unmyelinated axonal arborizations. Impairments in the storage of dopamine compound this stress due to cytosolic reactions that transform the vital neurotransmitter into an endogenous neurotoxicant, and this toxicity is thought to contribute to the dopamine neuron degeneration that occurs Parkinson's disease. We have previously identified synaptic vesicle glycoprotein 2C (SV2C) as a modifier of vesicular dopamine function, demonstrating that genetic ablation of SV2C in mice results in decreased dopamine content and evoked dopamine release in the striatum. Here, we adapted a previously published in vitro assay utilizing false fluorescent neurotransmitter 206 (FFN206) to visualize how SV2C regulates vesicular dopamine dynamics and determined that SV2C promotes the uptake and retention of FFN206 within vesicles. In addition, we present data indicating that SV2C enhances the retention of dopamine in the vesicular compartment with radiolabeled dopamine in vesicles isolated from immortalized cells and from mouse brain. Further, we demonstrate that SV2C enhances the ability of vesicles to store the neurotoxicant 1-methyl-4-phenylpyridinium (MPP+) and that genetic ablation of SV2C results in enhanced 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced vulnerability in mice. Together, these findings suggest that SV2C functions to enhance vesicular storage of dopamine and neurotoxicants, and helps maintain the integrity of dopaminergic neurons.
Collapse
Affiliation(s)
- Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Amy R Dunn
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Joshua M Bradner
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Kristen Stout Egerton
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - James P Burkett
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Michelle A Johnson
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10031, USA
| |
Collapse
|
20
|
Ray Chaudhuri K, Leta V, Bannister K, Brooks DJ, Svenningsson P. The noradrenergic subtype of Parkinson disease: from animal models to clinical practice. Nat Rev Neurol 2023:10.1038/s41582-023-00802-5. [PMID: 37142796 DOI: 10.1038/s41582-023-00802-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 05/06/2023]
Abstract
Many advances in understanding the pathophysiology of Parkinson disease (PD) have been based on research addressing its motor symptoms and phenotypes. Various data-driven clinical phenotyping studies supported by neuropathological and in vivo neuroimaging data suggest the existence of distinct non-motor endophenotypes of PD even at diagnosis, a concept further strengthened by the predominantly non-motor spectrum of symptoms in prodromal PD. Preclinical and clinical studies support early dysfunction of noradrenergic transmission in both the CNS and peripheral nervous system circuits in patients with PD that results in a specific cluster of non-motor symptoms, including rapid eye movement sleep behaviour disorder, pain, anxiety and dysautonomia (particularly orthostatic hypotension and urinary dysfunction). Cluster analyses of large independent cohorts of patients with PD and phenotype-focused studies have confirmed the existence of a noradrenergic subtype of PD, which had been previously postulated but not fully characterized. This Review discusses the translational work that unravelled the clinical and neuropathological processes underpinning the noradrenergic PD subtype. Although some overlap with other PD subtypes is inevitable as the disease progresses, recognition of noradrenergic PD as a distinct early disease subtype represents an important advance towards the delivery of personalized medicine for patients with PD.
Collapse
Affiliation(s)
- K Ray Chaudhuri
- Department of Basic and Clinical Neurosciences, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK.
| | - Valentina Leta
- Department of Basic and Clinical Neurosciences, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK
| | - Kirsty Bannister
- Central Modulation of Pain Lab, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - David J Brooks
- Institute of Translational and Clinical Research, University of Newcastle upon Tyne, Newcastle, UK
- Department of Nuclear Medicine, Aarhus University, Aarhus, Denmark
| | - Per Svenningsson
- Department of Basic and Clinical Neurosciences, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
21
|
Chen K, Zhang L, Wang F, Mao H, Tang Q, Shi G, You Y, Yuan Q, Chen B, Fang X. Altered functional connectivity within the brain fear circuit in Parkinson's disease with anxiety: A seed-based functional connectivity study. Heliyon 2023; 9:e15871. [PMID: 37305477 PMCID: PMC10256910 DOI: 10.1016/j.heliyon.2023.e15871] [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: 07/11/2022] [Revised: 04/06/2023] [Accepted: 04/24/2023] [Indexed: 06/13/2023] Open
Abstract
Objectives Aimed to investigate whether there are abnormal changes in the functional connectivity (FC) between the amygdala with other brain areas, in Parkinson's disease (PD) patients with anxiety. Methods Participants were enrolled prospectively, and the Hamilton Anxiety Rating (HAMA) Scale was used to quantify anxiety disorder. Rest-state functional MRI (rs-fMRI) was applied to analyze the amygdala FC patterns among anxious PD patients, non-anxious PD patients, and healthy controls. Results Thirty-three PD patients were recruited, 13 with anxiety, 20 without anxiety, and 19 non-anxious healthy controls. In anxious PD patients, FC between the amygdala with the hippocampus, putamen, intraparietal sulcus, and precuneus showed abnormal alterations compared with non-anxious PD patients and healthy controls. In particular, FC between the amygdala and hippocampus negatively correlated with the HAMA score (r = -0.459, p = 0.007). Conclusion Our results support the role of the fear circuit in emotional regulation in PD with anxiety. Also, the abnormal FC patterns of the amygdala could preliminarily explain the neural mechanisms of anxiety in PD.
Collapse
Affiliation(s)
- Kaidong Chen
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Liangxi District, Wuxi, 214023, Jiangsu Province, China
| | - Li Zhang
- Department of Neurology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Liangxi District, Wuxi, 214023, Jiangsu Province, China
| | - Feng Wang
- Department of Neurology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Liangxi District, Wuxi, 214023, Jiangsu Province, China
| | - Haixia Mao
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Liangxi District, Wuxi, 214023, Jiangsu Province, China
| | - Qunfeng Tang
- Department of Neurology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Liangxi District, Wuxi, 214023, Jiangsu Province, China
| | - Guofeng Shi
- Department of Neurology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Liangxi District, Wuxi, 214023, Jiangsu Province, China
| | - Yiping You
- Department of Neurology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Liangxi District, Wuxi, 214023, Jiangsu Province, China
| | - Qingfang Yuan
- Department of Neurology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Liangxi District, Wuxi, 214023, Jiangsu Province, China
| | - Bixue Chen
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Liangxi District, Wuxi, 214023, Jiangsu Province, China
| | - Xiangming Fang
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Liangxi District, Wuxi, 214023, Jiangsu Province, China
| |
Collapse
|
22
|
The Role of α-Synuclein in the Regulation of Serotonin System: Physiological and Pathological Features. Biomedicines 2023; 11:biomedicines11020541. [PMID: 36831077 PMCID: PMC9953742 DOI: 10.3390/biomedicines11020541] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
In patients affected by Parkinson's disease (PD), up to 50% of them experience cognitive changes, and psychiatric disturbances, such as anxiety and depression, often precede the onset of motor symptoms and have a negative impact on their quality of life. Pathologically, PD is characterized by the loss of dopamine (DA) neurons in the substantia nigra pars compacta (SNc) and the presence of intracellular inclusions, called Lewy bodies and Lewy neurites, composed mostly of α-synuclein (α-Syn). Much of PD research has focused on the role of α-Syn aggregates in the degeneration of SNc DA neurons due to the impact of striatal DA deficits on classical motor phenotypes. However, abundant Lewy pathology is also found in other brain regions including the midbrain raphe nuclei, which may contribute to non-motor symptoms. Indeed, dysfunction of the serotonergic (5-HT) system, which regulates mood and emotional pathways, occurs during the premotor phase of PD. However, little is known about the functional consequences of α-Syn inclusions in this neuronal population other than DA neurons. Here, we provide an overview of the current knowledge of α-Syn and its role in regulating the 5-HT function in health and disease. Understanding the relative contributions to α-Syn-linked alterations in the 5-HT system may provide a basis for identifying PD patients at risk for developing depression and could lead to a more targeted therapeutic approach.
