1
|
Tang X, Xue J, Chen R, Xing J, Lu X, Cui L. Behavioral, biochemical, and molecular characterization of MPTP/p-intoxicated mice. Exp Neurol 2025; 386:115168. [PMID: 39884331 DOI: 10.1016/j.expneurol.2025.115168] [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: 11/11/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model remains the most extensively utilized animal model for Parkinson's disease (PD). Treatment regimens are classified into three categories: acute, subacute, and chronic. Among these, the MPTP with probenecid (MPTP/p)-induced chronic mouse model is favored for its capacity to sustain long-term striatal dopamine depletion, though the resultant behavioral, biochemical, and molecular alterations require further validation. To systematically evaluate these abnormalities in the chronic MPTP/p mouse model, we conducted observations over a 12-month period. The results showed that these mice displayed reduced locomotor activity, minor gait abnormalities, and anxiety-like behavior within one week following the final MPTP/p injection. No significant motor disorders were observed from 1 to 12 months post-final injection, with exception of increased exploratory activity in the elevated plus maze from 2 to 8 months. One month after the final MPTP/p injection, there was a significant decrease in dopaminergic neurons in the ventral midbrain, which partially recovered after 12 months. A single MPTP/p injection temporarily increased striatal DA and its metabolites. Proteomics of ventral midbrain tissue indicated that the recovery of dopaminergic neurons might be linked to the upregulation of proteins like Bone morphogenetic protein type II receptor (Bmpr2) and Glutathione S-transferase mu 2 (Gstm2) once MPTP toxicity was removed. Our study indicated that the optimal time to evaluate behavioral abnormalities in chronic MPTP/p mouse model is within one week after modeling. Moreover, the upregulated expression of related proteins, such as Bmpr2 and Gstm2, in the ventral midbrain of the model mice after modeling may represent important targets for the future treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Xiaolu Tang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Department of Human Anatomy, School of Basic Medical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Jinhua Xue
- Department of Pathophysiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Rui Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Jiawei Xing
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Xiaying Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou 341000, China.
| | - Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; The Marine Biomedical Research Institute of Guangdong, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524000, China.
| |
Collapse
|
2
|
Liu Q, Huang B, Guiberson NGL, Chen S, Zhu D, Ma G, Ma XM, Crittenden JR, Yu J, Graybiel AM, Dawson TM, Dawson VL, Xiong Y. CalDAG-GEFI acts as a guanine nucleotide exchange factor for LRRK2 to regulate LRRK2 function and neurodegeneration. SCIENCE ADVANCES 2024; 10:eadn5417. [PMID: 39576856 PMCID: PMC11584015 DOI: 10.1126/sciadv.adn5417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 10/22/2024] [Indexed: 11/24/2024]
Abstract
Mutations in LRRK2 are the most common genetic cause of Parkinson's disease (PD). LRRK2 protein contains two enzymatic domains: a GTPase (Roc-COR) and a kinase domain. Disease-causing mutations are found in both domains. Now, studies have focused largely on LRRK2 kinase activity, while attention to its GTPase function is limited. LRRK2 is a guanine nucleotide-binding protein, but the mechanism of direct regulation of its GTPase activity remains unclear and its physiological GEF is not known. Here, we identified CalDAG-GEFI (CDGI) as a physiological GEF for LRRK2. CDGI interacts with LRRK2 and increases its GDP to GTP exchange activity. CDGI modulates LRRK2 cellular functions and LRRK2-induced neurodegeneration in both LRRK2 Drosophila and mouse models. Together, this study identified the physiological GEF for LRRK2 and provides strong evidence that LRRK2 GTPase is regulated by GAPs and GEFs. The LRRK2 GTPase, GAP, or GEF activities have the potential to serve as therapeutic targets, which is distinct from the direct LRRK2 kinase inhibition.
