1
|
Lind-Holm Mogensen F, Seibler P, Grünewald A, Michelucci A. Microglial dynamics and neuroinflammation in prodromal and early Parkinson's disease. J Neuroinflammation 2025; 22:136. [PMID: 40399949 DOI: 10.1186/s12974-025-03462-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/05/2025] [Indexed: 05/23/2025] Open
Abstract
Parkinson's disease (PD) is characterized by a drastic loss of dopaminergic neurons already at diagnosis. As this loss of neurons starts decades before diagnosis, understanding the prodromal stages of the disease might offer novel strategies to curb its progression. While the precise pathogenic mechanisms underlying PD remain incompletely understood, growing evidence suggests that neuroinflammation and immune dysregulation play a central role in the development and progression of the disease. Here, we delve into the emerging roles of microglia, the resident immune cells of the central nervous system, in the pathogenesis of prodromal and early-stage PD. We emphasize that microglia contribute to neuroinflammation, protein aggregation and neurodegeneration, although the underlying mechanisms are not yet known. Neuroimaging studies have provided valuable insights into the patterns of microglial activation detected in individuals with prodromal PD and at the time of clinical diagnosis. Furthermore, we highlight the complex interplay between immune dysregulation and neurodegeneration along PD development, including alterations in the peripheral immune system, brain-gut interactions and brain-immune interfaces. Lastly, we outline existing models for investigating microglial involvement in prodromal PD, along with the impact of anti-inflammatory therapies and strategies to modify risk factors. In conclusion, targeting microglial activation and immune dysfunctions in individuals at risk of PD could represent a promising preventive measure and may offer novel therapeutic strategies for early intervention and disease modification.
Collapse
Affiliation(s)
- Frida Lind-Holm Mogensen
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, Luxembourg, L-1210, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, 2, avenue de l'Université, Esch-sur-Alzette, L-4365, Luxembourg
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck and University Hospital Schleswig-Holstein, Ratzeburger Allee 160, Lübeck, 23562, Germany
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6, avenue du Swing, Belvaux, L-4367, Luxembourg
| | - Alessandro Michelucci
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, Luxembourg, L-1210, Luxembourg.
| |
Collapse
|
2
|
Huang YT, Yang TJ, Liu KC, Chen MC, Chan PYS, Chen JC. Intranasal α-Synuclein induces progressive behavioral impairments in mice. Behav Brain Res 2025; 485:115517. [PMID: 40024483 DOI: 10.1016/j.bbr.2025.115517] [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: 10/07/2024] [Revised: 12/23/2024] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
α-Synuclein (α-Syn) is implicated in the progression of Parkinson's disease, yet the disease's etiology remains unclear. This study aims to explore how α-Syn affects olfactory, motor, mood and cognitive functions if it initiates from the olfactory bulb. Mice were administered intranasal human AAV-α-Syn and subsequently evaluated for olfactory, motor, mood, and cognitive functions. Immunofluorescence was performed to assess dopaminergic neuronal damage. Results shown that olfactory dysfunction was evident as AAV-α-Syn-treated mice took longer to find buried pellets compared to controls at 3, 9, and 12 months post-instillation. Motor activity remained normal at 6 months but significantly declined at 9 months. Reduced tyrosine hydroxylase expression but increased amount of human α-Syn were observed in the substantia nigra at end of behavioral measurements. AAV-α-Syn mice showed reduced sucrose intake and decreased time in the center zone of the open field at 9 months. Cognitive deficits were observed in recognition function and social memory at 6 and 9 months, with impaired working memory at 12 months. Thus, intranasal AAV-α-Syn instillation in mice leads to progressive olfactory, motor, anxiety, depression-like, and cognitive dysfunctions, reflecting α-Syn pathology propagation.
Collapse
Affiliation(s)
- Yu-Ting Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tzu-Jung Yang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kou-Chen Liu
- Department of Electronic Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Min-Chi Chen
- Department of Public Health and Biostatistics Consulting Center, Chang Gung University, Taoyuan, Taiwan; Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Pei-Ying S Chan
- Department of Occupational Therapy, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Psychiatry, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Jin-Chung Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Neuroscience Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
3
|
Abraham JN, Rawat D, Srikanth P, Sunny LP, Abraham NM. Alpha-synuclein pathology and Parkinson's disease-related olfactory dysfunctions: an update on preclinical models and therapeutic approaches. Mamm Genome 2025:10.1007/s00335-025-10128-w. [PMID: 40293510 DOI: 10.1007/s00335-025-10128-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 04/03/2025] [Indexed: 04/30/2025]
Abstract
Olfactory dysfunction (OD) is considered one of the early signs of Parkinson's disease (PD), affecting over 90% of PD patients. OD often appears several years before the onset of motor symptoms and is therefore considered an early biomarker of PD. Recent studies have shown that COVID-19 infection might lead to worsening of symptoms and acceleration of disease progression in neurodegenerative disorders, where OD is a common symptom to both. Hence, it is essential to accurately monitor olfactory fitness in clinical settings using any of the currently available olfactory function tests. Even after a quarter of a century of the discovery of α-synuclein (α-syn) pathogenesis in PD, many aspects related to the α-syn pathogenesis in OD remain unknown. Currently, there is no definitive cure for PD; the disease management options include dopaminergic medications, deep brain stimulations, stem cells, and immunotherapy. Generating reliable PD animal models is critical for understanding the molecular pathways and neural circuits affected by disease conditions. This might contribute to the development and validation of new therapeutic approaches. This review discusses the known mechanisms of α-syn aggregated forms causing neuronal death, the recent developments in the PD preclinical models with ODs, and the treatment strategies employed.
Collapse
Affiliation(s)
- Jancy Nixon Abraham
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India.
- Department of Life Sciences, Centre of Excellence in Epigenetics, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh, 201314, India.
| | - Devesh Rawat
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Priyadharshini Srikanth
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Lisni P Sunny
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India
| | - Nixon M Abraham
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, Maharashtra, 411008, India.
| |
Collapse
|
4
|
Pimenta JC, Beltrami VA, Oliveira BDS, Queiroz-Junior CM, Barsalini J, Teixeira DC, de Souza-Costa LP, Lima ALD, Machado CA, Parreira BZSG, Santos FRDS, Costa PAC, Lacerda LDSB, Gonçalves MR, Chaves IDM, Couto MGG, Costa VRDM, Nóbrega NRC, Silva BL, Fonseca T, Resende F, Wnuk NT, Marim FM, Rocha FEO, Umezu HL, Campolina-Silva G, Andrade ACDSP, de Aguiar RS, Costa GMJ, Guimarães PPG, Silva GSF, Rachid MA, Vieira LB, Pinho V, Teixeira AL, Teixeira MM, Miranda AS, Costa VV. Neuropsychiatric sequelae in an experimental model of post-COVID syndrome in mice. Brain Behav Immun 2025; 128:16-36. [PMID: 40120834 DOI: 10.1016/j.bbi.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/17/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
The global impact of the COVID-19 pandemic has been unprecedented, and presently, the world is facing a new challenge known as post-COVID syndrome (PCS). Current estimates suggest that more than 100 million people are grappling with PCS, encompassing several manifestations, including pulmonary, musculoskeletal, metabolic, and neuropsychiatric sequelae (cognitive and behavioral). The mechanisms underlying PCS remain unclear. The present study aimed to: (i) comprehensively characterize the acute effects of pulmonary inoculation of the betacoronavirus MHV-A59 in immunocompetent mice at clinical, cellular, and molecular levels; (ii) examine potential acute and long-term pulmonary, musculoskeletal, and neuropsychiatric sequelae induced by the betacoronavirus MHV-A59; and to (iii) assess sex-specific differences. Male and female C57Bl/6 mice were initially inoculated with varying viral titers (3x103 to 3x105 PFU/30 μL) of the betacoronavirus MHV-A59 via the intranasal route to define the highest inoculum capable of inducing disease without causing mortality. Further experiments were conducted with the 3x104 PFU inoculum. Mice exhibited an altered neutrophil/lymphocyte ratio in the blood in the 2nd and 5th day post-infection (dpi). Marked lung lesions were characterized by hyperplasia of the alveolar walls, infiltration of polymorphonuclear leukocytes (PMN) and mononuclear leukocytes, hemorrhage, increased concentrations of CCL2, CCL3, CCL5, and CXCL1 chemokines, as well as high viral titers until the 5th dpi. While these lung inflammatory signs resolved, other manifestations were observed up to the 60 dpi, including mild brain lesions with gliosis and hyperemic blood vessels, neuromuscular dysfunctions, anhedonic-like behavior, deficits in spatial working memory, and short-term aversive memory. These musculoskeletal and neuropsychiatric complications were exclusive to female mice and prevented after ovariectomy. In summary, our study describes for the first time a novel sex-dependent model of PCS focused on neuropsychiatric and musculoskeletal disorders. This model provides a unique platform for future investigations regarding the effects of acute therapeutic interventions on the long-term sequelae unleashed by betacoronavirus infection.
Collapse
Affiliation(s)
- Jordane Clarisse Pimenta
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vinícius Amorim Beltrami
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bruna da Silva Oliveira
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jéssica Barsalini
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danielle Cunha Teixeira
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiz Pedro de Souza-Costa
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Anna Luiza Diniz Lima
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Caroline Amaral Machado
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Felipe Rocha da Silva Santos
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro Augusto Carvalho Costa
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Matheus Rodrigues Gonçalves
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ian de Meira Chaves
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Manoela Gonzaga Gontijo Couto
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Victor Rodrigues de Melo Costa
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Bárbara Luísa Silva
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Talita Fonseca
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Filipe Resende
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Natália Teixeira Wnuk
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fernanda Martins Marim
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Felipe Emanuel Oliveira Rocha
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Hanna L Umezu
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gabriel Campolina-Silva
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Ana Cláudia Dos Santos Pereira Andrade
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Department of Microbiology and Immunology, Université Laval, Quebec, Canada
| | - Renato Santana de Aguiar
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Guilherme Mattos Jardim Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro Pires Goulart Guimarães
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Glauber Santos Ferreira Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Milene Alvarenga Rachid
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luciene Bruno Vieira
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vanessa Pinho
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Antônio Lúcio Teixeira
- Biggs Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Aline Silva Miranda
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Vivian Vasconcelos Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
5
|
Soares ES, Queiroz LY, Canever JB, Griebner G, Stahler CU, Mansur DS, Prediger RDS, Cimarosti HI. SENP3 knockdown improves motor and cognitive impairments in the intranasal MPTP rodent model of Parkinson's disease. Physiol Behav 2025; 288:114725. [PMID: 39488250 DOI: 10.1016/j.physbeh.2024.114725] [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: 07/09/2024] [Revised: 09/04/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Several mechanisms underlying Parkinson's disease (PD) remain unclear, and effective treatments are still lacking. The conjugation of the small ubiquitin-like modifier (SUMO), known as SUMOylation, to key proteins in PD has shown potential beneficial effects. Considering that this process is reversed by SUMO-specific proteases (SENPs), this study addressed the effects of increased SUMO-2/3 conjugation, mediated by SENP3 knockdown, in the intranasal 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) rodent model of PD. Two weeks after infusion of the shRNA-containing lentiviral vector into the dorsolateral striatum and one week following intranasal MPTP administration, male Wistar rats were evaluated using cognitive and motor behavioural tests. Infection efficiency was confirmed by detecting GFP expression in the dorsolateral striatum. SENP3 knockdown, verified by Western blotting, resulted in increased SUMO-2/3 conjugation. MPTP-administered rats displayed impairments in both recognition and spatial memories, while SENP3 knockdown prevented these deficits. Rats exposed to MPTP also exhibited motor dysfunction, which was ameliorated by SENP3 knockdown. These findings underscore the involvement of SUMO-2/3 conjugation in PD and its potential as a novel therapeutic target to counteract cognitive and motor impairments induced by neurodegeneration.