Collapse
|
23
|
Song S, Tu D, Meng C, Liu J, Wilson B, Wang Q, Shih YYI, Gao HM, Hong JS. Dysfunction of the noradrenergic system drives inflammation, α-synucleinopathy, and neuronal loss in mouse colon. Front Immunol 2023; 14:1083513. [PMID: 36845109 PMCID: PMC9950510 DOI: 10.3389/fimmu.2023.1083513] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/01/2023] [Indexed: 02/12/2023] Open
Abstract
Clinical and pathological evidence revealed that α-synuclein (α-syn) pathology seen in PD patients starts in the gut and spreads via anatomically connected structures from the gut to the brain. Our previous study demonstrated that depletion of central norepinephrine (NE) disrupted brain immune homeostasis, producing a spatiotemporal order of neurodegeneration in the mouse brain. The purpose of this study was 1) to determine the role of peripheral noradrenergic system in the maintenance of gut immune homeostasis and in the pathogenesis of PD and 2) to investigate whether NE-depletion induced PD-like α-syn pathological changes starts from the gut. For these purposes, we investigated time-dependent changes of α-synucleinopathy and neuronal loss in the gut following a single injection of DSP-4 (a selective noradrenergic neurotoxin) to A53T-SNCA (human mutant α-syn) over-expression mice. We found DPS-4 significantly reduced the tissue level of NE and increased immune activities in gut, characterized by increased number of phagocytes and proinflammatory gene expression. Furthermore, a rapid-onset of α-syn pathology was observed in enteric neurons after 2 weeks and delayed dopaminergic neurodegeneration in the substantia nigra was detected after 3-5 months, associated with the appearance of constipation and impaired motor function, respectively. The increased α-syn pathology was only observed in large, but not in the small, intestine, which is similar to what was observed in PD patients. Mechanistic studies reveal that DSP-4-elicited upregulation of NADPH oxidase (NOX2) initially occurred only in immune cells during the acute intestinal inflammation stage, and then spread to enteric neurons and mucosal epithelial cells during the chronic inflammation stage. The upregulation of neuronal NOX2 correlated well with the extent of α-syn aggregation and subsequent enteric neuronal loss, suggesting that NOX2-generated reactive oxygen species play a key role in α-synucleinopathy. Moreover, inhibiting NOX2 by diphenyleneiodonium or restoring NE function by salmeterol (a β2-receptor agonist) significantly attenuated colon inflammation, α-syn aggregation/propagation, and enteric neurodegeneration in the colon and ameliorated subsequent behavioral deficits. Taken together, our model of PD shows a progressive pattern of pathological changes from the gut to the brain and suggests a potential role of the noradrenergic dysfunction in the pathogenesis of PD.
Collapse
Affiliation(s)
- Sheng Song
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
- Biomedical Research Imaging Center, University of North Caroline at Chapel Hill, Chapel Hill, NC, United States
| | - Dezhen Tu
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Institute for Brain Sciences, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of medicine, Nanjing University, Nanjing, China
| | - Chengbo Meng
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Jie Liu
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Belinda Wilson
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Qingshan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Yen-Yu Ian Shih
- Biomedical Research Imaging Center, University of North Caroline at Chapel Hill, Chapel Hill, NC, United States
| | - Hui-Ming Gao
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Institute for Brain Sciences, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of medicine, Nanjing University, Nanjing, China
| | - Jau-Shyong Hong
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| |
Collapse
|
24
|
Zeng F, Parker K, Zhan Y, Miller M, Zhu MY. Upregulated DNA Damage-Linked Biomarkers in Parkinson's Disease Model Mice. ASN Neuro 2023; 15:17590914231152099. [PMID: 36683340 PMCID: PMC9880594 DOI: 10.1177/17590914231152099] [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: 08/05/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/24/2023] Open
Abstract
SUMMARY STATEMENT The present study examined expression of DNA damage markers in VMAT2 Lo PD model mice. The results demonstrate there is a significant increase in these DNA damage markers mostly in the brain regions of 18- and 23-month-old model mice, indicating oxidative stress-induced DNA lesion is an important pathologic feature of this mouse model.
Collapse
Affiliation(s)
- Fei Zeng
- Department of Neurology, Renmin Hospital of the Wuhan University,
Wuhan, China
- Departments of Biomedical Sciences, Quillen College of Medicine, East Tennessee State
University, Johnson City, TN, USA
| | - Karsten Parker
- Departments of Biomedical Sciences, Quillen College of Medicine, East Tennessee State
University, Johnson City, TN, USA
| | - Yanqiang Zhan
- Department of Neurology, Renmin Hospital of the Wuhan University,
Wuhan, China
- Departments of Biomedical Sciences, Quillen College of Medicine, East Tennessee State
University, Johnson City, TN, USA
| | - Matthew Miller
- Departments of Biomedical Sciences, Quillen College of Medicine, East Tennessee State
University, Johnson City, TN, USA
| | - Meng-Yang Zhu
- Departments of Biomedical Sciences, Quillen College of Medicine, East Tennessee State
University, Johnson City, TN, USA
| |
Collapse
|
25
|
Weerasinghe-Mudiyanselage PD, Kang S, Kim JS, Moon C. Therapeutic Approaches to Non-Motor Symptoms of Parkinson's Disease: A Current Update on Preclinical Evidence. Curr Neuropharmacol 2023; 21:560-577. [PMID: 36200159 PMCID: PMC10207906 DOI: 10.2174/1570159x20666221005090126] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/22/2022] Open
Abstract
Despite being classified as a movement disorder, Parkinson's disease (PD) is characterized by a wide range of non-motor symptoms that significantly affect the patients' quality of life. However, clear evidence-based therapy recommendations for non-motor symptoms of PD are uncommon. Animal models of PD have previously been shown to be useful for advancing the knowledge and treatment of motor symptoms. However, these models may provide insight into and assess therapies for non-motor symptoms in PD. This paper highlights non-motor symptoms in preclinical models of PD and the current position regarding preclinical therapeutic approaches for these non-motor symptoms. This information may be relevant for designing future preclinical investigations of therapies for nonmotor symptoms in PD.
Collapse
Affiliation(s)
- Poornima D.E. Weerasinghe-Mudiyanselage
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, South Korea
| | - Sohi Kang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, South Korea
| | - Joong-Sun Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, South Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, South Korea
| |
Collapse
|
26
|
Tsarouchi M, Fanarioti E, Karathanos VT, Dermon CR. Protective Effects of Currants ( Vitis vinifera) on Corticolimbic Serotoninergic Alterations and Anxiety-like Comorbidity in a Rat Model of Parkinson's Disease. Int J Mol Sci 2022; 24:ijms24010462. [PMID: 36613906 PMCID: PMC9820698 DOI: 10.3390/ijms24010462] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the loss of nigral dopaminergic neurons. Increasing evidence supports that PD is not simply a motor disorder but a systemic disease leading to motor and non-motor symptoms, including memory loss and neuropsychiatric conditions, with poor management of the non-motor deficits by the existing dopaminergic medication. Oxidative stress is considered a contributing factor for nigrostriatal degeneration, while antioxidant/anti-inflammatory properties of natural phyto-polyphenols have been suggested to have beneficial effects. The present study aimed to determine the contribution of monoaminergic neurotransmission on the anxiety-like phenotype in a rat rotenone PD model and evaluate the possible neuroprotective effects of black Corinthian currant, Vitis vinifera, consisting of antioxidant polyphenols. Rotenone-treated rats showed anxiety-like behavior and exploratory deficits, accompanied by changes in 5-HT, SERT and β2-ARs expression in the prefrontal cortices, hippocampus and basolateral amygdala. Importantly, the motor and non-motor behavior, as well as 5-HT, SERT and β2-ARs expression patterns of the PD-like phenotype were partially recovered by a supplementary diet with currants. Overall, our results suggest that the neuroprotective effects of Corinthian currants in rotenone-induced anxiety-like behavior may be mediated via corticolimbic serotonergic transmission.
Collapse
Affiliation(s)
- Martha Tsarouchi
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, 265 00 Patras, Greece
| | - Eleni Fanarioti
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, 265 00 Patras, Greece
| | - Vaios T. Karathanos
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Dietetics and Nutrition, Harokopio University, 176 76 Kallithea, Greece
- Agricultural Cooperatives’ Union of Aeghion, Corinthou 201, 251 00 Aeghion, Greece
| | - Catherine R. Dermon
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, 265 00 Patras, Greece
- Correspondence:
| |
Collapse
|
27
|
How Well Do Rodent Models of Parkinson's Disease Recapitulate Early Non-Motor Phenotypes? A Systematic Review. Biomedicines 2022; 10:biomedicines10123026. [PMID: 36551782 PMCID: PMC9775565 DOI: 10.3390/biomedicines10123026] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
The prodromal phase of Parkinson's disease (PD) is characterised by many non-motor symptoms, and these have recently been posited to be predictive of later diagnosis. Genetic rodent models can develop non-motor phenotypes, providing tools to identify mechanisms underlying the early development of PD. However, it is not yet clear how reproducible non-motor phenotypes are amongst genetic PD rodent models, whether phenotypes are age-dependent, and the translatability of these phenotypes has yet to be explored. A systematic literature search was conducted on studies using genetic PD rodent models to investigate non-motor phenotypes; cognition, anxiety/depressive-like behaviour, gastrointestinal (GI) function, olfaction, circadian rhythm, cardiovascular and urinary function. In total, 51 genetic models of PD across 150 studies were identified. We found outcomes of most phenotypes were inconclusive due to inadequate studies, assessment at different ages, or variation in experimental and environmental factors. GI dysfunction was the most reproducible phenotype across all genetic rodent models. The mouse model harbouring mutant A53T, and the wild-type hα-syn overexpression (OE) model recapitulated the majority of phenotypes, albeit did not reliably produce concurrent motor deficits and nigral cell loss. Furthermore, animal models displayed different phenotypic profiles, reflecting the distinct genetic risk factors and heterogeneity of disease mechanisms. Currently, the inconsistent phenotypes within rodent models pose a challenge in the translatability and usefulness for further biomechanistic investigations. This review highlights opportunities to improve phenotype reproducibility with an emphasis on phenotypic assay choice and robust experimental design.