Collapse
Affiliation(s)
- Qinfang Liu
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Bingxu Huang
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Noah Guy Lewis Guiberson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shifan Chen
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Dong Zhu
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Gang Ma
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | - Xin-Ming Ma
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Jill R. Crittenden
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jianzhong Yu
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | - Ann M. Graybiel
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ted M. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130, USA
| | - Valina L. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yulan Xiong
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| |
Collapse
|
3
|
Serra M, Faustini G, Brembati V, Casu MA, Pizzi M, Morelli M, Pinna A, Bellucci A. Early α-synuclein/synapsin III co-accumulation, nigrostriatal dopaminergic synaptopathy and denervation in the MPTPp mouse model of Parkinson's Disease. Exp Neurol 2024; 383:115040. [PMID: 39500391 DOI: 10.1016/j.expneurol.2024.115040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/10/2024]
Abstract
Parkinson's disease (PD) is characterized by the loss of nigrostriatal dopaminergic neurons and the presence of Lewy bodies (LB), intraneuronal inclusions mainly composed of α-synuclein (α-Syn) fibrils. Compelling evidence supports that, in PD brains, synapses are the sites where neurodegeneration initiates several years before the manifestation of motor symptoms. Furthermore, the amount of α-Syn deposited at synaptic terminals is several orders greater than that constituting LB. This hints that pathological synaptic α-Syn aggregates may be the main trigger for the retrograde synapse-to-cell body degeneration pattern characterizing early prodromal phases of PD. Identifying reliable biomarkers of synaptopathy is therefore crucial for early diagnosis. Here, we studied the alterations of key dopaminergic and non-dopaminergic striatal synaptic markers during the initial phases of axonal and cell body degeneration in mice subjected to 3 or 10 administrations of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine + probenecid (MPTPp), a model for early prodromal PD. We found that MPTPp administration resulted in progressive deposition of α-Syn, advancing from synaptic terminals to axons and dopaminergic neuron cell bodies. This was accompanied by marked co-accumulation of Synapsin III (Syn III), a synaptic protein previously identified as a component of α-Syn fibrils in post-mortem PD brains and as a main stabilizer of α-Syn aggregates, as well as very early and severe reduction of vesicular monoamine transporter 2 (VMAT2), dopamine transporter (DAT) and tyrosine hydroxylase (TH) immunoreactivity in nigrostriatal neurons. Results also showed that striatal α-Syn accumulation and VMAT2 decrease, unlike other markers, did not recover following washout from 10 MPTPp administrations, supporting that these changes were precocious and severe. Finally, we found that early changes in striatal α-Syn, Syn III, VMAT2 and DAT observed following 3 MPTPp administrations, correlated with nigrostriatal neuron loss after 10 MPTPp administrations. These findings indicate that α-Syn/Syn III co-deposition characterizes very early stages of striatal dopaminergic dysfunction in the MPTPp model and highlight that VMAT2 and Syn III could be two reliable molecular imaging biomarkers to predict dopamine neuron denervation and estimate α-Syn-related synaptopathy in prodromal and early symptomatic phases of PD.
Collapse
Affiliation(s)
- Marcello Serra
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Gaia Faustini
- Department of Molecular and Translational Medicine University of Brescia, Brescia, Italy
| | - Viviana Brembati
- Department of Molecular and Translational Medicine University of Brescia, Brescia, Italy
| | - Maria Antonietta Casu
- National Research Council of Italy, Institute of Translational Pharmacology, UOS of Cagliari, Scientific and Technological Park of Sardinia POLARIS, Pula, Italy
| | - Marina Pizzi
- Department of Molecular and Translational Medicine University of Brescia, Brescia, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy; National Research Council of Italy, Neuroscience Institute, UOS of Cagliari, Italy
| | - Annalisa Pinna
- National Research Council of Italy, Neuroscience Institute, UOS of Cagliari, Italy.
| | - Arianna Bellucci
- Department of Molecular and Translational Medicine University of Brescia, Brescia, Italy.