Collapse
Affiliation(s)
- Ericks S Soares
- Department of Pharmacology, Postgraduate Program in Pharmacology, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Leticia Y Queiroz
- Department of Pharmacology, Postgraduate Program in Pharmacology, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil; Postgraduate Program in Neuroscience, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Jaquelini B Canever
- Postgraduate Program in Neuroscience, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Gustavo Griebner
- Department of Pharmacology, Postgraduate Program in Pharmacology, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Carolina U Stahler
- Department of Pharmacology, Postgraduate Program in Pharmacology, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Daniel S Mansur
- Department of Microbiology, Immunology, and Parasitology, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Rui Daniel S Prediger
- Department of Pharmacology, Postgraduate Program in Pharmacology, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil; Postgraduate Program in Neuroscience, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Helena I Cimarosti
- Department of Pharmacology, Postgraduate Program in Pharmacology, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil; Postgraduate Program in Neuroscience, Centre for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil.
| |
Collapse
|
6
|
Soni R, Mathur K, Rathod H, Khairnar A, Shah J. Hyperglycemia-Driven Insulin Signaling Defects Promote Parkinson's Disease-like Pathology in Mice. ACS Pharmacol Transl Sci 2024; 7:4155-4164. [PMID: 39698281 PMCID: PMC11650731 DOI: 10.1021/acsptsci.4c00586] [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: 10/07/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
This study aims to determine the effect of chronic hyperglycemia, induced by a high-fat diet and STZ-induced diabetes, on the development of Parkinson's disease-like characteristics. Understanding this relationship is crucial in pharmacology, neurology, and diabetes, as it could potentially lead to developing new therapeutic strategies for Parkinson's disease. Our study employed a comprehensive approach to investigate the effect of hyperglycemia on Parkinson's disease-like characteristics. Hyperglycemia was induced by a high-fat diet for 6- and 9-week duration with a single intraperitoneal STZ (100 mg/kg) injection at week 5 in C57/BL6 mice. Rotenone (10 mg/kg p.o.) was administered to C57/BL6 mice for 6 and 9 weeks. Time-dependent behavioral studies (wire-hang tests, pole tests, Y-maze tests, and round beam walk tests) were carried out to monitor pathology progression and deficits. Molecular protein levels (GLP1, PI3K, AKT, GSK-3β, NF-κB, and α-syn), oxidative stress (GSH and MDA) parameters, and histopathological alterations (H&E and Nissl staining) were determined after 6 weeks as well as 9 weeks. After 9 weeks of study, molecular protein expression (p-AKT and p-α-syn) was determined. Hyperglycemia induced by HFD and STZ induced significant motor impairment in mice, correlated with the rotenone group. Insulin receptor signaling (GLP1/PI3K/AKT) was found to be disrupted in the HFD+STZ group and also in rotenone-treated mice, which further enhanced phosphorylation of α-syn, suggesting its role in α-syn accumulation. Histopathological alterations indicating neuroinflammation and neurodegeneration were quite evident in the HFD+STZ and rotenone groups. Exposure to hyperglycemia induced by HFD+STZ administration exhibits PD-like characteristics after 9 weeks of duration, which was correlative with rotenone-induced PD-like symptoms.
Collapse
Affiliation(s)
- Ritu Soni
- Department
of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Kirti Mathur
- Department
of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Hritik Rathod
- Department
of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Amit Khairnar
- International
Clinical Research Centre, St. Anne’s
University Hospital, Brno, Czech Republic, ICRC, FNUSA, Brno 60200, Czechia
- Department
of Physiology, Faculty of Medicine, Masaryk
University, Kamenice
753/5, Brno 62500, Czechia
- International
Clinical Research Centre, Faculty of Medicine, Masaryk University, Kamenice 753/5, Brno 62500, Czechia
| | - Jigna Shah
- Department
of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| |
Collapse
|
7
|
Gorskaya AV, Vasilev DS. Problems in the Diagnosis of Dysfunctions of the Olfactory Analyzer in Laboratory Animals Based on Behavioral and Electrophysiological Study Methods. NEUROSCIENCE AND BEHAVIORAL PHYSIOLOGY 2024; 54:990-1002. [DOI: 10.1007/s11055-024-01702-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2025]
|
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
|
Oliveira BDS, Toscano ECDB, Abreu LKS, Fernandes HDB, Amorim RF, Ferreira RN, Machado CA, Carvalho BC, da Silva MCM, de Oliveira ACP, Rachid MA, Rocha NP, Teixeira AL, da Silva ER, de Miranda AS. Nigrostriatal Inflammation Is Associated with Nonmotor Symptoms in an Experimental Model of Prodromal Parkinson's Disease. Neuroscience 2024; 549:65-75. [PMID: 38750924 DOI: 10.1016/j.neuroscience.2024.05.011] [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: 01/02/2024] [Revised: 04/17/2024] [Accepted: 05/09/2024] [Indexed: 05/21/2024]
Abstract
Recent evidence has supported a pathogenic role for neuroinflammation in Parkinson's disease (PD). Inflammatory response has been associated with symptoms and subtypes of PD. However, it is unclear whether immune changes are involved in the initial pathogenesis of PD, leading to the non-motor symptoms (NMS) observed in its prodromal stage. The current study aimed to characterize the behavioral and cognitive changes in a toxin-induced model of prodromal PD-like syndrome. We also sought to investigate the role of neuroinflammation in prodromal PD-related NMS. Male mice were subjected to bilateral intranasal infusion with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or saline (control group), followed by comprehensive behavioral, pathological and neurochemical analysis. Intranasal MPTP infusion was able to cause the loss of dopaminergic neurons in the substantia nigra (SN). In parallel, it induced impairment in olfactory discrimination and social memory consolidation, compulsive and anxiety-like behaviors, but did not influence motor performance. Iba-1 and GFAP expressions were increased in the SN, suggesting an activated state of microglia and astrocytes. Consistent with this, MPTP mice had increased levels of IL-10 and IL-17A, and decreased levels of BDNF and TrkA mRNA in the SN. The striatum showed increased IL-17A, BDNF, and NFG levels compared to control mice. In conclusion, neuroinflammation may play an important role in the early stage of experimental PD-like syndrome, leading to cognitive and behavioral changes. Our results also indicate that intranasal administration of MPTP may represent a valuable mouse model for prodromal PD.
Collapse
Affiliation(s)
- Bruna da Silva Oliveira
- Laboratório de Neurobiologia "Conceição Machado", Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Eliana Cristina de Brito Toscano
- Departamento de Patologia, Faculdade de Medicina, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil; Programa de Pós-graduação em Saúde, Faculdade de Medicina, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Larissa Katharina Sabino Abreu
- Laboratório de Neurobiologia "Conceição Machado", Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Heliana de Barros Fernandes
- Laboratório de Neurobiologia "Conceição Machado", Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Renan Florindo Amorim
- Laboratório de Neurobiologia "Conceição Machado", Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rodrigo Novaes Ferreira
- Laboratório de Neurobiologia "Conceição Machado", Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Caroline Amaral Machado
- Laboratório de Neurobiologia "Conceição Machado", Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Brener Cunha Carvalho
- Laboratório de Genes Inflamatórios, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria Carolina Machado da Silva
- Laboratório de Neurofarmacologia, Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Antônio Carlos Pinheiro de Oliveira
- Laboratório de Neurofarmacologia, Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Milene Alvarenga Rachid
- Laboratório de Patologia Celular e Molecular, Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | - Natália Pessoa Rocha
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, TX, USA
| | - Antônio Lúcio Teixeira
- Instituto de Ensino e Pesquisa, Santa Casa BH, Belo Horizonte, Brazil; Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, TX, USA
| | - Elizabeth Ribeiro da Silva
- Laboratório de Neurobiologia "Conceição Machado", Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Aline Silva de Miranda
- Laboratório de Neurobiologia "Conceição Machado", Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
10
|
Pimenta J, Da Silva Oliveira B, Lima ALD, Machado CA, De Souza Barbosa Lacerda L, Rossi L, Queiroz-Junior CM, De Souza-Costa LP, Andrade ACSP, Gonçalves MR, Mota B, Marim FM, Aguiar RS, Guimarães PPG, Teixeira AL, Vieira LB, Guatimosim C, Teixeira MM, De Miranda AS, Costa VV. A suitable model to investigate acute neurological consequences of coronavirus infection. Inflamm Res 2023; 72:2073-2088. [PMID: 37837557 DOI: 10.1007/s00011-023-01798-w] [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: 06/02/2023] [Revised: 09/06/2023] [Accepted: 09/13/2023] [Indexed: 10/16/2023] Open
Abstract
OBJECTIVE AND DESIGN The present study aimed to investigate the neurochemical and behavioral effects of the acute consequences after coronavirus infection through a murine model. MATERIAL Wild-type C57BL/6 mice were infected intranasally (i.n) with the murine coronavirus 3 (MHV-3). METHODS Mice underwent behavioral tests. Euthanasia was performed on the fifth day after infection (5 dpi), and the brain tissue was subjected to plaque assays for viral titration, ELISA, histopathological, immunohistochemical and synaptosome analysis. RESULTS Increased viral titers and mild histological changes, including signs of neuronal degeneration, were observed in the cerebral cortex of infected mice. Importantly, MHV-3 infection induced an increase in cortical levels of glutamate and calcium, which is indicative of excitotoxicity, as well as increased levels of pro-inflammatory cytokines (IL-6, IFN-γ) and reduced levels of neuroprotective mediators (BDNF and CX3CL1) in the mice brain. Finally, behavioral analysis showed impaired motor, anhedonia-like and anxiety-like behaviors in animals infected with MHV-3. CONCLUSIONS In conclusion, the data presented emulate many aspects of the acute neurological outcomes seen in patients with COVID-19. Therefore, this model may provide a preclinical platform to study acute neurological sequelae induced by coronavirus infection and test possible therapies.