Collapse
|
28
|
Interactions of dopamine, iron, and alpha-synuclein linked to dopaminergic neuron vulnerability in Parkinson's disease and neurodegeneration with brain iron accumulation disorders. Neurobiol Dis 2022; 175:105920. [DOI: 10.1016/j.nbd.2022.105920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 10/21/2022] [Accepted: 11/04/2022] [Indexed: 11/08/2022] Open
|
29
|
Erritzoe D, Godlewska BR, Rizzo G, Searle GE, Agnorelli C, Lewis Y, Ashok AH, Colasanti A, Boura I, Farrell C, Parfitt H, Howes O, Passchier J, Gunn RN, Politis M, Nutt DJ, Cowen PJ, Knudsen GM, Rabiner EA. Brain Serotonin Release Is Reduced in Patients With Depression: A [ 11C]Cimbi-36 Positron Emission Tomography Study With a d-Amphetamine Challenge. Biol Psychiatry 2022:S0006-3223(22)01704-8. [PMID: 36635177 DOI: 10.1016/j.biopsych.2022.10.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/03/2022] [Accepted: 10/21/2022] [Indexed: 01/14/2023]
Abstract
BACKGROUND The serotonin hypothesis of depression proposes that diminished serotonergic (5-HT) neurotransmission is causal in the pathophysiology of the disorder. Although the hypothesis is over 50 years old, there is no firm in vivo evidence for diminished 5-HT neurotransmission. We recently demonstrated that the 5-HT2A receptor agonist positron emission tomography (PET) radioligand [11C]Cimbi-36 is sensitive to increases in extracellular 5-HT induced by an acute d-amphetamine challenge. Here we applied [11C]Cimbi-36 PET to compare brain 5-HT release capacity in patients experiencing a major depressive episode (MDE) to that of healthy control subjects (HCs) without depression. METHODS Seventeen antidepressant-free patients with MDE (3 female/14 male, mean age 44 ± 13 years, Hamilton Depression Rating Scale score 21 ± 4 [range 16-30]) and 20 HCs (3 female/17 male, mean age 32 ± 9 years) underwent 90-minute dynamic [11C]Cimbi-36 PET before and 3 hours after a 0.5-mg/kg oral dose of d-amphetamine. Frontal cortex (main region of interest) 5-HT2A receptor nondisplaceable binding was calculated from kinetic analysis using the multilinear analysis-1 approach with the cerebellum as the reference region. RESULTS Following d-amphetamine administration, frontal nondisplaceable binding potential (BPND) was significantly reduced in the HC group (1.04 ± 0.31 vs. 0.87 ± 0.24, p < .001) but not in the MDE group (0.97 ± 0.25 vs. 0.92 ± 0.22, not significant). ΔBPND of the MDE group was significantly lower than that of the HC group (HC: 15% ± 14% vs. MDE: 6.5% ± 20%, p = .041). CONCLUSIONS This first direct assessment of 5-HT release capacity in people with depression provides clear evidence for dysfunctional serotonergic neurotransmission in depression by demonstrating reduced 5-HT release capacity in patients experiencing an MDE.
Collapse
Affiliation(s)
- David Erritzoe
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom.
| | - Beata R Godlewska
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | | | | | - Claudio Agnorelli
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom; Department of Molecular Medicine, University of Siena, Siena, Italy
| | | | - Abhishekh H Ashok
- Department of Psychosis Studies, King's College London, London, United Kingdom; Department of Radiology, University of Cambridge & Addenbrooke's Hospital, Cambridge, United Kingdom
| | | | - Iro Boura
- Parkinson Foundation Centre of Excellence, King's College London, London, United Kingdom
| | - Chloe Farrell
- Parkinson Foundation Centre of Excellence, King's College London, London, United Kingdom
| | - Hollie Parfitt
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Oliver Howes
- Department of Psychosis Studies, King's College London, London, United Kingdom
| | | | | | - Marios Politis
- Neurodegeneration Imaging Group, University of Exeter, Exeter, United Kingdom
| | - David J Nutt
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Philip J Cowen
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Gitte M Knudsen
- Neurobiology Research Unit, University Hospital Rigshospitalet and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Eugenii A Rabiner
- Invicro, London, United Kingdom; Department of Neuroimaging, King's College London, London, United Kingdom
| |
Collapse
|
30
|
Combining CRISPR-Cas9 and brain imaging to study the link from genes to molecules to networks. Proc Natl Acad Sci U S A 2022; 119:e2122552119. [PMID: 36161926 DOI: 10.1073/pnas.2122552119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Receptors, transporters, and ion channels are important targets for therapy development in neurological diseases, but their mechanistic role in pathogenesis is often poorly understood. Gene editing and in vivo imaging approaches will help to identify the molecular and functional role of these targets and the consequence of their regional dysfunction on the whole-brain level. We combine CRISPR-Cas9 gene editing with in vivo positron emission tomography (PET) and functional MRI (fMRI) to investigate the direct link between genes, molecules, and the brain connectome. The extensive knowledge of the Slc18a2 gene encoding the vesicular monoamine transporter (VMAT2), involved in the storage and release of dopamine, makes it an excellent target for studying the gene network relationships while structurally preserving neuronal integrity and function. We edited the Slc18a2 in the substantia nigra pars compacta of adult rats and used in vivo molecular imaging besides behavioral, histological, and biochemical assessments to characterize the CRISPR-Cas9-mediated VMAT2 knockdown. Simultaneous PET/fMRI was performed to investigate molecular and functional brain alterations. We found that stage-specific adaptations of brain functional connectivity follow the selective impairment of presynaptic dopamine storage and release. Our study reveals that recruiting different brain networks is an early response to the dopaminergic dysfunction preceding neuronal cell loss. Our combinatorial approach is a tool to investigate the impact of specific genes on brain molecular and functional dynamics, which will help to develop tailored therapies for normalizing brain function.
Collapse
|
31
|
Ahnaou A, Whim D. REM sleep behavior and olfactory dysfunction: improving the utility and translation of animal models in the search for neuroprotective therapies for Parkinson's disease. Neurosci Biobehav Rev 2022; 143:104897. [PMID: 36183864 DOI: 10.1016/j.neubiorev.2022.104897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 11/25/2022]
Abstract
Parkinson's disease (PD) is a heterogeneous neurodegenerative disease that belongs to the family of synucleiopathies, varying in age, symptoms and progression. Hallmark of the disease is the accumulation of misfolded α-synuclein protein (α-Syn) in neuronal and non-neuronal brain cells. In past decades, diagnosis and treatment of PD has focused on motor deficits, which for the clinical endpoint, have contributed to the prevalence of deficits in the nigrostriatal dopaminergic system and animal models related to motor behavior to study disease. However, clinical trials have failed to translate results from animal models into effective treatments. PD as a multisystem disorder therefore requires additional assessment of motor and non-motor symptoms. Braak's staging revealed early α-Syn pathology in pontine brainstem and olfactory circuits controlling rapid eye movement sleep behavior disorder (RBD) and olfaction, respectively. Recent converging evidence from multicenter clinical studies supports that RBD is the most important risk factor for prodromal PD and the conduct of neuroprotective therapeutic trials in RBD-enriched cohorts has been recommended. Animal models of RBD and olfaction dysfunction can aid to fill the gap in translational research.
Collapse
Affiliation(s)
- A Ahnaou
- Department of Neuroscience, Janssen Research & Development, a Division of Janssen Pharmaceutica NV. Turnhoutseweg 30, B-2340 Beerse, Belgium.
| | - Drinkenburg Whim
- Department of Neuroscience, Janssen Research & Development, a Division of Janssen Pharmaceutica NV. Turnhoutseweg 30, B-2340 Beerse, Belgium
| |
Collapse
|
32
|
Castro AE, Domínguez-Ordoñez R, Young LJ, Camacho FJ, Ávila-González D, Paredes RG, Díaz NF, Portillo W. Pair-bonding and social experience modulate new neurons survival in adult male and female prairie voles ( Microtus ochrogaster). Front Neuroanat 2022; 16:987229. [PMID: 36189119 PMCID: PMC9520527 DOI: 10.3389/fnana.2022.987229] [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: 07/05/2022] [Accepted: 08/16/2022] [Indexed: 12/04/2022] Open
Abstract
Prairie voles are a socially monogamous species that, after cohabitation with mating, form enduring pair bonds. The plastic mechanisms involved in this social behavior are not well-understood. Neurogenesis in adult rodents is a plastic neural process induced in specific brain areas like the olfactory bulbs (OB) and dentate gyrus (DG) of the hippocampus. However, it is unknown how cell survival is modulated by social or sexual experience in prairie voles. This study aimed to evaluate if cohabitation with mating and/or social exposure to a vole of the opposite sex increased the survival of the new cells in the main and accessory OB and DG. To identify the new cells and evaluate their survival, voles were injected with the DNA synthesis marker 5-bromo-2'-deoxyuridine (BrdU) and were randomly distributed into one of the following groups: (A) Control (C), voles that did not receive any sexual stimulation and were placed alone during the behavioral test. (B) Social exposure (SE), voles were individually placed in a cage equally divided into two compartments by an acrylic screen with small holes. One male and one female were placed in opposite compartments. (C) Social cohabitation with mating (SCM), animals mated freely. Our findings demonstrated that SCM females had increases in the number of new cells (BrdU-positive cells) in the main olfactory bulb and new mature neurons (BrdU/NeuN-positive cells) in the glomerular layer (GlL). In contrast, these new cells decrease in males in the SE and SCM conditions. In the granular cell layer (GrL), SCM females had more new cells and neurons than the SE group. In the accessory olfactory bulb, in the anterior GlL, SCM decreased the number of new cells and neurons in females. On the other hand, in the DG, SCM and SE increase the number of new cells in the suprapyramidal blade in female voles. Males from SCM express more new cells and neurons in the infrapyramidal blade compared with SE group. Comparison between male and females showed that new cells/neurons survival was sex dependent. These results suggest that social interaction and sexual behavior modulate cell survival and influence the neuronal fate in a sex-dependent manner, in the OB and DG. This study will contribute to understand neural mechanisms of complex social and pair bond behaviors in the prairie voles; supporting adult neurogenesis as a plastic mechanism potentially involved in social monogamous strategy.