| |
Collapse
|
4
|
Chatterjee A, Mohapatra J, Sharma M, Jha A, Patro R, Das D, Patel H, Patel H, Chaudhari J, Borda N, Viswanathan K, Sharma B, Bhavsar H, Patel A, Ranvir R, Sundar R, Agarwal S, Jain M. A novel selective NLRP3 inhibitor shows disease-modifying potential in animal models of Parkinson's disease. Brain Res 2024; 1842:149129. [PMID: 39074525 DOI: 10.1016/j.brainres.2024.149129] [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: 01/27/2024] [Revised: 05/23/2024] [Accepted: 07/20/2024] [Indexed: 07/31/2024]
Abstract
Pathological activation of the Nod-like receptor family pyrin domain containing protein 3 (NLRP3) inflammasome signaling underlies many autoimmune and neuroinflammatory conditions. Here we report that, a rationally designed, novel, orally active, selective NLRP3 inflammasome inhibitor, Usnoflast (ZYIL1), showed potent inhibition of ATP, Nigericin and monosodium urate-mediated interleukin (IL)-1β release in THP-1 cells and human PBMC. In isolated microglia cells, the IC50 of ZYIL1 mediated inhibition of IL-1β was 43 nM. ZYIL1 displayed good pharmacokinetic profile in mice, rats and primates after oral administration and the concentrations found in the brain and cerebrospinal fluid (CSF) were markedly higher than the IC50 values. In an in vivo model of neuroinflammation, ZYIL1 demonstrated robust suppression of NLRP3 inflammasome activation and IL-1β upon oral administration. This translated into efficacy in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and 6-Hydroxydopamine (6-OHDA)-induced Parkinson's disease (PD) models in mice. In MPTP and/or 6-OHDA models, treatment with ZYIL1 ameliorated motor deficits, degeneration of nigrostriatal dopaminergic neurons and abnormal accumulation of α-synuclein. There were positive changes in the genes related to walking, locomotor activity, neurogenesis, neuroblast proliferation and neuronal differentiation in the PD brain indicating improvement in neural health which translated into improved mobility. These findings clearly indicate that selective NLRP3 inhibitor ZYIL1, ameliorates neuroinflammation and appears to have the potential for disease modification and progression associated with PD.
Collapse
Affiliation(s)
- Abhijit Chatterjee
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India.
| | - Jogeswar Mohapatra
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Manoranjan Sharma
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Abhishek Jha
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Randeep Patro
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Debajeet Das
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Hiren Patel
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Harilal Patel
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Jaimin Chaudhari
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Nilesh Borda
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Kasinath Viswanathan
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Bhavesh Sharma
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Harsh Bhavsar
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Ashvin Patel
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Ramchandra Ranvir
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Rajesh Sundar
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Sameer Agarwal
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| | - Mukul Jain
- Zydus Research Centre, Zydus Lifesciences Limited, Sharkhej-Bavla NH No. 8A, Village Moraiya, Changodar, Ahmedabad 382 213, Gujarat, India
| |
Collapse
|
5
|
Poniatowski ŁA, Joniec-Maciejak I, Wawer A, Sznejder-Pachołek A, Machaj E, Ziętal K, Mirowska-Guzel D. Dose-Ranging Effects of the Intracerebral Administration of Atsttrin in Experimental Model of Parkinson's Disease Induced by 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in Mice. Mol Neurobiol 2024; 61:9432-9458. [PMID: 38642286 PMCID: PMC11496375 DOI: 10.1007/s12035-024-04161-0] [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: 02/03/2024] [Accepted: 04/02/2024] [Indexed: 04/22/2024]
Abstract