Collapse
Affiliation(s)
- Jordane Pimenta
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, MG, 31270-901, Brazil
| | - Bruna Da Silva Oliveira
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, MG, 31270-901, Brazil
| | - Anna Luiza Diniz Lima
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Caroline Amaral Machado
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, MG, 31270-901, Brazil
| | - Larisse De Souza Barbosa Lacerda
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, MG, 31270-901, Brazil
| | - Leonardo Rossi
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, MG, 31270-901, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, MG, 31270-901, Brazil
| | - Luiz Pedro De Souza-Costa
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana Claudia Santos Pereira Andrade
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, MG, 31270-901, Brazil
| | - Matheus Rodrigues Gonçalves
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bárbara Mota
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, MG, 31270-901, Brazil
| | - Fernanda Martins Marim
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Renato Santana Aguiar
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro Pires Goulart Guimarães
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Antônio Lúcio Teixeira
- Department of Psychiatry and Behavioral Sciences, McGovern Medical Houston, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Luciene Bruno Vieira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Cristina Guatimosim
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, MG, 31270-901, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Aline Silva De Miranda
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, MG, 31270-901, Brazil.
| | - Vivian Vasconcelos Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
11
|
Kim W, Tripathi M, Kim C, Vardhineni S, Cha Y, Kandi SK, Feitosa M, Kholiya R, Sah E, Thakur A, Kim Y, Ko S, Bhatia K, Manohar S, Kong YB, Sindhu G, Kim YS, Cohen B, Rawat DS, Kim KS. An optimized Nurr1 agonist provides disease-modifying effects in Parkinson's disease models. Nat Commun 2023; 14:4283. [PMID: 37463889 DOI: 10.1038/s41467-023-39970-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 07/05/2023] [Indexed: 07/20/2023] Open
Abstract
The nuclear receptor, Nurr1, is critical for both the development and maintenance of midbrain dopamine neurons, representing a promising molecular target for Parkinson's disease (PD). We previously identified three Nurr1 agonists (amodiaquine, chloroquine and glafenine) that share an identical chemical scaffold, 4-amino-7-chloroquinoline (4A7C), suggesting a structure-activity relationship. Herein we report a systematic medicinal chemistry search in which over 570 4A7C-derivatives were generated and characterized. Multiple compounds enhance Nurr1's transcriptional activity, leading to identification of an optimized, brain-penetrant agonist, 4A7C-301, that exhibits robust neuroprotective effects in vitro. In addition, 4A7C-301 protects midbrain dopamine neurons in the MPTP-induced male mouse model of PD and improves both motor and non-motor olfactory deficits without dyskinesia-like behaviors. Furthermore, 4A7C-301 significantly ameliorates neuropathological abnormalities and improves motor and olfactory dysfunctions in AAV2-mediated α-synuclein-overexpressing male mouse models. These disease-modifying properties of 4A7C-301 may warrant clinical evaluation of this or analogous compounds for the treatment of patients with PD.
Collapse
Affiliation(s)
- Woori Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Mohit Tripathi
- Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Chunhyung Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | | | - Young Cha
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | | | - Melissa Feitosa
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Rohit Kholiya
- Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Eric Sah
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Anuj Thakur
- Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Yehan Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Sanghyeok Ko
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Kaiya Bhatia
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Sunny Manohar
- Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Young-Bin Kong
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Gagandeep Sindhu
- Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Yoon-Seong Kim
- Institute for Neurological Therapeutics, Rutgers University, Piscataway, NJ, 08854, USA
| | - Bruce Cohen
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Diwan S Rawat
- Department of Chemistry, University of Delhi, Delhi, 110007, India.
| | - Kwang-Soo Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA.
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA.
| |
Collapse
|
12
|
Savall ASP, Fidelis EM, de Mello JD, Quines CB, Denardin CC, Marques LS, Klann IP, Nogueira CW, Sampaio TB, Pinton S. Neuroprotective effect of Eugenia uniflora against intranasal MPTP-induced memory impairments in rats: The involvement of pro-BDNF/p75 NTR pathway. Life Sci 2023; 324:121711. [PMID: 37088413 DOI: 10.1016/j.lfs.2023.121711] [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: 12/20/2022] [Revised: 04/17/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
Parkinson's disease is a multisystemic neurodegenerative disorder that includes motor and non-motor symptoms, and common symptoms include memory loss and learning difficulties. Thus, we investigated the neuroprotective potential of a hydroalcoholic extract of Brazilian purple cherry (Eugenia uniflora) (HAE-BC) on memory impairments induced by intranasal 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration in rats and the involvement of hippocampal BDNF/TrkB/p75NTR pathway in its effects. Adult male Wistar rats were exposed to MPTP (1 mg/nostril) or vehicle. Twenty-four hours later, the HAE-BC treatments began at doses of 300 or 2000 mg/kg/day or vehicle for 14 days. From 7 days after the MPTP induction, the animals were subjected to behavioral tests to evaluate several cognitive paradigms. HAE-BC treatments, at both doses, blocked the MPTP-caused disruption in the social recognition memory, short- and long-term object recognition memories, and working memory. Furthermore, MPTP-induced motor deficit linked to striatal tyrosine hydroxylase levels decreased, which was blocked by HAE-BC. Our findings demonstrated that HAE-BC blocked the MPTP-induced increase in the hippocampal pro-BDNF, TrkB.t1, and p75NTR levels. The pro-BDNF/p75NTR interaction negatively regulates synaptic transmission and plasticity, and the neuroprotective effect of HAE-BC was related, at least partly, to the modulation of this hippocampal signaling pathway. Thus, our study reports the first evidence of the potential therapeutic of E. uniflora in a Parkinson's disease model in rodents.
Collapse
Affiliation(s)
| | | | | | | | | | - Luiza Souza Marques
- Federal University of Santa Maria - Campus Camobi, Santa Maria CEP 97105-900, RS, Brazil
| | | | | | | | - Simone Pinton
- Federal University of Pampa - Campus Uruguaiana, Uruguaiana CEP 97500-970, RS, Brazil.
| |
Collapse
|
13
|
Gallegos CE, Bartos M, Gumilar F, Minetti A, Baier CJ. Behavioral and neurochemical impairments after intranasal administration of chlorpyrifos formulation in mice. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 189:105315. [PMID: 36549818 DOI: 10.1016/j.pestbp.2022.105315] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/17/2022] [Accepted: 12/06/2022] [Indexed: 06/17/2023]
Abstract
Among the most relevant environmental factors associated with the etiology of neurodegenerative disorders are pesticides. Spray drift or volatilization generates pesticide dispersion after its application. In addition, inhalation or intranasal (IN) administration of xenobiotics constitutes a feasible route for substance delivery to the brain. This study investigates the behavioral and neurochemical effects of IN exposure to a commercial formulation of chlorpyrifos (fCPF). Adult male CF-1 mice were intranasally administered with fCPF (3-10 mg/kg/day) three days a week, for 2 weeks. Behavioral and biochemical analyses were conducted 20 and 30 days after the last IN fCPF administration, respectively. No significant behavioral or biochemical effects were observed in the 3 mg/kg fCPF IN exposure group. However, animals exposed to 10 mg/kg fCPF showed anxiogenic behavior and recognition memory impairment, with no effects on locomotor activity. In addition, the IN administration of 10 mg/kg fCPF altered the redox balance, modified the activity of enzymes belonging to the cholinergic and glutamatergic pathways, and affected glucose metabolism, and cholesterol levels in different brain areas. Taken together, these observations suggest that these biochemical imbalances could be responsible for the neurobehavioral disturbances observed after IN administration of fCPF in mice.
Collapse
Affiliation(s)
- Cristina Eugenia Gallegos
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Departamento de Biología, Bioquímica y Farmacia (DBByF), San Juan 670, B8000ICN Bahía Blanca, Argentina
| | - Mariana Bartos
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Departamento de Biología, Bioquímica y Farmacia (DBByF), San Juan 670, B8000ICN Bahía Blanca, Argentina
| | - Fernanda Gumilar
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Departamento de Biología, Bioquímica y Farmacia (DBByF), San Juan 670, B8000ICN Bahía Blanca, Argentina
| | - Alejandra Minetti
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Departamento de Biología, Bioquímica y Farmacia (DBByF), San Juan 670, B8000ICN Bahía Blanca, Argentina
| | - Carlos Javier Baier
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Departamento de Biología, Bioquímica y Farmacia (DBByF), San Juan 670, B8000ICN Bahía Blanca, Argentina.
| |
Collapse
|
14
|
Chen HC, Ma YZ, Cao JX, Zhang YS, Zhang L, Gao LP, Jing YH. Synergistic effects of hIAPP and Aβ 1-42 impaired the olfactory function associated with the decline of adult neurogenesis in SVZ. Neuropeptides 2022; 96:102268. [PMID: 35841876 DOI: 10.1016/j.npep.2022.102268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/22/2022] [Accepted: 06/26/2022] [Indexed: 10/17/2022]
Abstract
According to many in the field,the prevalence of Alzheimer's disease (AD) in type II diabetes (T2DM) populations is considerably higher than that in the normal population. Human islet amyloid polypeptide (hIAPP) is considered to be a common risk factor for T2DM and AD. Preliminary observations around T2DM animal model show that the decrease of adult neural stem cells (NSCs) in the subventricular zone (SVZ) is accompanied by olfactory dysfunction. Furthermore, impaired olfactory function could serve as to an early predictor of neurodegeneration,which is associated with cognitive impairment. However, the synergistic effects between hIAPP and amyloid-beta (Aβ) 1-42 in the brain and the neurodegeneration remains to be further clarified. In this study, olfactory capacity, synaptic density, status of NSC in SVZ, and status of newborn neurons in olfactory bulb (OB) were assessed 6 months after stereotactic injection of oligomer Aβ1-42 into the dens gyrus (DG) of hIAPP-/+ mice or wild-type homogenous mice. Our results set out that Aβ42 and amylin co-localized into OB and raised Aβ42 deposition in hIAPP-/+ mice compared with wild-type brood mice. In addition, 6 months after injection of Aβ1-42 in hIAPP-/+ mice, these mice showed increased olfactory dysfunction, significant loss of synapses, depletion of NSC in SVZ, and impaired cell renewal in OB. Our present study suggested that the synergistic effects between hIAPP and Aβ1-42 impairs olfactory function and was associated with decreased neurogenesis in adults with SVZ.
Collapse
Affiliation(s)
- Hai-Chao Chen
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yue-Zhang Ma
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Jia-Xin Cao
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yi-Shu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Lu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Li-Ping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China; Key Laboratory of Preclinical Study for New Drugs of Gansu province, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yu-Hong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China; Key Laboratory of Preclinical Study for New Drugs of Gansu province, Lanzhou University, Lanzhou, Gansu, People's Republic of China.
| |
Collapse
|
15
|
The effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) on the cognitive and motor functions in rodents: A systematic review and meta-analysis. Neurosci Biobehav Rev 2022; 140:104792. [PMID: 35872230 DOI: 10.1016/j.neubiorev.2022.104792] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 07/16/2022] [Accepted: 07/20/2022] [Indexed: 11/24/2022]
Abstract
Memory and motor deficits are commonly identified in Parkinson's disease (PD). 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is transformed to MPP+ via monoamine oxidase B (MAOB), which causes oxidative stress and destroys dopaminergic (DA) neurons in substantia nigra pars compacta (SNc) and is widely used to create animal models of PD. However, to-date, a comprehensive analysis of the MPTP effects on various aspects of PD does not exist. Here, we provide a systematic review and meta-analysis on the MPTP effects on memory and motor functions by analyzing 51 studies on more than one thousand animals mainly including rats and mice. The results showed that in addition to motor functions such as coordination, balance and locomotor activity, MPTP significantly affects various mnemonic processes including spatial memory, working memory, recognition memory, and associative memory compared with the control group with some differences between systemic and intra-nigral injections on spatial memory, familiar object recognition, and anxiety-like behaviors. Nevertheless, our analysis failed to find systematic relationship between MPTP injection protocol parameters reported and the extent of the induced PD symptoms that can be a cause of concern for replicability of MPTP studies.