Collapse
Affiliation(s)
- Analía E. Castro
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| | - Raymundo Domínguez-Ordoñez
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
- Benemérita Universidad Autónoma de Puebla, Complejo Regional Centro, Puebla, Mexico
| | - Larry J. Young
- Silvio O. Conte Center for Oxytocin and Social Cognition, Center for Translational Social Neuroscience, Department of Psychiatry and Behavioral Sciences, Emory National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Francisco J. Camacho
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| | - Daniela Ávila-González
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| | - Raúl G. Paredes
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Nestor F. Díaz
- Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - Wendy Portillo
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| |
Collapse
|
33
|
Mendonça IP, de Paiva IHR, Duarte-Silva EP, de Melo MG, da Silva RS, do Nascimento MIX, Peixoto CA. Metformin improves depressive-like behavior in experimental Parkinson's disease by inducing autophagy in the substantia nigra and hippocampus. Inflammopharmacology 2022; 30:1705-1716. [PMID: 35931897 DOI: 10.1007/s10787-022-01043-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/16/2022] [Indexed: 11/05/2022]
Abstract
Parkinson's disease (PD) remains a disease of little known etiology. In addition to the motor symptoms, depression is present in about 40% of patients, contributing to the loss of quality of life. Recently, the involvement of the autophagy mechanism in the pathogenesis of depression has been studied, in addition to its involvement in PD as well. In this study, we tested the effects of metformin, an antidiabetic drug also with antidepressant effects, on depressive-like behavior in a rotenone-induced PD model and on the autophagy process. Mice 8-week-old male C57BL/6 were induced with rotenone for 20 consecutive days (2.5 mg/kg/day) and treated with metformin (200 mg/kg/day) from the 5th day of induction. All the animals were submitted to rotarod, sucrose preference and tail suspension tests. After euthanasia, the substantia nigra and hippocampus were removed for analysis by western blotting or fixed and analyzed by immunofluorescence. The results show that there was an impairment of autophagy in animals induced by rotenone both in nigral and extranigral regions as well as a depressive-like behavior. Metformin was able to inhibit depressive-like behavior and increase signaling pathway proteins, transcription factors and autophagosome-forming proteins, thus inducing autophagy in both the hippocampus and the substantia nigra. In conclusion, we show that metformin has an antidepressant effect in a rotenone-induced PD model, which may result, at least in part, from the induction of the autophagy process.
Collapse
Affiliation(s)
- Ingrid Prata Mendonça
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), Recife, PE, Brazil. .,Postgraduate Program in Biological Sciences (PPGCB), Federal University of Pernambuco (UFPE), Recife, Brazil.
| | - Igor Henrique Rodrigues de Paiva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), Recife, PE, Brazil.,Postgraduate Program in Biological Sciences (PPGCB), Federal University of Pernambuco (UFPE), Recife, Brazil
| | - Eduardo Pereira Duarte-Silva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), Recife, PE, Brazil.,Postgraduate Program in Biosciences and Biotechnology for Health (PPGBBS), Oswaldo Cruz Foundation (FIOCRUZ-PE)/Aggeu Magalhães Institute (IAM), Recife, PE, Brazil
| | - Michel Gomes de Melo
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), Recife, PE, Brazil.,Postgraduate Program in Biological Sciences (PPGCB), Federal University of Pernambuco (UFPE), Recife, Brazil
| | - Rodrigo S da Silva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), Recife, PE, Brazil.,Postgraduate Program in Biological Sciences (PPGCB), Federal University of Pernambuco (UFPE), Recife, Brazil
| | | | - Christina Alves Peixoto
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), Recife, PE, Brazil. .,National Institute of Science and Technology On Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|
34
|
Goldstein DS, Pekker MJ, Sullivan P, Isonaka R, Sharabi Y. Modeling the Progression of Cardiac Catecholamine Deficiency in Lewy Body Diseases. J Am Heart Assoc 2022; 11:e024411. [PMID: 35621196 PMCID: PMC9238705 DOI: 10.1161/jaha.121.024411] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/15/2022] [Indexed: 11/16/2022]
Abstract
Background Lewy body diseases (LBDs) feature deficiency of the sympathetic neurotransmitter norepinephrine in the left ventricular myocardium and sympathetic intra-neuronal deposition of the protein alpha-synuclein (αS). LBDs therefore are autonomic synucleinopathies. Computational modeling has revealed multiple functional abnormalities in residual myocardial sympathetic noradrenergic nerves in LBDs, including decreased norepinephrine synthesis, vesicular storage, and recycling. We report an extended model that enables predictions about the progression of LBDs and effects of genetic predispositions and treatments on that progression. Methods and Results The model combines cardiac sympathetic activation with autotoxicity mediated by the dopamine metabolite 3,4-dihydroxyphenylacetaldehyde. We tested the model by its ability to predict longitudinal empirical data based on cardiac sympathetic neuroimaging, effects of genetic variations related to particular intra-neuronal reactions, treatment by monoamine oxidase inhibition to decrease 3,4-dihydroxyphenylacetaldehyde production, and post-mortem myocardial tissue contents of catecholamines and αS. The new model generated a triphasic decline in myocardial norepinephrine content. This pattern was confirmed by empirical data from serial cardiac 18F-dopamine positron emission tomographic scanning in patients with LBDs. The model also correctly predicted empirical data about effects of genetic variants and monoamine oxidase inhibition and about myocardial levels of catecholamines and αS. Conclusions The present computational model predicts a triphasic decline in myocardial norepinephrine content as LBDs progress. According to the model, disease-modifying interventions begun at the transition from the first to the second phase delay the onset of symptomatic disease. Computational modeling coupled with biomarkers of preclinical autonomic synucleinopathy may enable early detection and more effective treatment of LBDs.
Collapse
Affiliation(s)
- David S. Goldstein
- Autonomic Medicine SectionClinical Neurosciences ProgramDivision of Intramural ResearchNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMD
| | - Mark J. Pekker
- Mathematical SciencesUniversity of Alabama at HuntsvilleHuntsvilleAL
| | - Patti Sullivan
- Autonomic Medicine SectionClinical Neurosciences ProgramDivision of Intramural ResearchNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMD
| | - Risa Isonaka
- Autonomic Medicine SectionClinical Neurosciences ProgramDivision of Intramural ResearchNational Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMD
| | - Yehonatan Sharabi
- Tel Aviv University Sackler Faculty of Medicine and Chaim Sheba Medical CenterTel HaShomerIsrael
| |
Collapse
|
35
|
Rajendran R, Ragavan RP, Al-Sehemi AG, Uddin MS, Aleya L, Mathew B. Current understandings and perspectives of petroleum hydrocarbons in Alzheimer's disease and Parkinson's disease: a global concern. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:10928-10949. [PMID: 35000177 DOI: 10.1007/s11356-021-17931-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 11/30/2021] [Indexed: 06/14/2023]
Abstract
Over the last few decades, the global prevalence of neurodevelopmental and neurodegenerative illnesses has risen rapidly. Although the aetiology remains unclear, evidence is mounting that exposure to persistent hydrocarbon pollutants is a substantial risk factor, predisposing a person to neurological diseases later in life. Epidemiological studies correlate environmental hydrocarbon exposure to brain disorders including neuropathies, cognitive, motor and sensory impairments; neurodevelopmental disorders like autism spectrum disorder (ASD); and neurodegenerative disorders like Alzheimer's disease (AD) and Parkinson's disease (PD). Particulate matter, benzene, toluene, ethylbenzene, xylenes, polycyclic aromatic hydrocarbons and endocrine-disrupting chemicals have all been linked to neurodevelopmental problems in all class of people. There is mounting evidence that supports the prevalence of petroleum hydrocarbon becoming neurotoxic and being involved in the pathogenesis of AD and PD. More study is needed to fully comprehend the scope of these problems in the context of unconventional oil and natural gas. This review summarises in vitro, animal and epidemiological research on the genesis of neurodegenerative disorders, highlighting evidence that supports inexorable role of hazardous hydrocarbon exposure in the pathophysiology of AD and PD. In this review, we offer a summary of the existing evidence gathered through a Medline literature search of systematic reviews and meta-analyses of the most important epidemiological studies published so far.