Parkinson's disease is one of the most common neurodegenerative disorders characterized by a multitude of motor and non-motor clinical symptoms resulting from the progressive and long-lasting abnormal loss of nigrostriatal dopaminergic neurons. Currently, the available treatments for patients with Parkinson's disease are limited and exert only symptomatic effects, without adequate signs of delaying or stopping the progression of the disease. Atsttrin constitutes the bioengineered protein which ultrastructure is based on the polypeptide chain frame of the progranulin (PGRN), which exerts anti-inflammatory effects through the inhibition of TNFα. The conducted preclinical studies suggest that the therapeutic implementation of Atsttrin may be potentially effective in the treatment of neurodegenerative diseases that are associated with the occurrence of neuroinflammatory processes. The aim of the proposed study was to investigate the effect of direct bilateral intracerebral administration of Atsttrin using stereotactic methods in the preclinical C57BL/6 mouse model of Parkinson's disease inducted by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) intoxication. The analysis of the dose dependency effects of the increasing doses of Atsttrin has covered a number of parameters and markers regarding neurodegenerative processes and inflammatory responses including IL-1α, TNFα, IL-6, TH, and TG2 mRNA expressions. Accordingly, the evaluation of the changes in the neurochemical profile included DA, DOPAC, 3-MT, HVA, NA, MHPG, 5-HT, and 5-HIAA concentration levels. The intracerebral administration of Atsttrin into the striatum effectively attenuated the neuroinflammatory reaction in evaluated neuroanatomical structures. Furthermore, the partial restoration of monoamine content and its metabolic turnover were observed. In this case, taking into account the previously described pharmacokinetic profile and extrapolated bioavailability as well as the stability characteristics of Atsttrin, an attempt was made to describe as precisely as possible the quantitative and qualitative effects of increasing doses of the compound within the brain tissue microenvironment in the presented preclinical model of the disease. Collectively, this findings demonstrated that the intracerebral administration of Atsttrin may represent a potential novel therapeutic method for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Łukasz A Poniatowski
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
- Department of Neurosurgery, Dietrich-Bonhoeffer-Klinikum, Salvador-Allende-Straße 30, 17036, Neubrandenburg, Germany
| | - Ilona Joniec-Maciejak
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland.
| | - Adriana Wawer
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Anna Sznejder-Pachołek
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Ewa Machaj
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Katarzyna Ziętal
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| |
Collapse
|
6
|
He S, Ru Q, Chen L, Xu G, Wu Y. Advances in animal models of Parkinson's disease. Brain Res Bull 2024; 215:111024. [PMID: 38969066 DOI: 10.1016/j.brainresbull.2024.111024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
Parkinson's disease is a complex neurodegenerative disease characterized by progressive movement impairments. Predominant symptoms encompass resting tremor, bradykinesia, limb rigidity, and postural instability. In addition, it also includes a series of non-motor symptoms such as sleep disorders, hyposmia, gastrointestinal dysfunction, autonomic dysfunction and cognitive impairment. Pathologically, the disease manifests through dopaminergic neuronal loss and the presence of Lewy bodies. At present, no significant breakthrough has been achieved in clinical Parkinson's disease treatment. Exploring treatment modalities necessitate the establishment of scientifically sound animal models. In recent years, researchers have focused on replicating the symptoms of human Parkinson's disease, resulting in the establishment of various experimental animal models primarily through drugs and transgenic methods to mimic relevant pathologies and identify more effective treatments. This review examines traditional neurotoxin and transgenic animal models as well as α-synuclein pre-formed fibrils models, non-human primate models and non-mammalian specie models. Additionally, it introduces emerging models, including models based on optogenetics, induced pluripotent stem cells, and gene editing, aiming to provide a reference for the utilization of experimental animal models and clinical research for researchers in this field.