Collapse
|
16
|
Song B, Feldmann JW, Cao S, Feitosa M, Kong Y, Kim W, Schweitzer A, Leblanc P, Schweitzer JS, Kim KS. A Pitx3-deficient developmental mouse model for fine motor, olfactory, and gastrointestinal symptoms of Parkinson's disease. Neurobiol Dis 2022; 170:105777. [PMID: 35636646 PMCID: PMC9425627 DOI: 10.1016/j.nbd.2022.105777] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/01/2022] [Accepted: 05/21/2022] [Indexed: 10/26/2022] Open
|
17
|
Avanipully JN, Thekkekkara D, M S, Parihar VK, Manjula SN. The Role of Olfactory System in the Etiogenesis of Parkinson’s Diseases: An Overview. J Pharmacol Pharmacother 2022. [DOI: 10.1177/0976500x221085802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Parkinson’s disease (PD) mainly affects the dopaminergic neuronal networks of the substantia nigra, which leads to both motor and nonmotor symptoms of the disease. Based on the reports from the previous studies, 95% of the cases are presented along with olfactory dysfunction. The relevant publications from 2002 to 2021 were searched and shortlisted using PubMed and Google Scholar. In this review, we have discussed the correlation between olfactory dysfunction and PD. Olfactory damage presents earlier than the motor symptoms. Because there are no current methodologies for the early detection of PD, olfactory dysfunction can be used as a potential marker for the early detection of PD and hence paving the way for better therapeutic approaches.
Collapse
Affiliation(s)
| | - Dithu Thekkekkara
- Department of Pharmacology, JSS College of Pharmacy, JSSAHER, Mysuru-570015, Karnataka, India
| | - Sahyadri M
- Department of Pharmacology, JSS College of Pharmacy, JSSAHER, Mysuru-570015, Karnataka, India
| | - Vipan K. Parihar
- Department of Pharmacology and Toxicology, NIPER-Hajipur, Bihar-844102, India
| | | |
Collapse
|
18
|
Pereira MCL, Boese AC, Murad R, Yin J, Hamblin MH, Lee JP. Reduced dopaminergic neuron degeneration and global transcriptional changes in Parkinson's disease mouse brains engrafted with human neural stems during the early disease stage. Exp Neurol 2022; 352:114042. [PMID: 35271839 DOI: 10.1016/j.expneurol.2022.114042] [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] [Received: 12/15/2021] [Revised: 02/16/2022] [Accepted: 03/03/2022] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Current stem cell therapies for Parkinson's disease (PD) focus on a neurorestorative approach that aims to repair the CNS during the symptomatic phase. However, the pleiotropic and supportive effects of human neural stem cells (hNSCs) may make them effective for PD treatment during the disease's earlier stages. In the current study, we investigated the therapeutic effects of transplanting hNSCs during the early stages of PD development when most dopaminergic neurons are still present and before symptoms appear. Previous studies on hNSCs in Parkinson's disease focus on the substantia nigra and its immediate surroundings, but other brain structures are affected in PD as well. Here, we investigated the therapeutic effects of hNSCs on the entire PD-afflicted brain transcriptome using RNA sequencing (RNA-seq). METHODS PD was induced with a single intranasal infusion of 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) and hNSCs were transplanted unilaterally into the striatum one week later. The timepoint for hNSC transplantation coincided with upregulation of endogenous proinflammatory cytokines in the CNS, which play a role in stem cell migration. At 3 weeks post-transplantation (4 weeks post-MPTP), we assessed motor symptoms through behavioral tests, quantified dopaminergic neurons in the substantia nigra, and performed global transcriptional profiling to understand the mechanism underlying the effect of hNSCs on dopaminergic neuron degeneration. RESULTS We found that early hNSC engraftment mitigated motor symptoms induced by MPTP, and also reduced MPTP-induced loss of dopaminergic neurons. In this study, we uniquely presented the first comprehensive analysis of the effect of hNSC transplantation on the transcriptional profiling of PD mouse brains showing decreased expression of 249 and increased expression of 200 genes. These include genes implicated in mitochondrial bioenergetics, proteostasis, and other signaling pathways associated with improved PD outcome following hNSC transplantation. CONCLUSION These findings indicate that NSC transplantation during the asymptomatic phase of PD may limit or halt the progression of this neurodegenerative disorder. Transcriptional profiling of hNSC-engrafted PD mouse brains provides mechanistic insight that could lead to novel approaches to ameliorating degeneration of dopaminergic neurons and improving behavioral dysfunction in PD.
Collapse
Affiliation(s)
- Marcia C L Pereira
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Austin C Boese
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Rabi Murad
- Bioinformatics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Jun Yin
- Bioinformatics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Milton H Hamblin
- Tulane University Health Sciences Center, Tulane University, New Orleans, LA 70112, USA
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
19
|
Kim J, Park I, Jang S, Choi M, Kim D, Sun W, Choe Y, Choi JW, Moon C, Park SH, Choe HK, Kim K. Pharmacological Rescue with SR8278, a Circadian Nuclear Receptor REV-ERBα Antagonist as a Therapy for Mood Disorders in Parkinson's Disease. Neurotherapeutics 2022; 19:592-607. [PMID: 35322351 PMCID: PMC9226214 DOI: 10.1007/s13311-022-01215-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2022] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease is a neurodegenerative disease characterized by progressive dopaminergic neuronal loss. Motor deficits experienced by patients with Parkinson's disease are well documented, but non-motor symptoms, including mood disorders associated with circadian disturbances, are also frequent features. One common phenomenon is "sundowning syndrome," which is characterized by the occurrence of neuropsychiatric symptoms at a specific time (dusk), causing severe quality of life challenges. This study aimed to elucidate the underlying mechanisms of sundowning syndrome in Parkinson's disease and their molecular links with the circadian clock. We demonstrated that 6-hydroxydopamine (6-OHDA)-lesioned mice, as Parkinson's disease mouse model, exhibit increased depression- and anxiety-like behaviors only at dawn (the equivalent of dusk in human). Administration of REV-ERBα antagonist, SR8278, exerted antidepressant and anxiolytic effects in a circadian time-dependent manner in 6-OHDA-lesioned mice and restored the circadian rhythm of mood-related behaviors. 6-OHDA-lesion altered DAergic-specific Rev-erbα and Nurr1 transcription, and atypical binding activities of REV-ERBα and NURR1, which are upstream nuclear receptors for the discrete tyrosine hydroxylase promoter region. SR8278 treatment restored the binding activities of REV-ERBα and NURR1 to the tyrosine hydroxylase promoter and the induction of enrichment of the R/N motif, recognized by REV-ERBα and NURR1, as revealed by ATAC-sequencing; therefore, tyrosine hydroxylase expression was elevated in the ventral tegmental area of 6-OHDA-injected mice, especially at dawn. These results indicate that REV-ERBα is a potential therapeutic target, and its antagonist, SR8278, is a potential drug for mood disorders related to circadian disturbances, namely sundowning syndrome, in Parkinson's disease.
Collapse
Affiliation(s)
- Jeongah Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
- Department of Anatomy, College of Medicine, Korea University, Seoul, Korea
| | - Inah Park
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
| | - Sangwon Jang
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
| | - Mijung Choi
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
| | - Doyeon Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Woong Sun
- Department of Anatomy, College of Medicine, Korea University, Seoul, Korea
| | | | - Ji-Woong Choi
- Department of Electrical Engineering and Computer Science, DGIST, Daegu, Korea
| | - Cheil Moon
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
- Convergence Research Advanced Centre for Olfaction, DGIST, Daegu, Korea
| | - Sung Ho Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Korea
| | - Han Kyoung Choe
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
| | - Kyungjin Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea.
- Convergence Research Advanced Centre for Olfaction, DGIST, Daegu, Korea.
| |
Collapse
|
20
|
Chen H, Wang K, Scheperjans F, Killinger B. Environmental triggers of Parkinson's disease - Implications of the Braak and dual-hit hypotheses. Neurobiol Dis 2022; 163:105601. [PMID: 34954321 PMCID: PMC9525101 DOI: 10.1016/j.nbd.2021.105601] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/05/2021] [Accepted: 12/22/2021] [Indexed: 11/22/2022] Open
Abstract
Idiopathic Parkinson's disease (PD) may take decades to develop, during which many risk or protective factors may come into play to initiate the pathogenesis or modify its progression to clinical PD. The lack of understanding of this prodromal phase of PD and the factors involved has been a major hurdle in the study of PD etiology and preventive strategies. Although still controversial, the Braak and dual-hit hypotheses that PD may start peripherally in the olfactory structures and/or the gut provides a theoretical platform to identify the triggers and modifiers of PD prodromal development and progression. This is particularly true for the search of environmental causes of PD as the olfactory structures and gut are the major human mucosal interfaces with the environment. In this review, we lay out our personal views about how the Braak and dual-hit hypotheses may help us search for the environmental triggers and modifiers for PD, summarize available experimental and epidemiological evidence, and discuss research gaps and strategies.
Collapse
Affiliation(s)
- Honglei Chen
- Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA.
| | - Keran Wang
- Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Filip Scheperjans
- Department of Neurology, Helsinki University Hospital, and Clinicum, University of Helsinki, Haartmaninkatu 4, 00290 Helsinki, Finland
| | - Bryan Killinger
- Graduate College, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
21
|
Carta AR, Pisanu A, Palmas MF, Barcia C, Cuenca-Bermejo L, Herrero MT. MPTP: Advances from an Evergreen Neurotoxin. HANDBOOK OF NEUROTOXICITY 2022:485-516. [DOI: 10.1007/978-3-031-15080-7_104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
22
|
Fan Y, Han J, Zhao L, Wu C, Wu P, Huang Z, Hao X, Ji Y, Chen D, Zhu M. Experimental Models of Cognitive Impairment for Use in Parkinson's Disease Research: The Distance Between Reality and Ideal. Front Aging Neurosci 2021; 13:745438. [PMID: 34912207 PMCID: PMC8667076 DOI: 10.3389/fnagi.2021.745438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/01/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease. Cognitive impairment is one of the key non-motor symptoms of PD, affecting both mortality and quality of life. However, there are few experimental studies on the pathology and treatments of PD with mild cognitive impairment (PD-MCI) and PD dementia (PDD) due to the lack of representative models. To identify new strategies for developing representative models, we systematically summarized previous studies on PD-MCI and PDD and compared differences between existing models and diseases. Our initial search identified 5432 articles, of which 738 were duplicates. A total of 227 articles met our inclusion criteria and were included in the analysis. Models fell into three categories based on model design: neurotoxin-induced, transgenic, and combined. Although the neurotoxin-induced experimental model was the most common type that was used during every time period, transgenic and combined experimental models have gained significant recent attention. Unfortunately, there remains a big gap between ideal and actual experimental models. While each model has its own disadvantages, there have been tremendous advances in the development of PD models of cognitive impairment, and almost every model can verify a hypothesis about PD-MCI or PDD. Finally, our proposed strategies for developing novel models are as follows: a set of plans that integrate symptoms, biochemistry, neuroimaging, and other objective indicators to judge and identify that the novel model plays a key role in new strategies for developing representative models; novel models should simulate different clinical features of PD-MCI or PDD; inducible α-Syn overexpression and SH-SY5Y-A53T cellular models are good candidate models of PD-MCI or PDD.