Collapse
Affiliation(s)
- Rajalakshmi Rajendran
- Department of Pharmacy Practice, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Roshni Pushpa Ragavan
- Research Center for Advanced Materials Science, King Khalid University, Abha, 61413, Saudi Arabia.
| | - Abdullah G Al-Sehemi
- Research Center for Advanced Materials Science, King Khalid University, Abha, 61413, Saudi Arabia
- Department of Chemistry, King Khalid University, Abha, 61413, Saudi Arabia
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Lotfi Aleya
- Laboratoire Chrono-Environment, CNRS6249, Universite de Bourgogne Franche-Comte, Besancon, France
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, 682 041, India.
| |
Collapse
|
36
|
Hunt J, Coulson EJ, Rajnarayanan R, Oster H, Videnovic A, Rawashdeh O. Sleep and circadian rhythms in Parkinson's disease and preclinical models. Mol Neurodegener 2022; 17:2. [PMID: 35000606 PMCID: PMC8744293 DOI: 10.1186/s13024-021-00504-w] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [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: 11/30/2021] [Indexed: 12/21/2022] Open
Abstract
The use of animals as models of human physiology is, and has been for many years, an indispensable tool for understanding the mechanisms of human disease. In Parkinson's disease, various mouse models form the cornerstone of these investigations. Early models were developed to reflect the traditional histological features and motor symptoms of Parkinson's disease. However, it is important that models accurately encompass important facets of the disease to allow for comprehensive mechanistic understanding and translational significance. Circadian rhythm and sleep issues are tightly correlated to Parkinson's disease, and often arise prior to the presentation of typical motor deficits. It is essential that models used to understand Parkinson's disease reflect these dysfunctions in circadian rhythms and sleep, both to facilitate investigations into mechanistic interplay between sleep and disease, and to assist in the development of circadian rhythm-facing therapeutic treatments. This review describes the extent to which various genetically- and neurotoxically-induced murine models of Parkinson's reflect the sleep and circadian abnormalities of Parkinson's disease observed in the clinic.
Collapse
Affiliation(s)
- Jeremy Hunt
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Elizabeth J. Coulson
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | | | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | - Aleksandar Videnovic
- Movement Disorders Unit and Division of Sleep Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Oliver Rawashdeh
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Australia
| |
Collapse
|
37
|
Khan AH, Lee LK, Smith DJ. Single-cell analysis of gene expression in the substantia nigra pars compacta of a pesticide-induced mouse model of Parkinson's disease. Transl Neurosci 2022; 13:255-269. [PMID: 36117858 PMCID: PMC9438968 DOI: 10.1515/tnsci-2022-0237] [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: 05/26/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 12/02/2022] Open
Abstract
Exposure to pesticides in humans increases the risk of Parkinson’s disease (PD), but the mechanisms remain poorly understood. To elucidate these pathways, we dosed C57BL/6J mice with a combination of the pesticides maneb and paraquat. Behavioral analysis revealed motor deficits consistent with PD. Single-cell RNA sequencing of substantia nigra pars compacta revealed both cell-type-specific genes and genes expressed differentially between pesticide and control, including Fam241b, Emx2os, Bivm, Gm1439, Prdm15, and Rai2. Neurons had the largest number of significant differentially expressed genes, but comparable numbers were found in astrocytes and less so in oligodendrocytes. In addition, network analysis revealed enrichment in functions related to the extracellular matrix. These findings emphasize the importance of support cells in pesticide-induced PD and refocus our attention away from neurons as the sole agent of this disorder.
Collapse
Affiliation(s)
- Arshad H. Khan
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Box 951735, 23-151 A CHS, Los Angeles, CA 90095-1735, United States of America
| | - Lydia K. Lee
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095-6928, United States of America
| | - Desmond J. Smith
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Box 951735, 23-151 A CHS, Los Angeles, CA 90095-1735, United States of America
| |
Collapse
|
38
|
Capsoni S, Fogli Iseppe A, Casciano F, Pignatelli A. Unraveling the Role of Dopaminergic and Calretinin Interneurons in the Olfactory Bulb. Front Neural Circuits 2021; 15:718221. [PMID: 34690707 PMCID: PMC8531203 DOI: 10.3389/fncir.2021.718221] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/31/2021] [Indexed: 11/27/2022] Open
Abstract
The perception and discriminating of odors are sensory activities that are an integral part of our daily life. The first brain region where odors are processed is the olfactory bulb (OB). Among the different cell populations that make up this brain area, interneurons play an essential role in this sensory activity. Moreover, probably because of their activity, they represent an exception compared to other parts of the brain, since OB interneurons are continuously generated in the postnatal and adult period. In this review, we will focus on periglomerular (PG) cells which are a class of interneurons found in the glomerular layer of the OB. These interneurons can be classified into distinct subtypes based on their neurochemical nature, based on the neurotransmitter and calcium-binding proteins expressed by these cells. Dopaminergic (DA) periglomerular cells and calretinin (CR) cells are among the newly generated interneurons and play an important role in the physiology of OB. In the OB, DA cells are involved in the processing of odors and the adaptation of the bulbar network to external conditions. The main role of DA cells in OB appears to be the inhibition of glutamate release from olfactory sensory fibers. Calretinin cells are probably the best morphologically characterized interneurons among PG cells in OB, but little is known about their function except for their inhibitory effect on noisy random excitatory signals arriving at the main neurons. In this review, we will mainly describe the electrophysiological properties related to the excitability profiles of DA and CR cells, with a particular view on the differences that characterize DA mature interneurons from cells in different stages of adult neurogenesis.
Collapse
Affiliation(s)
- Simona Capsoni
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
| | - Alex Fogli Iseppe
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Fabio Casciano
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
- Interdepartmental Research Centre for the Study of Multiple Sclerosis and Inflammatory and Degenerative Diseases of the Nervous System, University of Ferrara, Ferrara, Italy
| | - Angela Pignatelli
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| |
Collapse
|
39
|
Langley MR, Ghaisas S, Palanisamy BN, Ay M, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Characterization of nonmotor behavioral impairments and their neurochemical mechanisms in the MitoPark mouse model of progressive neurodegeneration in Parkinson's disease. Exp Neurol 2021; 341:113716. [PMID: 33839143 PMCID: PMC9797183 DOI: 10.1016/j.expneurol.2021.113716] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/24/2021] [Accepted: 04/05/2021] [Indexed: 12/30/2022]
Abstract
Mitochondrial dysfunction has been implicated as a key player in the pathogenesis of Parkinson's disease (PD). The MitoPark mouse, a transgenic mitochondrial impairment model developed by specific inactivation of TFAM in dopaminergic neurons, spontaneously exhibits progressive motor deficits and neurodegeneration, recapitulating several features of PD. Since nonmotor symptoms are now recognized as important features of the prodromal stage of PD, we comprehensively assessed the clinically relevant motor and nonmotor deficiencies from ages 8-24 wk in both male and female MitoPark mice and their littermate controls. As expected, motor deficits in MitoPark mice began around 12-14 wk and became severe by 16-24 wk. Interestingly, MitoPark mice exhibited olfactory deficits in the novel and social scent tests as early as 10-12 wk as compared to age-matched littermate controls. Additionally, male MitoPark mice showed spatial memory deficits before female mice, beginning at 8 wk and becoming most severe at 16 wk, as determined by the Morris water maze. MitoPark mice between 16 and 24 wk spent more time immobile in forced swim and tail suspension tests, and made fewer entries into open arms of the elevated plus maze, indicating a depressive and anxiety-like phenotype, respectively. Importantly, depressive behavior as determined by immobility in forced swim test was reversible by antidepressant treatment with desipramine. Neurochemical and mechanistic studies revealed significant changes in CREB phosphorylation, BDNF, and catecholamine levels as well as neurogenesis in key brain regions. Collectively, our results indicate that MitoPark mice progressively exhibit deficits in olfactory discrimination, cognitive learning and memory, and anxiety- and depression-like behaviors as well as key neurochemical signaling associated with nonmotor deficits in PD. Thus, MitoPark mice can serve as an invaluable model for studying nonmotor deficits in addition to studying the motor deficits related to pathology in PD.