Collapse
Affiliation(s)
- Sui He
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Guodong Xu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
7
|
Semenova EI, Rudenok MM, Rybolovlev IN, Shulskaya MV, Lukashevich MV, Partevian SA, Budko AI, Nesterov MS, Abaimov DA, Slominsky PA, Shadrina MI, Alieva AK. Effects of Age and MPTP-Induced Parkinson's Disease on the Expression of Genes Associated with the Regulation of the Sleep-Wake Cycle in Mice. Int J Mol Sci 2024; 25:7721. [PMID: 39062963 PMCID: PMC11276692 DOI: 10.3390/ijms25147721] [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: 06/10/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Parkinson's disease (PD) is characterized by a long prodromal period, during which patients often have sleep disturbances. The histaminergic system and circadian rhythms play an important role in the regulation of the sleep-wake cycle. Changes in the functioning of these systems may be involved in the pathogenesis of early stages of PD and may be age-dependent. Here, we have analyzed changes in the expression of genes associated with the regulation of the sleep-wake cycle (Hnmt, Hrh1, Hrh3, Per1, Per2, and Chrm3) in the substantia nigra (SN) and striatum of normal male mice of different ages, as well as in young and adult male mice with an MPTP-induced model of the early symptomatic stage (ESS) of PD. Age-dependent expression analysis in normal mouse brain tissue revealed changes in Hrh3, Per1, Per2, and Chrm3 genes in adult mice relative to young mice. When gene expression was examined in mice with the MPTP-induced model of the ESS of PD, changes in the expression of all studied genes were found only in the SN of adult mice with the ESS model of PD. These data suggest that age is a significant factor influencing changes in the expression of genes associated with sleep-wake cycle regulation in the development of PD.
Collapse
Affiliation(s)
- Ekaterina I. Semenova
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Margarita M. Rudenok
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Ivan N. Rybolovlev
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Marina V. Shulskaya
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Maria V. Lukashevich
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Suzanna A. Partevian
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Alexander I. Budko
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Maxim S. Nesterov
- Scientific Center for Biomedical Technologies of the Federal Biomedical Agency of Russia, 119435 Krasnogorsk, Russia;
| | - Denis A. Abaimov
- Research Center of Neurology, Volokolamskoye Shosse 80, 125367 Moscow, Russia;
| | - Petr A. Slominsky
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Maria I. Shadrina
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Anelya Kh. Alieva
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| |
Collapse
|
8
|
Ayerra L, Abellanas MA, Basurco L, Tamayo I, Conde E, Tavira A, Trigo A, Vidaurre C, Vilas A, San Martin-Uriz P, Luquin E, Clavero P, Mengual E, Hervás-Stubbs S, Aymerich MS. Nigrostriatal degeneration determines dynamics of glial inflammatory and phagocytic activity. J Neuroinflammation 2024; 21:92. [PMID: 38610019 PMCID: PMC11015575 DOI: 10.1186/s12974-024-03091-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Glial cells are key players in the initiation of innate immunity in neurodegeneration. Upon damage, they switch their basal activation state and acquire new functions in a context and time-dependent manner. Since modulation of neuroinflammation is becoming an interesting approach for the treatment of neurodegenerative diseases, it is crucial to understand the specific contribution of these cells to the inflammatory reaction and to select experimental models that recapitulate what occurs in the human disease. Previously, we have characterized a region-specific activation pattern of CD11b+ cells and astrocytes in the α-synuclein overexpression mouse model of Parkinson´s disease (PD). In this study we hypothesized that the time and the intensity of dopaminergic neuronal death would promote different glial activation states. Dopaminergic degeneration was induced with two administration regimens of the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), subacute (sMPTP) and chronic (cMPTP). Our results show that in the sMPTP mouse model, the pro-inflammatory phenotype of striatal CD11b+ cells was counteracted by an anti-inflammatory astrocytic profile. In the midbrain the roles were inverted, CD11b+ cells exhibited an anti-inflammatory profile and astrocytes were pro-inflammatory. The overall response generated resulted in decreased CD4 T cell infiltration in both regions. Chronic MPTP exposure resulted in a mild and prolonged neuronal degeneration that generated a pro-inflammatory response and increased CD4 T cell infiltration in both regions. At the onset of the neurodegenerative process, microglia and astrocytes cooperated in the removal of dopaminergic terminals. With time, only microglia maintained the phagocytic activity. In the ventral midbrain, astrocytes were the main phagocytic mediators at early stages of degeneration while microglia were the major phagocytic cells in the chronic state. In this scenario, we questioned which activation pattern recapitulates better the features of glial activation in PD. Glial activation in the cMPTP mouse model reflects many pathways of their corresponding counterparts in the human brain with advanced PD. Altogether, our results point toward a context-dependent cooperativity of microglia/myeloid cells and astrocytes in response to neuronal damage and the relevance of selecting the right experimental models for the study of neuroinflammation.