Collapse
Affiliation(s)
- Yaohua Fan
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jiajun Han
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lijun Zhao
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Chunxiao Wu
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China.,Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peipei Wu
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zifeng Huang
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xiaoqian Hao
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - YiChun Ji
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Dongfeng Chen
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Meiling Zhu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
23
|
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
|
24
|
Vecchia DD, Kanazawa LKS, Wendler E, Hocayen PDAS, Vital MABF, Takahashi RN, Da Cunha C, Miyoshi E, Andreatini R. Ketamine reversed short-term memory impairment and depressive-like behavior in animal model of Parkinson's disease. Brain Res Bull 2021; 168:63-73. [PMID: 33359641 DOI: 10.1016/j.brainresbull.2020.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/01/2020] [Accepted: 12/18/2020] [Indexed: 12/15/2022]
Abstract
The most common features of Parkinson's disease (PD) are motor impairments, but many patients also present depression and memory impairment. Ketamine, an N-methyl-d-aspartate (NMDA) receptor antagonist, has been shown to be effective in patients with treatment-resistant major depression. Thus, the present study evaluated the action of ketamine on memory impairment and depressive-like behavior in an animal model of PD. Male Wistar rats received a bilateral infusion of 6 μg/side 6-hydroxydopamine (6-OHDA) into the substantia nigra pars compacta (SNc). Short-term memory was evaluated by the social recognition test, and depressive-like behaviors were evaluated by the sucrose preference and forced swimming tests (FST). Drug treatments included vehicle (i.p., once a week); ketamine (5, 10 and 15 mg/kg, i.p., once a week); and imipramine (20 mg/kg, i.p., daily). The treatments were administered 21 days after the SNc lesion and lasted for 28 days. The SNc lesion impaired short-term social memory, and all ketamine doses reversed the memory impairment and anhedonia (reduction of sucrose preference) induced by 6-OHDA. In the FST, 6-OHDA increased immobility, and all doses of ketamine and imipramine reversed this effect. The anti-immobility effect of ketamine was associated with an increase in swimming but not in climbing, suggesting a serotonergic effect. Ketamine and imipramine did not reverse the 6-OHDA-induced reduction in tyrosine hydroxylase immunohistochemistry in the SNc. In conclusion, ketamine reversed depressive-like behaviors and short-term memory impairment in rats with SNc bilateral lesions, indicating a promising profile for its use in PD patients.
Collapse
Affiliation(s)
- Débora Dalla Vecchia
- Department of Pharmacology, Setor de Ciências Biológicas, Universidade Federal do Paraná, Centro Politécnico, C.P. 19031, 81531-980, Curitiba, PR, Brazil; Uniandrade, Centro Universitário Campos de Andrade, Santa Quiteria, 80310-310, Curitiba, PR, Brazil
| | - Luiz Kae Sales Kanazawa
- Department of Pharmacology, Setor de Ciências Biológicas, Universidade Federal do Paraná, Centro Politécnico, C.P. 19031, 81531-980, Curitiba, PR, Brazil
| | - Etiéli Wendler
- Department of Pharmacology, Setor de Ciências Biológicas, Universidade Federal do Paraná, Centro Politécnico, C.P. 19031, 81531-980, Curitiba, PR, Brazil; Uniandrade, Centro Universitário Campos de Andrade, Santa Quiteria, 80310-310, Curitiba, PR, Brazil
| | - Palloma de Almeida Soares Hocayen
- Department of Pharmacology, Setor de Ciências Biológicas, Universidade Federal do Paraná, Centro Politécnico, C.P. 19031, 81531-980, Curitiba, PR, Brazil
| | - Maria Aparecida Barbato Frazão Vital
- Department of Pharmacology, Setor de Ciências Biológicas, Universidade Federal do Paraná, Centro Politécnico, C.P. 19031, 81531-980, Curitiba, PR, Brazil
| | - Reinaldo Naoto Takahashi
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, Trindade, 88049-900, Florianópolis, SC, Brazil
| | - Claudio Da Cunha
- Department of Pharmacology, Setor de Ciências Biológicas, Universidade Federal do Paraná, Centro Politécnico, C.P. 19031, 81531-980, Curitiba, PR, Brazil
| | - Edmar Miyoshi
- Department of Pharmaceutical Sciences, State University of Ponta Grossa, Avenida General Carlos Cavalcanti 4748, 84030-900, Ponta Grossa, PR, Brazil
| | - Roberto Andreatini
- Department of Pharmacology, Setor de Ciências Biológicas, Universidade Federal do Paraná, Centro Politécnico, C.P. 19031, 81531-980, Curitiba, PR, Brazil.
| |
Collapse
|
25
|
Antunes MS, Ladd FVL, Ladd AABL, Moreira AL, Boeira SP, Cattelan Souza L. Hesperidin protects against behavioral alterations and loss of dopaminergic neurons in 6-OHDA-lesioned mice: the role of mitochondrial dysfunction and apoptosis. Metab Brain Dis 2021; 36:153-167. [PMID: 33057922 DOI: 10.1007/s11011-020-00618-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 09/14/2020] [Indexed: 01/01/2023]
Abstract
Hesperidin is a flavonoid glycoside that is frequently found in citrus fruits. Our group have demonstrated that hesperidin has neuroprotective effect in 6-hydroxydopamine (6-OHDA) model of Parkinson's disease (PD), mainly by antioxidant mechanisms. Although the pathophysiology of PD remains uncertain, a large body of evidence has demonstrated that mitochondrial dysfunction and apoptosis play a critical role in dopaminergic nigrostriatal degeneration. However, the ability of hesperidin in modulating these mechanisms has not yet been investigated. In the present study, we examined the potential of a 28-day hesperidin treatment (50 mg/kg/day, p.o.) in preventing behavioral alterations induced by 6-OHDA injection via regulating mitochondrial dysfunction, apoptosis and dopaminergic neurons in the substantia nigra pars compacta (SNpc) in C57BL/6 mice. Our results demonstrated that hesperidin treatment improved motor, olfactory and spatial memory impairments elicited by 6-OHDA injection. Moreover, hesperidin treatment attenuated the loss of dopaminergic neurons (TH+ cells) in the SNpc and the depletion of dopamine (DA) and its metabolities 3,4-dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA) in the striatum of 6-OHDA-lesioned mice. Hesperidin also protected against the inhibition of mitochondrial respiratory chain complex-I, -IV and V, the decrease of Na + -K + -ATPase activity and the increase of caspase-3 and -9 activity in the striatum. Taken together, our findings indicate that hesperidin mitigates the degeneration of dopaminergic neurons in the SNpc by preventing mitochondrial dysfunction and modulating apoptotic pathways in the striatum of 6-OHDA-treated mice, thus improving behavioral alterations. These results provide new insights on neuroprotective mechanisms of hesperidin in a relevant preclinical model of PD.
Collapse
Affiliation(s)
- Michelle S Antunes
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui, RS, CEP 97650-000, Brazil
| | - Fernando Vagner Lobo Ladd
- Department of Morphology/Laboratory of Neuroanatom, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Aliny Antunes Barbosa Lobo Ladd
- Laboratory of Stochastic Stereology and Chemical Anatomy, Department of Surgery, College of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Amanda Lopez Moreira
- Laboratory of Stochastic Stereology and Chemical Anatomy, Department of Surgery, College of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Silvana Peterini Boeira
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui, RS, CEP 97650-000, Brazil
| | - Leandro Cattelan Souza
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui, RS, CEP 97650-000, Brazil.
| |
Collapse
|
26
|
Cerri S, Blandini F. In vivo modeling of prodromal stage of Parkinson’s disease. J Neurosci Methods 2020; 342:108801. [DOI: 10.1016/j.jneumeth.2020.108801] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 05/31/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022]
|
27
|
Neuroprotective effects of melatonin against neurotoxicity induced by intranasal sodium dimethyldithiocarbamate administration in mice. Neurotoxicology 2020; 80:144-154. [PMID: 32738267 DOI: 10.1016/j.neuro.2020.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/30/2020] [Accepted: 07/22/2020] [Indexed: 12/14/2022]
Abstract
Exposure to fungicide ziram (zinc dimethyldithiocarbamate) has been associated with increased incidence of Parkinson's disease (PD). We recently demonstrated that the intranasal (i.n.) administration of sodium dimethyldithiocarbamate (NaDMDC, a more soluble salt than ziram) induces PD-like behavioral and neurochemical alterations in mice. We now investigated the putative neuroprotective effects of melatonin on behavioral dificits and neurochemical alterations induced by i.n. NaDMDC. Melatonin treatment (3, 10 or 30 mg/kg, i.p.) was given 1 h before NaDMDC administration (1 mg/nostril) during 4 consecutive days and we evaluated early (up to 7 days) and late (up to 35 days) NaDMDC-induced behavioral and neurochemical alterations. Melatonin treatment protected against early motor and general neurological impairments observed in the open field and neurological score of severity, respectively, and late deficits in rotarod test. Melatonin prevented the NaDMDC-induced alterations in the striatal tyrosine hydroxylase immunocontent. Melatonin also protected against increased levels of oxidative stress markers (4-hydroxynonenal and 3-nitrotyrosine) in the striatum, as well as the NaDMDC-induced increase of 4-hydroxynonenal and TNF, markers of oxidative stress and inflammation, respectively, in the olfactory bulb. These results further detail the mechanisms underlying NaDMDC toxicity and demonstrate the neuroprotective effects of melatonin against the neuronal damage induced by NaDMDC.
Collapse
|
28
|
Gallegos CE, Bartos M, Gumilar F, Raisman-Vozari R, Minetti A, Baier CJ. Intranasal glyphosate-based herbicide administration alters the redox balance and the cholinergic system in the mouse brain. Neurotoxicology 2020; 77:205-215. [PMID: 31991143 DOI: 10.1016/j.neuro.2020.01.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 01/01/2023]
Abstract
Pesticide exposure is associated with cognitive and psychomotor disorders. Glyphosate-based herbicides (GlyBH) are among the most used agrochemicals, and inhalation of GlyBH sprays may arise from frequent aerial pulverizations. Previously, we described that intranasal (IN) administration of GlyBH in mice decreases locomotor activity, increases anxiety, and impairs recognition memory. Then, the aim of the present study was to investigate the mechanisms involved in GlyBH neurotoxicity after IN administration. Adult male CF-1 mice were exposed to GlyBH IN administration (equivalent to 50 mg/kg/day of Gly acid, 3 days a week, during 4 weeks). Total thiol content and the activity of the enzymes catalase, acetylcholinesterase and transaminases were evaluated in different brain areas. In addition, markers of the cholinergic and the nigrostriatal pathways, as well as of astrocytes were evaluated by fluorescence microscopy in coronal brain sections. The brain areas chosen for analysis were those seen to be affected in our previous study. GlyBH IN administration impaired the redox balance of the brain and modified the activities of enzymes involved in cholinergic and glutamatergic pathways. Moreover, GlyBH treatment decreased the number of cholinergic neurons in the medial septum as well as the expression of the α7-acetylcholine receptor in the hippocampus. Also, the number of astrocytes increased in the anterior olfactory nucleus of the exposed mice. Taken together, these disturbances may contribute to the neurobehavioural impairments reported previously by us after IN GlyBH administration in mice.