Collapse
Affiliation(s)
- Monica R Langley
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Shivani Ghaisas
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Bharathi N Palanisamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Muhammet Ay
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Huajun Jin
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Vellareddy Anantharam
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Arthi Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America
| | - Anumantha G Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States of America.
| |
Collapse
|
40
|
Neuropsychiatric and Cognitive Deficits in Parkinson's Disease and Their Modeling in Rodents. Biomedicines 2021; 9:biomedicines9060684. [PMID: 34204380 PMCID: PMC8234051 DOI: 10.3390/biomedicines9060684] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 11/29/2022] Open
Abstract
Parkinson’s disease (PD) is associated with a large burden of non-motor symptoms including olfactory and autonomic dysfunction, as well as neuropsychiatric (depression, anxiety, apathy) and cognitive disorders (executive dysfunctions, memory and learning impairments). Some of these non-motor symptoms may precede the onset of motor symptoms by several years, and they significantly worsen during the course of the disease. The lack of systematic improvement of these non-motor features by dopamine replacement therapy underlines their multifactorial origin, with an involvement of monoaminergic and cholinergic systems, as well as alpha-synuclein pathology in frontal and limbic cortical circuits. Here we describe mood and neuropsychiatric disorders in PD and review their occurrence in rodent models of PD. Altogether, toxin-based rodent models of PD indicate a significant but non-exclusive contribution of mesencephalic dopaminergic loss in anxiety, apathy, and depressive-like behaviors, as well as in learning and memory deficits. Gene-based models display significant deficits in learning and memory, as well as executive functions, highlighting the contribution of alpha-synuclein pathology to these non-motor deficits. Collectively, neuropsychiatric and cognitive deficits are recapitulated to some extent in rodent models, providing partial but nevertheless useful options to understand the pathophysiology of non-motor symptoms and develop therapeutic options for these debilitating symptoms of PD.
Collapse
|
41
|
Vecchio LM, Sullivan P, Dunn AR, Bermejo MK, Fu R, Masoud ST, Gregersen E, Urs NM, Nazari R, Jensen PH, Ramsey A, Goldstein DS, Miller GW, Salahpour A. Enhanced tyrosine hydroxylase activity induces oxidative stress, causes accumulation of autotoxic catecholamine metabolites, and augments amphetamine effects in vivo. J Neurochem 2021; 158:960-979. [PMID: 33991113 PMCID: PMC8376767 DOI: 10.1111/jnc.15432] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/10/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022]
Abstract
In Parkinson's disease, dopamine‐containing nigrostriatal neurons undergo profound degeneration. Tyrosine hydroxylase (TH) is the rate‐limiting enzyme in dopamine biosynthesis. TH increases in vitro formation of reactive oxygen species, and previous animal studies have reported links between cytosolic dopamine build‐up and oxidative stress. To examine effects of increased TH activity in catecholaminergic neurons in vivo, we generated TH‐over‐expressing mice (TH‐HI) using a BAC‐transgenic approach that results in over‐expression of TH with endogenous patterns of expression. The transgenic mice were characterized by western blot, qPCR, and immunohistochemistry. Tissue contents of dopamine, its metabolites, and markers of oxidative stress were evaluated. TH‐HI mice had a 3‐fold increase in total and phosphorylated TH levels and an increased rate of dopamine synthesis. Coincident with elevated dopamine turnover, TH‐HI mice showed increased striatal production of H2O2 and reduced glutathione levels. In addition, TH‐HI mice had elevated striatal levels of the neurotoxic dopamine metabolites 3,4‐dihydroxyphenylacetaldehyde and 5‐S‐cysteinyl‐dopamine and were more susceptible than wild‐type mice to the effects of amphetamine and methamphetamine. These results demonstrate that increased TH alone is sufficient to produce oxidative stress in vivo, build up autotoxic dopamine metabolites, and augment toxicity.
Collapse
Affiliation(s)
- Laura M Vecchio
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Patricia Sullivan
- Autonomic Medicine Section, Clinical Neurosciences Program, Division of Intramural Research, National Institute of Neurological, Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Amy R Dunn
- The Jackson Laboratory. Bar Harbor, Maine, USA
| | - Marie Kristel Bermejo
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Rong Fu
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | - Shababa T Masoud
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Emil Gregersen
- Danish Research Institute of Translational Neuroscience - DANDRITE, Department of Biomedicine, Faculty of Health, Aarhus University, Aarhus C., Denmark
| | - Nikhil M Urs
- Department of Pharmacology and Therapeutics, University of Florida, Gainsville, FL, USA
| | - Reza Nazari
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Poul Henning Jensen
- Danish Research Institute of Translational Neuroscience - DANDRITE, Department of Biomedicine, Faculty of Health, Aarhus University, Aarhus C., Denmark
| | - Amy Ramsey
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - David S Goldstein
- Autonomic Medicine Section, Clinical Neurosciences Program, Division of Intramural Research, National Institute of Neurological, Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University Medical Centre, New York, NY, USA
| | - Ali Salahpour
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
42
|
Cui K, Yang F, Tufan T, Raza MU, Zhan Y, Fan Y, Zeng F, Brown RW, Price JB, Jones TC, Miller GW, Zhu MY. Restoration of Noradrenergic Function in Parkinson's Disease Model Mice. ASN Neuro 2021; 13:17590914211009730. [PMID: 33940943 PMCID: PMC8114769 DOI: 10.1177/17590914211009730] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dysfunction of the central noradrenergic and dopaminergic systems is the primary neurobiological characteristic of Parkinson’s disease (PD). Importantly, neuronal loss in the locus coeruleus (LC) that occurs in early stages of PD may accelerate progressive loss of dopaminergic neurons. Therefore, restoring the activity and function of the deficient noradrenergic system may be an important therapeutic strategy for early PD. In the present study, the lentiviral constructions of transcription factors Phox2a/2b, Hand2 and Gata3, either alone or in combination, were microinjected into the LC region of the PD model VMAT2 Lo mice at 12 and 18 month age. Biochemical analysis showed that microinjection of lentiviral expression cassettes into the LC significantly increased mRNA levels of Phox2a, and Phox2b, which were accompanied by parallel increases of mRNA and proteins of dopamine β-hydroxylase (DBH) and tyrosine hydroxylase (TH) in the LC. Furthermore, there was considerable enhancement of DBH protein levels in the frontal cortex and hippocampus, as well as enhanced TH protein levels in the striatum and substantia nigra. Moreover, these manipulations profoundly increased norepinephrine and dopamine concentrations in the striatum, which was followed by a remarkable improvement of the spatial memory and locomotor behavior. These results reveal that over-expression of these transcription factors in the LC improves noradrenergic and dopaminergic activities and functions in this rodent model of PD. It provides the necessary groundwork for the development of gene therapies of PD, and expands our understanding of the link between the LC-norepinephrine and dopamine systems during the progression of PD.
Collapse
Affiliation(s)
- Kui Cui
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| | - Fan Yang
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States.,Hong Kong Institute, Asia Metropolitan University, Hong Kong, China
| | - Turan Tufan
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| | - Muhammad U Raza
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| | - Yanqiang Zhan
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States.,Department of Neurology, Renmin Hospital of the Wuhan University, Wuhan, China
| | - Yan Fan
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States.,Department of Biochemistry, Nantong University College of Medicine, Nantong, China
| | - Fei Zeng
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States.,Department of Neurology, Renmin Hospital of the Wuhan University, Wuhan, China
| | - Russell W Brown
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| | - Jennifer B Price
- Department of Biological Sciences, College of Arts and Sciences; East Tennessee State University, Johnson City, United States
| | - Thomas C Jones
- Department of Biological Sciences, College of Arts and Sciences; East Tennessee State University, Johnson City, United States
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailmen School of Public Health, Columbia University, New York, New York, United States
| | - Meng-Yang Zhu
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| |
Collapse
|
43
|
Bradner JM, Kalia V, Lau FK, Sharma M, Bucher ML, Johnson M, Chen M, Walker DI, Jones DP, Miller GW. Genetic or Toxicant-Induced Disruption of Vesicular Monoamine Storage and Global Metabolic Profiling in Caenorhabditis elegans. Toxicol Sci 2021; 180:313-324. [PMID: 33538833 PMCID: PMC8041460 DOI: 10.1093/toxsci/kfab011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The proper storage and release of monoamines contributes to a wide range of neuronal activity. Here, we examine the effects of altered vesicular monoamine transport in the nematode Caenorhabditis elegans. The gene cat-1 is responsible for the encoding of the vesicular monoamine transporter (VMAT) in C. elegans and is analogous to the mammalian vesicular monoamine transporter 2 (VMAT2). Our laboratory has previously shown that reduced VMAT2 activity confers vulnerability on catecholamine neurons in mice. The purpose of this article was to determine whether this function is conserved and to determine the impact of reduced VMAT activity in C. elegans. Here we show that deletion of cat-1/VMAT increases sensitivity to the neurotoxicant 1-methyl-4-phenylpyridinium (MPP+) as measured by enhanced degeneration of dopamine neurons. Reduced cat-1/VMAT also induces changes in dopamine-mediated behaviors. High-resolution mass spectrometry-based metabolomics in the whole organism reveals changes in amino acid metabolism, including tyrosine metabolism in the cat-1/VMAT mutants. Treatment with MPP+ disrupted tryptophan metabolism. Both conditions altered glycerophospholipid metabolism, suggesting a convergent pathway of neuronal dysfunction. Our results demonstrate the evolutionarily conserved nature of monoamine function in C. elegans and further suggest that high-resolution mass spectrometry-based metabolomics can be used in this model to study environmental and genetic contributors to complex human disease.