Collapse
Grants
- PI20/01063 Instituto de Salud Carlos III
- PI20/01063 Instituto de Salud Carlos III
- PI20/01063 Instituto de Salud Carlos III
- PI20/01063 Instituto de Salud Carlos III
- PI20/01063 Instituto de Salud Carlos III
- PI20/01063 Instituto de Salud Carlos III
- PI20/01063 Instituto de Salud Carlos III
- FPU19/03255 Ministerio de Ciencia, Innovación y Universidades
- PC060-061 Dirección General de Industria, Energia y Proyectos Estrategicos S3, Gobierno de Navarra
- PC060-061 Dirección General de Industria, Energia y Proyectos Estrategicos S3, Gobierno de Navarra
- PC060-061 Dirección General de Industria, Energia y Proyectos Estrategicos S3, Gobierno de Navarra
- PC060-061 Dirección General de Industria, Energia y Proyectos Estrategicos S3, Gobierno de Navarra
- PC060-061 Dirección General de Industria, Energia y Proyectos Estrategicos S3, Gobierno de Navarra
- FPU18/02244 Ministerio de Ciencia, Innovación y Universidades,Spain
- FPU21/01545 Ministerio de Ciencia, Innovación y Universidades,Spain
Collapse
Affiliation(s)
- Leyre Ayerra
- Facultad de Ciencias, Departamento de Bioquímica y Genética, Universidad de Navarra, Pamplona, Spain
- CIMA-Universidad de Navarra, Pamplona, España
| | - Miguel Angel Abellanas
- Facultad de Ciencias, Departamento de Bioquímica y Genética, Universidad de Navarra, Pamplona, Spain
- CIMA-Universidad de Navarra, Pamplona, España
| | - Leyre Basurco
- Facultad de Ciencias, Departamento de Bioquímica y Genética, Universidad de Navarra, Pamplona, Spain
- CIMA-Universidad de Navarra, Pamplona, España
| | - Ibon Tamayo
- CIMA-Universidad de Navarra, Pamplona, España
| | | | - Adriana Tavira
- Facultad de Ciencias, Departamento de Bioquímica y Genética, Universidad de Navarra, Pamplona, Spain
- CIMA-Universidad de Navarra, Pamplona, España
| | - Amaya Trigo
- Facultad de Ciencias, Departamento de Bioquímica y Genética, Universidad de Navarra, Pamplona, Spain
- CIMA-Universidad de Navarra, Pamplona, España
| | - Clara Vidaurre
- Facultad de Ciencias, Departamento de Bioquímica y Genética, Universidad de Navarra, Pamplona, Spain
- CIMA-Universidad de Navarra, Pamplona, España
| | - Amaia Vilas
- CIMA-Universidad de Navarra, Pamplona, España
| | | | - Esther Luquin
- Facultad de Medicina, Departamento de Patología, Anatomía y Fisiología, Universidad de Navarra, Pamplona, Spain
| | - Pedro Clavero
- Servicio de Neurología, Hospital Universitario de Navarra, Pamplona, Spain
| | - Elisa Mengual
- Facultad de Medicina, Departamento de Patología, Anatomía y Fisiología, Universidad de Navarra, Pamplona, Spain
| | - Sandra Hervás-Stubbs
- CIMA-Universidad de Navarra, Pamplona, España
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Maria S Aymerich
- Facultad de Ciencias, Departamento de Bioquímica y Genética, Universidad de Navarra, Pamplona, Spain.
- CIMA-Universidad de Navarra, Pamplona, España.