Collapse
Affiliation(s)
- Cristina Eugenia Gallegos
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Universidad Nacional del Sur-CONICET, San Juan 670, 8000 Bahía Blanca, Buenos Aires, Argentina
| | - Mariana Bartos
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Universidad Nacional del Sur-CONICET, San Juan 670, 8000 Bahía Blanca, Buenos Aires, Argentina
| | - Fernanda Gumilar
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Universidad Nacional del Sur-CONICET, San Juan 670, 8000 Bahía Blanca, Buenos Aires, Argentina
| | - Rita Raisman-Vozari
- INSERM UMR 1127, CNRS UMR 7225, UPMC, ThérapeutiqueExpérimentale de la Neurodégénérescence, Hôpital de la Salpetrière-ICM (Institut du cerveau et de la moelleépinière), Paris, France
| | - Alejandra Minetti
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Universidad Nacional del Sur-CONICET, San Juan 670, 8000 Bahía Blanca, Buenos Aires, Argentina
| | - Carlos Javier Baier
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Universidad Nacional del Sur-CONICET, San Juan 670, 8000 Bahía Blanca, Buenos Aires, Argentina.
| |
Collapse
|
29
|
Wang Y, Feng L, Liu S, Zhou X, Yin T, Liu Z, Yang Z. Transcranial Magneto-Acoustic Stimulation Improves Neuroplasticity in Hippocampus of Parkinson's Disease Model Mice. Neurotherapeutics 2019; 16:1210-1224. [PMID: 30993592 PMCID: PMC6985386 DOI: 10.1007/s13311-019-00732-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
In this study, we have, for the first time, demonstrated the beneficial effects of transcranial magneto-acoustic stimulation (TMAS), a technique based on focused ultrasound stimulation within static magnetic field, on the learning and memory abilities and neuroplasticity of the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease (PD). Our results showed that chronic TMAS treatment (2 weeks) improved the outcome of Morris water maze, long-term potentiation (LTP), and dendritic spine densities in the dentate gyrus (DG) region of the hippocampus of PD model mice. To further investigate into the underlying mechanisms of these beneficial effects by TMAS, we quantified the proteins in the hippocampus that regulated neuroplasticity. Results showed that the level of postsynaptic density protein 95 was elevated in the brain of TMAS-treated PD model mice while the level of synaptophysin (SYP) did not show any change. We further quantified proteins that mediated neuroplasticity mechanisms, such as brain-derived neurotrophic factor (BDNF) and other important proteins that mediated neuroplasticity. Results showed that TMAS treatment elevated the levels of BDNF, cAMP response element-binding protein (CREB), and protein kinase B (p-Akt) in the PD model mouse hippocampus, but not in the non-PD mouse hippocampus. These results suggest that the beneficial effects on the neuroplasticity of PD model mice treated with TMAS could possibly be conducted through postsynaptic regulations and mediated by BDNF.
Collapse
Affiliation(s)
- Yuexiang Wang
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University, Tianjin, 300071, China
| | - Lina Feng
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University, Tianjin, 300071, China
| | - Shikun Liu
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Xiaoqing Zhou
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Tao Yin
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Zhipeng Liu
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China.
| | - Zhuo Yang
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
30
|
Samal J, Rebelo AL, Pandit A. A window into the brain: Tools to assess pre-clinical efficacy of biomaterials-based therapies on central nervous system disorders. Adv Drug Deliv Rev 2019; 148:68-145. [PMID: 30710594 DOI: 10.1016/j.addr.2019.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/04/2019] [Accepted: 01/28/2019] [Indexed: 12/13/2022]
Abstract
Therapeutic conveyance into the brain is a cardinal requirement for treatment of diverse central nervous system (CNS) disorders and associated pathophysiology. Effectual shielding of the brain by the blood-brain barrier (BBB) sieves out major proportion of therapeutics with the exception of small lipophilic molecules. Various nano-delivery systems (NDS) provide an effective solution around this obstacle owing to their small size and targeting properties. To date, these systems have been used for several pre-clinical disease models including glioma, neurodegenerative diseases and psychotic disorders. An efficacy screen for these systems involves a test battery designed to probe into the multiple facets of therapeutic delivery. Despite their wide application in redressing various disease targets, the efficacy evaluation strategies for all can be broadly grouped into four modalities, namely: histological, bio-imaging, molecular and behavioural. This review presents a comprehensive insight into all of these modalities along with their strengths and weaknesses as well as perspectives on an ideal design for a panel of tests to screen brain nano-delivery systems.
Collapse
Affiliation(s)
- Juhi Samal
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Ana Lucia Rebelo
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
31
|
Paul G, Sullivan AM. Trophic factors for Parkinson's disease: Where are we and where do we go from here? Eur J Neurosci 2019; 49:440-452. [DOI: 10.1111/ejn.14102] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/25/2018] [Accepted: 07/22/2018] [Indexed: 01/15/2023]
Affiliation(s)
- Gesine Paul
- Translational Neurology GroupDepartment of Clinical ScienceLund University Lund Sweden
- Wallenberg Center for Molecular MedicineLund University Lund Sweden
- Department of NeurologyScania University Hospital Lund Sweden
| | - Aideen M. Sullivan
- Department of Anatomy and NeuroscienceUniversity College Cork Cork Ireland
| |
Collapse
|
32
|
Prediger RD, Schamne MG, Sampaio TB, Moreira ELG, Rial D. Animal models of olfactory dysfunction in neurodegenerative diseases. HANDBOOK OF CLINICAL NEUROLOGY 2019; 164:431-452. [PMID: 31604561 DOI: 10.1016/b978-0-444-63855-7.00024-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Olfactory dysfunction seems to occur earlier than classic motor and cognitive symptoms in many neurodegenerative diseases, including Parkinson's disease (PD) and Alzheimer's disease (AD). Thus, the use of the olfactory system as a clinical marker for neurodegenerative diseases is helpful in the characterization of prodromal stages of these diseases, early diagnostic strategies, differential diagnosis, and, potentially, prediction of treatment success. The use of genetic and neurotoxin animal models has contributed to the understanding of the mechanisms underlying olfactory dysfunction in a number of neurodegenerative diseases. In this chapter, we provide an overview of behavioral and neurochemical alterations observed in animal models of different neurodegenerative diseases (such as genetic and Aβ infusion models for AD and neurotoxins and genetic models of PD), in which olfactory dysfunction has been described.
Collapse
Affiliation(s)
- Rui D Prediger
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil.
| | - Marissa G Schamne
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Tuane B Sampaio
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Eduardo L G Moreira
- Department of Physiological Sciences, Center of Biological Sciences¸ Federal University of Santa Catarina, Florianópolis, Brazil
| | - Daniel Rial
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
33
|
Grandi LC, Di Giovanni G, Galati S. Reprint of “Animal models of early-stage Parkinson's disease and acute dopamine deficiency to study compensatory neurodegenerative mechanisms”. J Neurosci Methods 2018; 310:75-88. [DOI: 10.1016/j.jneumeth.2018.10.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/06/2018] [Accepted: 08/09/2018] [Indexed: 12/19/2022]
|
34
|
Deltamethrin Intranasal administration induces memory, emotional and tyrosine hydroxylase immunoreactivity alterations in rats. Brain Res Bull 2018; 142:297-303. [DOI: 10.1016/j.brainresbull.2018.08.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 07/24/2018] [Accepted: 08/11/2018] [Indexed: 02/04/2023]
|
35
|
Grandi LC, Di Giovanni G, Galati S. Animal models of early-stage Parkinson's disease and acute dopamine deficiency to study compensatory neurodegenerative mechanisms. J Neurosci Methods 2018; 308:205-218. [PMID: 30107207 DOI: 10.1016/j.jneumeth.2018.08.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/06/2018] [Accepted: 08/09/2018] [Indexed: 12/21/2022]
Abstract
Parkinson's disease is a common neurodegenerative disease characterized by a widely variety of motor and non-motor symptoms. While the motor deficits are only visible following a severe dopamine depletion, neurodegenerative process and some non-motor symptoms are manifested years before the motor deficits. Importantly, chronic degeneration of dopaminergic neurons leads to the development of compensatory mechanisms that play roles in the progression of the disease and the response to anti-parkinsonian therapies. The identification of these mechanisms will be of great importance for improving our understanding of factors with important contributions to the disease course and the underlying adaptive process. To date, most of the data obtained from animal models reflect the late, chronic, dopamine-depleted states, when compensatory mechanisms have already been established. Thus, adequate animal models with which researchers are able to dissect early- and late-phase mechanisms are necessary. Here, we reviewed the literature related to animal models of early-stage PD and pharmacological treatments capable of inducing acute dopamine impairments and/or depletion, such as reserpine, haloperidol and tetrodotoxin. We highlighted the advantages, limitations and the future prospective uses of these models, as well as their applications in the identification of novel agents for treating this neurological disorder.
Collapse
Affiliation(s)
- Laura Clara Grandi
- Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Switzerland
| | - Giuseppe Di Giovanni
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Malta; Neuroscience Division, School of Biosciences, Cardiff University, Cardiff, UK.
| | - Salvatore Galati
- Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Switzerland.
| |
Collapse
|
36
|
Biju KC, Evans RC, Shrestha K, Carlisle DCB, Gelfond J, Clark RA. Methylene Blue Ameliorates Olfactory Dysfunction and Motor Deficits in a Chronic MPTP/Probenecid Mouse Model of Parkinson's Disease. Neuroscience 2018; 380:111-122. [PMID: 29684508 DOI: 10.1016/j.neuroscience.2018.04.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/03/2018] [Accepted: 04/09/2018] [Indexed: 12/30/2022]
Abstract
Mitochondrial dysfunction and oxidative stress are very prominent and early features in Parkinson's disease (PD) and in animal models of PD. Thus, antioxidant therapy for PD has been proposed, but in clinical trials such strategies have met with very limited success. Methylene blue (MB), a small-molecule synthetic heterocyclic organic compound that acts as a renewable electron cycler in the mitochondrial electron transport chain, manifesting robust antioxidant and cell energetics-enhancing properties, has recently been shown to have significant beneficial effects in reducing nigrostriatal dopaminergic loss and motor impairment in acute toxin models of PD. However, no studies have investigated the impact of this promising agent in chronic models or for olfactory dysfunction, an early non-motor feature of PD. To test the efficacy of low-dose MB for olfactory dysfunction, motor symptoms, and dopaminergic neurodegeneration, mice were injected with ten subcutaneous doses of 25 mg/kg MPTP, plus 250 mg/kg intraperitoneal probenecid or saline/probenecid at 3.5-day intervals. Following the onset of olfactory dysfunction, MPTP/probenecid (MPTP/p) and saline/probenecid mice were provided drinking water with or without 1 mg/kg/day MB. Oral delivery of low-dose MB significantly ameliorated MPTP/p-induced deficits in motor coordination, as well as degeneration of tyrosine hydroxylase (TH)-positive neurons of the substantia nigra and TH-positive terminals in the striatum. Importantly, olfactory dysfunction was ameliorated by MB treatment, whereas this benefit is not observed with currently available anti-Parkinsonian medications. These results indicate that low-dose MB is a promising neuroprotective intervention for both motor and non-motor features of PD.