Collapse
Affiliation(s)
- Joshua M Bradner
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| | - Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| | - Fion K Lau
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| | - Monica Sharma
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| | - Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| | - Michelle Johnson
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia 30322, USA
| | - Merry Chen
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia 30322, USA
| | - Douglas I Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Dean P Jones
- Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia 30303, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| |
Collapse
|
44
|
Criaud M, Kim JH, Zurowski M, Lobaugh N, Chavez S, Houle S, Strafella AP. Anxiety in Parkinson's disease: Abnormal resting activity and connectivity. Brain Res 2021; 1753:147235. [PMID: 33412150 DOI: 10.1016/j.brainres.2020.147235] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 11/19/2020] [Accepted: 12/07/2020] [Indexed: 11/25/2022]
Abstract
Anxiety is a very common yet poorly understood symptom of Parkinson's disease. We investigated whether Parkinson's disease patients experiencing anxiety share neural mechanisms described in the general population with involvement of critical regions for the control of behaviour and movement. Thirty-nine patients with PD were recruited for this study, 20 with higher anxiety scores and 19 with lower anxiety scores. They all underwent a resting-state fMRI scan, while they were on medication. The amplitude of low-frequency fluctuation (ALFF) and seed-based connectivity were investigated to reveal the changes of the spontaneous activity and the interaction among different related regions. The results provided evidence that anxiety in Parkinson's disease is associated with the over-activation of the amygdala and impaired inter-relationship of regions involved in behavior (i.e. medial prefrontal cortex, insula) and motor control (i.e. basal ganglia).
Collapse
Affiliation(s)
- Marion Criaud
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada; Division of Brain, Imaging and Behaviour - Systems Neuroscience, Krembil Research Institute, UHN, University of Toronto, Ontario, Canada.
| | - Jin-Hee Kim
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada; Division of Brain, Imaging and Behaviour - Systems Neuroscience, Krembil Research Institute, UHN, University of Toronto, Ontario, Canada
| | - Mateusz Zurowski
- Division of Brain, Imaging and Behaviour - Systems Neuroscience, Krembil Research Institute, UHN, University of Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Ontario, Canada
| | - Nancy Lobaugh
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sofia Chavez
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada
| | - Sylvain Houle
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada
| | - Antonio P Strafella
- Morton and Gloria Shulman Movement Disorder Unit & E.J. Safra Parkinson Disease Program, Toronto Western Hospital, UHN, University of Toronto, Ontario, Canada; Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada; Division of Brain, Imaging and Behaviour - Systems Neuroscience, Krembil Research Institute, UHN, University of Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
45
|
Reduction of falls in a rat model of PD falls by the M1 PAM TAK-071. Psychopharmacology (Berl) 2021; 238:1953-1964. [PMID: 33735392 PMCID: PMC7969347 DOI: 10.1007/s00213-021-05822-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 03/08/2021] [Indexed: 01/03/2023]
Abstract
RATIONALE In addition to the disease-defining motor symptoms, patients with Parkinson's disease (PD) exhibit gait dysfunction, postural instability, and a propensity for falls. These dopamine (DA) replacement-resistant symptoms in part have been attributed to loss of basal forebrain (BF) cholinergic neurons and, in interaction with striatal dopamine (DA) loss, to the resulting disruption of the attentional control of balance and complex movements. Rats with dual cholinergic-DA losses ("DL rats") were previously demonstrated to model PD falls and associated impairments of gait and balance. OBJECTIVES We previously found that the muscarinic M1-positive allosteric modulator (PAM) TAK-071 improved the attentional performance of rats with BF cholinergic losses. Here, we tested the hypotheses that TAK-071 reduces fall rates in DL rats. RESULTS Prior to DL surgery, female rats were trained to traverse a rotating straight rod as well as a rod with two zigzag segments. DL rats were refamiliarized with such traversals post-surgery and tested over 7 days on increasingly demanding testing conditions. TAK-071 (0.1, 0.3 mg/kg, p.o.) was administered prior to daily test sessions over this 7-day period. As before, DL rats fell more frequently than sham-operated control rats. Treatment of DL rats with TAK-071 reduced falls from the rotating rod and the rotating zigzag rod, specifically when the angled part of the zigzag segment, upon entering, was at a steep, near vertical angle. CONCLUSIONS TAK-071 may benefit complex movement control, specifically in situations which disrupt the patterning of forward movement and require the interplay between cognitive and motor functions to modify movement based on information about the state of dynamic surfaces, balance, and gait.
Collapse
|
46
|
Black CA, Bucher ML, Bradner JM, Jonas L, Igarza K, Miller GW. Assessing Vesicular Monoamine Transport and Toxicity Using Fluorescent False Neurotransmitters. Chem Res Toxicol 2020; 34:1256-1264. [PMID: 33378168 DOI: 10.1021/acs.chemrestox.0c00380] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Impairments in the vesicular packaging of dopamine result in an accumulation of dopamine in the cytosol. Cytosolic dopamine is vulnerable to two metabolic processes-enzymatic catabolism and enzymatic- or auto-oxidation-that form toxic metabolites and generate reactive oxygen species. Alterations in the expression or activity of the vesicular monoamine transporter 2 (VMAT2), which transports monoamines such as dopamine from the cytosol into the synaptic vesicle, result in dysregulated dopamine packaging. Here, we developed a series of assays using the fluorescent false neurotransmitter 206 (FFN206) to visualize VMAT2-mediated vesicular packaging at baseline and following pharmacological and toxicological manipulations. As a proof of principle, we observed a significant reduction in vesicular FFN206 packaging after treatment with the VMAT2 inhibitors reserpine (IC50: 73.1 nM), tetrabenazine (IC50: 30.4 nM), methamphetamine (IC50: 2.4 μM), and methylphenidate (IC50: 94.3 μM). We then applied the assay to investigate the consequences on vesicular packaging by environmental toxicants including the pesticides paraquat, rotenone, and chlorpyrifos, as well as the halogenated compounds unichlor, perfluorooctanesulfonic acid, Paroil, Aroclor 1260, and hexabromocyclododecane. Several of the environmental toxicants showed minor impairment of the vesicular FFN206 loading, suggesting that the toxicants are weak VMAT2 inhibitors at the concentrations tested. The assay presented here can be applied to investigate the effect of additional pharmacological compounds and environmental toxicants on vesicular function, which will provide insight into how exposures to such factors are involved in the pathogenesis of monoaminergic diseases such as Parkinson's disease, and the assay can be used to identify pharmacological agents that influence VMAT2 activity.
Collapse
Affiliation(s)
- Carlie A Black
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia 30322, United States
| | - Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Joshua M Bradner
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Lauren Jonas
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia 30322, United States
| | - Kenny Igarza
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia 30322, United States
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| |
Collapse
|
47
|
Moreira CG, Morawska MM, Baumann A, Masneuf S, Linnebank M, Sommerauer M, Landolt HP, Noain D, Baumann CR. Improved functional and histochemical outcomes in l-DOPA plus tolcapone treated VMAT2-deficient mice. Neuropharmacology 2020; 181:108353. [PMID: 33038358 DOI: 10.1016/j.neuropharm.2020.108353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/03/2020] [Accepted: 10/05/2020] [Indexed: 01/02/2023]
Abstract
Parkinson disease is typically treated with L-3,4-dihydroxyphenylalanine (or levodopa) co-prescribed with concentration stabilizers to prevent undesired motor fluctuations. However, the beneficial role of the chronic combined therapy on disease progression has not been thoroughly explored. We hypothesized that tolcapone, a catechol-O-methyl-transferase inhibitor, co-administered with levodopa may offer beneficial long-term disease-modifying effects through its dopamine stabilization actions. Here, we followed vesicular monoamine transporter 2-deficient and wild-type mice treated twice daily per os with vehicle, levodopa (20 mg/kg), tolcapone (15 mg/kg) or levodopa (12.5 mg/kg) + tolcapone (15 mg/kg) for 17 weeks. We assessed open field, bar test and rotarod performances at baseline and every 4th week thereafter, corresponding to OFF-medication weeks. Finally, we collected coronal sections from the frontal caudate-putamen and determined the reactivity level of dopamine transporter. Vesicular monoamine transporter 2-deficient mice responded positively to chronic levodopa + tolcapone intervention in the bar test during OFF-periods. Neither levodopa nor tolcapone interventions offered significant improvements on their own. Similarly, chronic levodopa + tolcapone intervention was associated with partially rescued dopamine transporter levels, whereas animals treated solely with levodopa or tolcapone did not present this effect. Interestingly, 4-month progression of bar test scores correlated significantly with dopamine-transporter-label density. Overall, we observed a moderate functional and histopathological improvement effect by chronic dopamine replacement when combined with tolcapone in vesicular monoamine transporter 2-deficient mice. Altogether, chronic stabilization of dopamine levels by catechol-O-methyl-transferase inhibition, besides its intended immediate actions, arises as a potential long-term beneficial approach during the progression of Parkinson disease.