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain.
| |
Collapse
|
9
|
Tassan Mazzocco M, Serra M, Maspero M, Coliva A, Presotto L, Casu MA, Morelli M, Moresco RM, Belloli S, Pinna A. Positive relation between dopamine neuron degeneration and metabolic connectivity disruption in the MPTP plus probenecid mouse model of Parkinson's disease. Exp Neurol 2024; 374:114704. [PMID: 38281587 DOI: 10.1016/j.expneurol.2024.114704] [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: 10/11/2023] [Revised: 01/15/2024] [Accepted: 01/25/2024] [Indexed: 01/30/2024]
Abstract
The clinical manifestation of Parkinson's disease (PD) appears when neurodegeneration is already advanced, compromising the efficacy of disease-modifying treatment approaches. Biomarkers to identify the early stages of PD are therefore of paramount importance for the advancement of the therapy of PD. In the present study, by using a mouse model of PD obtained by subchronic treatment with the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and the clearance inhibitor probenecid (MPTPp), we identified prodromal markers of PD by combining in vivo positron emission tomography (PET) imaging and ex vivo immunohistochemistry. Longitudinal PET imaging of the dopamine transporter (DAT) by [18F]-N-(3-fluoropropyl)-2β-carboxymethoxy-3β-(4-iodophenyl) nortropane ([18F]-FP-CIT), and brain glucose metabolism by 2-deoxy-2-[18F]-fluoroglucose ([18F]-FDG) were performed before MPTPp treatment and after 1, 3, and 10 MPTPp administrations, in order to assess relation between dopamine neuron integrity and brain connectivity. The results show that in vivo [18F]-FP-CIT in the dorsal striatum was not modified after the first administration of MPTPp, tended to decrease after 3 administrations, and significantly decreased after 10 MPTPp administrations. Post-mortem immunohistochemical analyses of DAT and tyrosine hydroxylase (TH) in the striatum showed a positive correlation with [18F]-FP-CIT, confirming the validity of repeated MPTPp-treated mice as a model that can reproduce the progressive pathological changes in the early phases of PD. Analysis of [18F]-FDG uptake in several brain areas connected to the striatum showed that metabolic connectivity was progressively disrupted, starting from the first MPTPp administration, and that significant connections between cortical and subcortical regions were lost after 10 MPTPp administrations, suggesting an association between dopamine neuron degeneration and connectivity disruption in this PD model. The results of this study provide a relevant model, where new drugs that can alleviate neurodegeneration in PD could be evaluated preclinically.
Collapse
Affiliation(s)
- Margherita Tassan Mazzocco
- PhD Program in Neuroscience, Medicine and Surgery Department, University of Milano-Bicocca, Monza, Italy; Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Marcello Serra
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Marco Maspero
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; National Research Council of Italy, Institute of Molecular Bioimaging and Physiology, UOS of Segrate, Italy
| | - Angela Coliva
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Luca Presotto
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; Department of Physics "G. Occhialini", University of Milano - Bicocca, Milan, Italy
| | - Maria Antonietta Casu
- National Research Council of Italy, Institute of Translational Pharmacology, UOS of Cagliari, Scientific and Technological Park of Sardinia POLARIS, Pula, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy; National Research Council of Italy, Neuroscience Institute, UOS of Cagliari, Italy
| | - Rosa Maria Moresco
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; National Research Council of Italy, Institute of Molecular Bioimaging and Physiology, UOS of Segrate, Italy; School of Medicine and Surgery, University of Milano - Bicocca, Monza, Italy.