Collapse
Affiliation(s)
- K C Biju
- Department of Medicine, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Robert C Evans
- Department of Medicine, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Kripa Shrestha
- Department of Medicine, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Daniel C B Carlisle
- Department of Medicine, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Jonathan Gelfond
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Robert A Clark
- Department of Medicine, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States; South Texas Veterans Health Care System, 7400 Merton Minter Blvd, San Antonio, TX 78229, United States.
| |
Collapse
|
37
|
The Gender-Biased Effects of Intranasal MPTP Administration on Anhedonic- and Depressive-Like Behaviors in C57BL/6 Mice: the Role of Neurotrophic Factors. Neurotox Res 2018; 34:808-819. [DOI: 10.1007/s12640-018-9912-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 04/24/2018] [Accepted: 05/16/2018] [Indexed: 01/10/2023]
|
38
|
Crespo C, Liberia T, Blasco-Ibáñez JM, Nácher J, Varea E. Cranial Pair I: The Olfactory Nerve. Anat Rec (Hoboken) 2018; 302:405-427. [PMID: 29659152 DOI: 10.1002/ar.23816] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/29/2017] [Accepted: 06/15/2017] [Indexed: 12/13/2022]
Abstract
The olfactory nerve constitutes the first cranial pair. Compared with other cranial nerves, it depicts some atypical features. First, the olfactory nerve does not form a unique bundle. The olfactory axons join other axons and form several small bundles or fascicles: the fila olfactoria. These fascicles leave the nasal cavity, pass through the lamina cribrosa of the ethmoid bone and enter the brain. The whole of these fascicles is what is known as the olfactory nerve. Second, the olfactory sensory neurons, whose axons integrate the olfactory nerve, connect the nasal cavity and the brain without any relay. Third, the olfactory nerve is composed by unmyelinated axons. Fourth, the olfactory nerve contains neither Schwann cells nor oligodendrocytes wrapping its axons. But it contains olfactory ensheathing glia, which is a type of glia unique to this nerve. Fifth, the olfactory axons participate in the circuitry of certain spherical structures of neuropil that are unique in the brain: the olfactory glomeruli. Sixth, the axons of the olfactory nerve are continuously replaced and their connections in the central nervous system are remodeled continuously. Therefore, the olfactory nerve is subject to lifelong plasticity. Finally seventh, the olfactory nerve can be a gateway for the direct entrance of viruses, neurotoxins and other xenobiotics to the brain. In the same way, it can be used as a portal of entry to the brain for therapeutic substances, bypassing the blood-brain barrier. In this article, we analyze some features of the anatomy and physiology of the first cranial pair. Anat Rec, 302:405-427, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Carlos Crespo
- Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
| | - Teresa Liberia
- Departments of Neurosurgery and Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - José Miguel Blasco-Ibáñez
- Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
| | - Juan Nácher
- Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
| | - Emilio Varea
- Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
| |
Collapse
|
39
|
Mack JM, Moura TM, Lanznaster D, Bobinski F, Massari CM, Sampaio TB, Schmitz AE, Souza LF, Walz R, Tasca CI, Poli A, Doty RL, Dafre AL, Prediger RD. Intranasal administration of sodium dimethyldithiocarbamate induces motor deficits and dopaminergic dysfunction in mice. Neurotoxicology 2018; 66:107-120. [PMID: 29605442 DOI: 10.1016/j.neuro.2018.03.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 01/26/2023]
Abstract
The primary etiology of Parkinson's disease (PD) remains unclear, but likely reflects a combination of genetic and environmental factors. Exposure to some pesticides, including ziram (zinc dimethyldithiocarbamate), is a relevant risk factor for PD. Like some other environmental neurotoxicants, we hypothesized that ziram can enter the central nervous system from the nasal mucosa via the olfactory nerves. To address this issue, we evaluated the effects of 1, 2 or 4 days of intranasal (i.n., 1 mg/nostril/day) infusions of sodium dimethyldithiocarbamate (NaDMDC), a dimethyldithiocarbamate more soluble than ziram, on locomotor activity in the open field, neurological severity score and rotarod performance. We also addressed the effects of four daily i.n. NaDMDC infusions on olfactory bulb (OB) and striatal measures of cell death, reactive oxygen species (ROS), tyrosine hydroxylase, and the levels of dopamine, noradrenaline, serotonin, and their metabolites. A single i.n. administration of NaDMDC did not significantly alter the behavioral measures. Two consecutive days of i.n. NaDMDC administrations led to a transient neurological deficit that spontaneously resolved within a week. However, the i.n. infusions of NaDMDC for 4 consecutive days induced motor and neurological deficits for up to 7 days after the last NaDMDC administration and increased striatal TH immunocontent and dopamine degradation within a day of the last infusion. Pharmacological treatment with the anti-parkinsonian drugs l-DOPA and apomorphine improved the NaDMDC-induced locomotor deficits. NaDMDC increased serotonin levels and noradrenaline metabolism in the OB 24 h after the last NaDMDC infusion, ROS levels in the OB 2 h after the last infusion, and striatum 2 and 24 h after the last infusion. These results demonstrate, for the first time, that i.n. NaDMDC administration induces neurobehavioral and neurochemical impairments in mice. This accords with evidence that dimethyldithio-carbamate exposure increases the risk of PD and highlights the possibility that olfactory system could be a major route for NaDMDC entry to central nervous system.
Collapse
Affiliation(s)
- Josiel M Mack
- Department of Pharmacology, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Tainara M Moura
- Department of Pharmacology, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Débora Lanznaster
- Department of Biochemistry, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Franciane Bobinski
- Experimental Neuroscience Laboratory (LANEX), Graduate Program in Health Sciences, University of Southern of Santa Catarina (UNISUL), Palhoça, SC, Brazil
| | - Caio M Massari
- Department of Biochemistry, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Tuane B Sampaio
- Department of Pharmacology, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Ariana E Schmitz
- Department of Biochemistry, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Luiz F Souza
- Department of Biochemistry, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Roger Walz
- Department of Clinical Medical, Center of Health Sciences, University Hospital, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Carla I Tasca
- Department of Biochemistry, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Anicleto Poli
- Department of Pharmacology, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Richard L Doty
- Smell & Taste Center, Department of Otorhinolaryngology: Head and Neck Surgery, Perelman School of Medicine, Philadelphia, PA 19104 USA
| | - Alcir L Dafre
- Department of Biochemistry, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Rui D Prediger
- Department of Pharmacology, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil.
| |
Collapse
|
40
|
Whole-Transcriptome Analysis of Mouse Models with MPTP-Induced Early Stages of Parkinson’s Disease Reveals Stage-Specific Response of Transcriptome and a Possible Role of Myelin-Linked Genes in Neurodegeneration. Mol Neurobiol 2018; 55:7229-7241. [DOI: 10.1007/s12035-018-0907-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/11/2018] [Indexed: 01/19/2023]
|
41
|
Marques NF, Castro AA, Mancini G, Rocha FL, Santos ARS, Prediger RD, De Bem AF, Tasca CI. Atorvastatin Prevents Early Oxidative Events and Modulates Inflammatory Mediators in the Striatum Following Intranasal 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) Administration in Rats. Neurotox Res 2017; 33:549-559. [DOI: 10.1007/s12640-017-9840-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/20/2017] [Accepted: 11/03/2017] [Indexed: 10/18/2022]
|
42
|
Behavioral impairments following repeated intranasal glyphosate-based herbicide administration in mice. Neurotoxicol Teratol 2017; 64:63-72. [DOI: 10.1016/j.ntt.2017.10.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/12/2017] [Accepted: 10/18/2017] [Indexed: 12/31/2022]
|
43
|
Titova N, Schapira AHV, Chaudhuri KR, Qamar MA, Katunina E, Jenner P. Nonmotor Symptoms in Experimental Models of Parkinson's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 133:63-89. [PMID: 28802936 DOI: 10.1016/bs.irn.2017.05.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nonmotor symptoms of Parkinson's disease (PD) range from neuropsychiatric, cognitive to sleep and sensory disorders and can arise from the disease process as well as from drug treatment. The clinical heterogeneity of nonmotor symptoms of PD is underpinned by a wide range of neuropathological and molecular pathology, affecting almost the entire range of neurotransmitters present in brain and the periphery. Understanding the neurobiology and pathology of nonmotor symptoms is crucial to the effective treatment of PD and currently a key unmet need. This bench-to-bedside translational concept can only be successful if robust animal models of PD charting the genesis and natural history of nonmotor symptoms can be devised. Toxin-based and transgenic rodent and primate models of PD have given us important clues to the underlying basis of motor symptomatology and in addition, can provide a snapshot of some nonmotor aspects of PD, although the data are far from complete. In this chapter, we discuss some of the nonmotor aspects of the available experimental models of PD and how the development of robust animal models to understand and treat nonmotor symptoms needs to become a research priority.
Collapse
Affiliation(s)
- Nataliya Titova
- Federal State Budgetary Educational Institution of Higher Education "N.I. Pirogov Russian National Research Medical University" of the Ministry of Healthcare of the Russian Federation, Moscow, Russia.
| | | | - K Ray Chaudhuri
- National Parkinson Foundation International Centre of Excellence, King's College London and King's College Hospital, London, United Kingdom; The Maurice Wohl Clinical Neuroscience Institute, King's College London, National Institute for Health Research (NIHR) South London and Maudsley NHS Foundation Trust and King's College London, London, United Kingdom
| | - Mubasher A Qamar
- National Parkinson Foundation International Centre of Excellence, King's College London and King's College Hospital, London, United Kingdom; The Maurice Wohl Clinical Neuroscience Institute, King's College London, National Institute for Health Research (NIHR) South London and Maudsley NHS Foundation Trust and King's College London, London, United Kingdom
| | | | - Peter Jenner
- Neurodegenerative Diseases Research Group, Institute of Pharmaceutical Science, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| |
Collapse
|
44
|
Fortuna JTS, Gralle M, Beckman D, Neves FS, Diniz LP, Frost PS, Barros-Aragão F, Santos LE, Gonçalves RA, Romão L, Zamberlan DC, Soares FAA, Braga C, Foguel D, Gomes FCA, De Felice FG, Ferreira ST, Clarke JR, Figueiredo CP. Brain infusion of α-synuclein oligomers induces motor and non-motor Parkinson's disease-like symptoms in mice. Behav Brain Res 2017; 333:150-160. [PMID: 28668282 DOI: 10.1016/j.bbr.2017.06.047] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/16/2017] [Accepted: 06/27/2017] [Indexed: 01/05/2023]
Abstract
Parkinson's disease (PD) is characterized by motor dysfunction, which is preceded by a number of non-motor symptoms including olfactory deficits. Aggregation of α-synuclein (α-syn) gives rise to Lewy bodies in dopaminergic neurons and is thought to play a central role in PD pathology. However, whether amyloid fibrils or soluble oligomers of α-syn are the main neurotoxic species in PD remains controversial. Here, we performed a single intracerebroventricular (i.c.v.) infusion of α-syn oligomers (α-SYOs) in mice and evaluated motor and non-motor symptoms. Familiar bedding and vanillin essence discrimination tasks showed that α-SYOs impaired olfactory performance of mice, and decreased TH and dopamine levels in the olfactory bulb early after infusion. The olfactory deficit persisted until 45days post-infusion (dpi). α- SYO-infused mice behaved normally in the object recognition and forced swim tests, but showed increased anxiety-like behavior in the open field and elevated plus maze tests 20 dpi. Finally, administration of α-SYOs induced late motor impairment in the pole test and rotarod paradigms, along with reduced TH and dopamine content in the caudate putamen, 45 dpi. Reduced number of TH-positive cells was also seen in the substantia nigra of α-SYO-injected mice compared to control. In conclusion, i.c.v. infusion of α-SYOs recapitulated some of PD-associated non-motor symptoms, such as increased anxiety and olfactory dysfunction, but failed to recapitulate memory impairment and depressive-like behavior typical of the disease. Moreover, α-SYOs i.c.v. administration induced motor deficits and loss of TH and dopamine levels, key features of PD. Results point to α-syn oligomers as the proximal neurotoxins responsible for early non-motor and motor deficits in PD and suggest that the i.c.v. infusion model characterized here may comprise a useful tool for identification of PD novel therapeutic targets and drug screening.