Collapse
Affiliation(s)
- Carlos G Moreira
- Neurology Department, University Hospital Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
| | - Marta M Morawska
- Neurology Department, University Hospital Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
| | - Aron Baumann
- Neurology Department, University Hospital Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
| | - Sophie Masneuf
- Neurology Department, University Hospital Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
| | - Michael Linnebank
- Neurology Department, University Hospital Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
| | - Michael Sommerauer
- Neurology Department, University Hospital Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
| | - Hans-Peter Landolt
- Institute of Pharmacology and Toxicology, University of Zurich, Irchel Campus Y17, Winterthurerstrasse 190, 8057 Zurich, Switzerland; University Center of Competence Sleep & Health Zurich, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
| | - Daniela Noain
- Neurology Department, University Hospital Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland.
| | - Christian R Baumann
- Neurology Department, University Hospital Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland; University Center of Competence Sleep & Health Zurich, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland.
| |
Collapse
|
48
|
Bucher ML, Barrett CW, Moon CJ, Mortimer AD, Burton EA, Greenamyre JT, Hastings TG. Acquired dysregulation of dopamine homeostasis reproduces features of Parkinson's disease. NPJ PARKINSONS DISEASE 2020; 6:34. [PMID: 33298952 PMCID: PMC7666186 DOI: 10.1038/s41531-020-00134-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 10/08/2020] [Indexed: 12/24/2022]
Abstract
The catecholamine neurotransmitter dopamine has the potential to act as an endogenous neurotoxin when its vesicular sequestration is dysregulated. Despite postmortem analyses from patients with Parkinson’s disease that demonstrate decreased vesicular sequestration of dopamine with a corresponding increase in dopamine metabolism, dopamine’s contribution to nigrostriatal dopaminergic degeneration in Parkinson’s disease has been debated. Here, we present a new in vivo model demonstrating the induction of Parkinson’s disease-associated pathogenic mechanisms of degeneration resulting from acquired dysregulation of dopamine sequestration in nigrostriatal dopaminergic neurons in adult rats. Utilizing adeno-associated virus (serotype 2), viral-mediated small-hairpin RNA interference of endogenous vesicular monoamine transporter 2 (VMAT2) expression resulted in a loss of VMAT2 protein expression in transduced dopaminergic cell bodies in the substantia nigra with a corresponding loss of VMAT2 protein within the striatal terminals. The loss of VMAT2 resulted in an accumulation of cytosolic dopamine and subsequent increased dopamine metabolism, deficits in dopamine-mediated behaviors, and degeneration of nigrostriatal dopaminergic neurons that was rescued through reintroduction of exogenous VMAT2, demonstrating that the toxicity was specific to the loss of VMAT2. Analysis of parkinsonian pathogenic mechanisms of degeneration identified oxidative damage, activation of Parkinson’s disease-associated kinase LRRK2, and the formation of aberrant α-synuclein. This model demonstrates that a progressive acquired loss of VMAT2 expression in adulthood is sufficient to induce Parkinson’s disease-associated pathogenic mechanisms of degeneration and provides a new model to further investigate the consequences of cytosolic dopamine.
Collapse
Affiliation(s)
- Meghan L Bucher
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neuroscience, University of Pittsburgh School of Arts and Sciences, Pittsburgh, PA, USA.,Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Caitlyn W Barrett
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Connor J Moon
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neuroscience, University of Pittsburgh School of Arts and Sciences, Pittsburgh, PA, USA
| | - Amanda D Mortimer
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Edward A Burton
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Geriatric Research, Education and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Teresa G Hastings
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA. .,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Department of Neuroscience, University of Pittsburgh School of Arts and Sciences, Pittsburgh, PA, USA.
| |
Collapse
|
49
|
Hu Y, Korovaichuk A, Astiz M, Schroeder H, Islam R, Barrenetxea J, Fischer A, Oster H, Bringmann H. Functional Divergence of Mammalian TFAP2a and TFAP2b Transcription Factors for Bidirectional Sleep Control. Genetics 2020; 216:735-752. [PMID: 32769099 PMCID: PMC7648577 DOI: 10.1534/genetics.120.303533] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/20/2020] [Indexed: 11/18/2022] Open
Abstract
Sleep is a conserved behavioral state. Invertebrates typically show quiet sleep, whereas in mammals, sleep consists of periods of nonrapid-eye-movement sleep (NREMS) and REM sleep (REMS). We previously found that the transcription factor AP-2 promotes sleep in Caenorhabditiselegans and Drosophila In mammals, several paralogous AP-2 transcription factors exist. Sleep-controlling genes are often conserved. However, little is known about how sleep genes evolved from controlling simpler types of sleep to govern complex mammalian sleep. Here, we studied the roles of Tfap2a and Tfap2b in sleep control in mice. Consistent with our results from C. elegans and Drosophila, the AP-2 transcription factors Tfap2a and Tfap2b also control sleep in mice. Surprisingly, however, the two AP-2 paralogs play contrary roles in sleep control. Tfap2a reduction of function causes stronger delta and theta power in both baseline and homeostasis analysis, thus indicating increased sleep quality, but did not affect sleep quantity. By contrast, Tfap2b reduction of function decreased NREM sleep time specifically during the dark phase, reduced NREMS and REMS power, and caused a weaker response to sleep deprivation. Consistent with the observed signatures of decreased sleep quality, stress resistance and memory were impaired in Tfap2b mutant animals. Also, the circadian period was slightly shortened. Taken together, AP-2 transcription factors control sleep behavior also in mice, but the role of the AP-2 genes functionally diversified to allow for a bidirectional control of sleep quality. Divergence of AP-2 transcription factors might perhaps have supported the evolution of more complex types of sleep.
Collapse
Affiliation(s)
- Yang Hu
- Max Planck Research Group "Sleep and Waking", Max Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany
| | - Alejandra Korovaichuk
- Max Planck Research Group "Sleep and Waking", Max Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany
| | - Mariana Astiz
- Institute of Neurobiology, University of Lübeck, 23562, Germany
| | - Henning Schroeder
- German Center for Neurodegenerative Diseases, Göttingen 37075, Germany
| | - Rezaul Islam
- German Center for Neurodegenerative Diseases, Göttingen 37075, Germany
| | - Jon Barrenetxea
- Max Planck Research Group "Sleep and Waking", Max Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany
| | - Andre Fischer
- German Center for Neurodegenerative Diseases, Göttingen 37075, Germany
- Department for Psychiatry and Psychotherapy, University Medical Center, Göttingen 37075, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37073, Germany
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, 23562, Germany
| | - Henrik Bringmann
- Max Planck Research Group "Sleep and Waking", Max Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany
- Department of Animal Physiology/Neurophysiology, Philipps University Marburg, Marburg 35043, Germany
- BIOTEC of the Technical University Dresden, Dresden 01307, Germany
| |
Collapse
|
50
|
Shin T, Hiraoka Y, Yamasaki T, Marth JD, Penninger JM, Kanai-Azuma M, Tanaka K, Kofuji S, Nishina H. MKK7 deficiency in mature neurons impairs parental behavior in mice. Genes Cells 2020; 26:5-17. [PMID: 33098150 PMCID: PMC7839552 DOI: 10.1111/gtc.12816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 11/28/2022]
Abstract
c‐Jun N‐terminal kinases (JNKs) are constitutively activated in mammalian brains and are indispensable for their development and neural functions. MKK7 is an upstream activator of all JNKs. However, whether the common JNK signaling pathway regulates the brain's control of social behavior remains unclear. Here, we show that female mice in which Mkk7 is deleted specifically in mature neurons (Mkk7flox/floxSyn‐Cre mice) give birth to a normal number of pups but fail to raise them due to a defect in pup retrieval. To explore the mechanism underlying this abnormality, we performed comprehensive behavioral tests. Mkk7flox/floxSyn‐Cre mice showed normal locomotor functions and cognitive ability but exhibited depression‐like behavior. cDNA microarray analysis of mutant brain revealed an altered gene expression pattern. Quantitative RT‐PCR analysis demonstrated that mRNA expression levels of genes related to neural signaling pathways and a calcium channel were significantly different from controls. In addition, loss of neural MKK7 had unexpected regulatory effects on gene expression patterns in oligodendrocytes. These findings indicate that MKK7 has an important role in regulating the gene expression patterns responsible for promoting normal social behavior and staving off depression.
Collapse
Affiliation(s)
- Tadashi Shin
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yuichi Hiraoka
- Department of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tokiwa Yamasaki
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Jamey D Marth
- Center for Nanomedicine, Department of Molecular, Cellular and Developmental Biology, SBP Medical Discovery Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria.,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Masami Kanai-Azuma
- Department of Experimental Animal Model for Human Disease, Center for Experimental Animals, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kohichi Tanaka
- Department of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Satoshi Kofuji
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiroshi Nishina
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|