Collapse
Affiliation(s)
- Juliana T S Fortuna
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Matthias Gralle
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Danielle Beckman
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Fernanda S Neves
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Luan P Diniz
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Paula S Frost
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Fernanda Barros-Aragão
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Luís E Santos
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Rafaella A Gonçalves
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Luciana Romão
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Campus Xerém, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Daniele C Zamberlan
- Department of Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Felix A A Soares
- Department of Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Carolina Braga
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Campus Xerém, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Debora Foguel
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Flávia C A Gomes
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Julia R Clarke
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil.
| | - Cláudia P Figueiredo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil.
| |
Collapse
|
45
|
Schapira AHV, Chaudhuri KR, Jenner P. Non-motor features of Parkinson disease. Nat Rev Neurosci 2017; 18:435-450. [PMID: 28592904 DOI: 10.1038/nrn.2017.62] [Citation(s) in RCA: 1179] [Impact Index Per Article: 147.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Many of the motor symptoms of Parkinson disease (PD) can be preceded, sometimes for several years, by non-motor symptoms that include hyposmia, sleep disorders, depression and constipation. These non-motor features appear across the spectrum of patients with PD, including individuals with genetic causes of PD. The neuroanatomical and neuropharmacological bases of non-motor abnormalities in PD remain largely undefined. Here, we discuss recent advances that have helped to establish the presence, severity and effect on the quality of life of non-motor symptoms in PD, and the neuroanatomical and neuropharmacological mechanisms involved. We also discuss the potential for the non-motor features to define a prodrome that may enable the early diagnosis of PD.
Collapse
Affiliation(s)
- Anthony H V Schapira
- Department of Clinical Neurosciences, University College London (UCL) Institute of Neurology, Royal Free Campus, Rowland Hill Street, London NW3 2PF, UK
| | - K Ray Chaudhuri
- National Parkinson Foundation International Centre of Excellence, King's College Hospital, King's College London, Camberwell Road, London SE5 9RS, UK
| | - Peter Jenner
- Neurodegenerative Diseases Research Group, Institute of Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, Newcomen Street, London SE1 1UL, UK
| |
Collapse
|
46
|
Blaylock RL. Parkinson's disease: Microglial/macrophage-induced immunoexcitotoxicity as a central mechanism of neurodegeneration. Surg Neurol Int 2017; 8:65. [PMID: 28540131 PMCID: PMC5421223 DOI: 10.4103/sni.sni_441_16] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 02/01/2017] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease is one of the several neurodegenerative disorders that affects aging individuals, with approximately 1% of those over the age of 60 years developing the disorder in their lifetime. The disease has the characteristics of a progressive disorder in most people, with a common pattern of pathological change occurring in the nervous system that extends beyond the classical striatal degeneration of dopaminergic neurons. Earlier studies concluded that the disease was a disorder of alpha-synuclein, with the formation of aggregates of abnormal alpha-synuclein being characteristic. More recent studies have concluded that inflammation plays a central role in the disorder and that the characteristic findings can be accounted for by either mutation or oxidative damage to alpha-synuclein, with resulting immune reactions from surrounding microglia, astrocytes, and macrophages. What has been all but ignored in most of these studies is the role played by excitotoxicity and that the two processes are intimately linked, with inflammation triggered cell signaling enhancing the excitotoxic cascade. Further, there is growing evidence that it is the excitotoxic reactions that actually cause the neurodegeneration. I have coined the name immunoexcitotoxicity to describe this link between inflammation and excitotoxicity. It appears that the two processes are rarely, if ever, separated in neurodegenerative diseases.
Collapse
|
47
|
de Oliveira PA, Ben J, Matheus FC, Schwarzbold ML, Moreira ELG, Rial D, Walz R, Prediger RD. Moderate traumatic brain injury increases the vulnerability to neurotoxicity induced by systemic administration of 6-hydroxydopamine in mice. Brain Res 2017; 1663:78-86. [PMID: 28288867 DOI: 10.1016/j.brainres.2017.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 02/06/2017] [Accepted: 03/02/2017] [Indexed: 11/30/2022]
Abstract
Moderate traumatic brain injury (TBI) might increase the vulnerability to neuronal neurodegeneration, but the basis of such selective neuronal susceptibility has remained elusive. In keeping with the disruption of the blood-brain barrier (BBB) caused by TBI, changes in BBB permeability following brain injury could facilitate the access of xenobiotics into the brain. To test this hypothesis, here we evaluated whether TBI would increase the susceptibility of nigrostriatal dopaminergic fibers to the systemic administration of 6-hydroxydopamine (6-OHDA), a classic neurotoxin used to trigger a PD-like phenotype in mice, but that in normal conditions is unable to cross the BBB. Adult Swiss mice were submitted to a moderate TBI using a free weight-drop device and, 5h later, they were injected intraperitoneally with a single dose of 6-OHDA (100mg/kg). Afterwards, during a period of 4weeks, the mice were submitted to a battery of behavioral tests, including the neurological severity score (NSS), the open field and the rotarod. Animals from the TBI plus 6-OHDA group displayed significant motor and neurological impairments that were improved by acute l-DOPA administration (25mg/kg, i.p.). Moreover, the observation of the motor deficits correlates with (i) a significant decrease in the tyrosine hydroxylase levels mainly in the rostral striatum and (ii) a significant increase in the levels of striatal glial fibrillary acidic protein (GFAP) levels. On the whole, the present findings demonstrate that a previous moderate TBI event increases the susceptibility to motor, neurological and neurochemical alterations induced by systemic administration of the dopaminergic neurotoxin 6-OHDA in mice.
Collapse
Affiliation(s)
| | - Juliana Ben
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Brazil; Programa de Pós-Graduação em Neurociências, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Brazil
| | - Filipe Carvalho Matheus
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Brazil
| | - Marcelo Liborio Schwarzbold
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Brazil; Centro de Neurociências Aplicadas (CeNAp), Departamento de Clínica Médica, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Brazil
| | - Eduardo Luiz Gasnhar Moreira
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Brazil; Departamento de Ciências Fisiológicas, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Brazil
| | - Daniel Rial
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Brazil
| | - Roger Walz
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Brazil; Centro de Neurociências Aplicadas (CeNAp), Departamento de Clínica Médica, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Brazil
| | - Rui Daniel Prediger
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Brazil; Programa de Pós-Graduação em Neurociências, Universidade Federal de Santa Catarina, Florianópolis 88049-900, Brazil.
| |
Collapse
|
48
|
Gonçalves N, Simões AT, Prediger RD, Hirai H, Cunha RA, Pereira de Almeida L. Caffeine alleviates progressive motor deficits in a transgenic mouse model of spinocerebellar ataxia. Ann Neurol 2017; 81:407-418. [DOI: 10.1002/ana.24867] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 12/12/2016] [Accepted: 12/18/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Nélio Gonçalves
- Center for Neuroscience and Cell Biology; University of Coimbra; Coimbra Portugal
- Faculty of Pharmacy; University of Coimbra; Coimbra Portugal
| | - Ana T. Simões
- Center for Neuroscience and Cell Biology; University of Coimbra; Coimbra Portugal
- Faculty of Pharmacy; University of Coimbra; Coimbra Portugal
| | - Rui D. Prediger
- Center for Neuroscience and Cell Biology; University of Coimbra; Coimbra Portugal
- Department of Pharmacology; Federal University of Santa Catarina; Florianópolis, SC Brazil
| | - Hirokazu Hirai
- Department of Neurophysiology; Gunma University Graduate School of Medicine; Maebashi Gunma Japan
| | - Rodrigo A. Cunha
- Center for Neuroscience and Cell Biology; University of Coimbra; Coimbra Portugal
- Faculty of Medicine; University of Coimbra; Coimbra Portugal
| | - Luís Pereira de Almeida
- Center for Neuroscience and Cell Biology; University of Coimbra; Coimbra Portugal
- Faculty of Pharmacy; University of Coimbra; Coimbra Portugal
| |
Collapse
|
49
|
Ekimova IV, Simonova VV, Guzeev MA, Lapshina KV, Chernyshev MV, Pastukhov YF. Changes in sleep characteristics of rat preclinical model of Parkinson’s disease based on attenuation of the ubiquitin—proteasome system activity in the brain. J EVOL BIOCHEM PHYS+ 2017. [DOI: 10.1134/s1234567816060057] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
50
|
Transcriptome Profile Changes in Mice with MPTP-Induced Early Stages of Parkinson's Disease. Mol Neurobiol 2016; 54:6775-6784. [PMID: 27757834 DOI: 10.1007/s12035-016-0190-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/30/2016] [Indexed: 01/26/2023]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. Despite progress in the study of the molecular, genetic, and pathogenic mechanisms of PD, it is unclear which processes trigger the development of the pathology associated with PD. Models of the presymptomatic and early symptomatic stages of PD induced by MPTP have been used to analyze changes in transcriptome profile in brain tissues, to identify specific patterns and mechanisms underlying neurodegeneration in PD. The whole-transcriptome analysis in the brain tissues of the mice with MPTP-induced PD showed that striatum is involved in the pathogenesis in the earliest stages and the processes associated with vesicular transport may be altered. The expression profiles of the genes studied in the substantia nigra and peripheral blood confirm that lymphocytes from peripheral blood may reflect processes occurring in the brain. These data suggest that messenger RNA (mRNA) levels in peripheral blood may provide potential biomarkers of the neurodegeneration occurring in PD. The changes in expression at the mRNA and protein levels suggest that Snca may be involved in neurodegeneration and Drd2 may participate in the development of the compensatory mechanisms in the early stages of PD pathogenesis. Our data suggest that the brain cortex may be involved in the pathological processes in the early stages of PD, including the presymptomatic stage.
Collapse
|