| | - Sara Belloli
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; National Research Council of Italy, Institute of Molecular Bioimaging and Physiology, UOS of Segrate, Italy
| | - Annalisa Pinna
- National Research Council of Italy, Neuroscience Institute, UOS of Cagliari, Italy
| |
Collapse
|
10
|
Lee MH, Kang S, Um KH, Lee SW, Hwang H, Baek K, Choi JW. Brain-targeted delivery of neuroprotective survival gene minimizing hematopoietic cell contamination: implications for Parkinson's disease treatment. J Transl Med 2024; 22:53. [PMID: 38218903 PMCID: PMC10790275 DOI: 10.1186/s12967-023-04816-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/18/2023] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Neurodegenerative diseases, including Parkinson's disease, Amyotropic Lateral Sclerosis (ALS) and Alzheimer's disease, present significant challenges for therapeutic development due to drug delivery restrictions and toxicity concerns. Prevailing strategies often employ adeno-associated viral (AAV) vectors to deliver neuroprotective survival genes directly into the central nervous system (CNS). However, these methods have been limited by triggering immunogenic responses and risk of tumorigenicity, resulting from overexpression of survival genes in peripheral blood mononuclear cells (PBMC), thereby increasing the risk of tumorigenicity in specific immune cells. Thus, by coding selectively suppressive microRNA (miRNA) target sequences in AAV genome, we designed CNS-targeted neuroprotective gene expression vector system without leakage to blood cells. METHODS To minimize the potential for transgene contamination in the blood, we designed a CNS-specific AAV system. Our system utilized a self-complementary AAV (scAAV), encoding a quadruple repeated target sequence of the hematopoietic cell-specific miR142-3p at the 3' untranslated region (UTR). As a representative therapeutic survival gene for Parkinson's disease treatment, we integrated DX2, an antagonistic splice variant of the apoptotic gene AIMP2, known to be implicated in Parkinson's disease, into the vector. RESULTS This configuration ensured that transgene expression was stringently localized to the CNS, even if the vector found its way into the blood cells. A single injection of scAAV-DX2 demonstrated marked improvement in behavior and motor activity in animal models of Parkinson's disease induced by either Rotenone or 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Importantly, comprehensive preclinical data adhering to Good Laboratory Practice (GLP) standards revealed no adverse effects in the treated animals. CONCLUSIONS Our CNS-specific vector system, which encodes a survival transgene DX2, signifies a promising avenue for safe gene therapy, avoiding unintended expression of survival gene in blood cells, applicable to various neurodegenerative diseases.
Collapse
Affiliation(s)
- Min Hak Lee
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Biological and Medicinal Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Pharmacology, Institute of Regulatory Innovation Through Science, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sukyeong Kang
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Biological and Medicinal Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ki-Hwan Um
- Department of Biological and Medicinal Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Pharmacology, Institute of Regulatory Innovation Through Science, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Seok Won Lee
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Biological and Medicinal Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyorin Hwang
- Generoath Ltd., Seoul, 04168, Republic of Korea
- Department of Pharmacology, College of Dentistry and Research Institute of Oral Science, Gangneung-Wonju National University, Gangneung-si, Gangwon-do, 25457, Republic of Korea
| | - Kyunghwa Baek
- Generoath Ltd., Seoul, 04168, Republic of Korea.
- Department of Pharmacology, College of Dentistry and Research Institute of Oral Science, Gangneung-Wonju National University, Gangneung-si, Gangwon-do, 25457, Republic of Korea.
| | - Jin Woo Choi
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Biological and Medicinal Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Pharmacology, Institute of Regulatory Innovation Through Science, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
11
|
Park M, Ha J, Lee Y, Choi HS, Kim BS, Jeong YK. Low-moderate dose whole-brain γ-ray irradiation modulates the expressions of glial fibrillary acidic protein and intercellular adhesion molecule-1 in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson's disease mouse model. Neurobiol Aging 2023; 132:175-184. [PMID: 37837733 DOI: 10.1016/j.neurobiolaging.2023.06.015] [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: 02/28/2023] [Revised: 06/22/2023] [Accepted: 06/24/2023] [Indexed: 10/16/2023]
Abstract
The anti-inflammatory efficacy of radiation therapy (RT) with single fractions below 1.0 Gy has been demonstrated in Alzheimer's disease mouse models. As neuroinflammation is also a major pathological feature of Parkinson's disease (PD), RT may also be effective in PD treatment. Therefore, this study aimed to investigate the anti-inflammatory effect of low-moderate dose RT (LMDRT, 0.6 Gy/single dose, for 5 days) exposure in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP; 30 mg/kg, intraperitoneally, for 5 consecutive days)-induced PD mouse model. Importantly, LMDRT reduced the levels of glial fibrillary acidic protein and intercellular adhesion molecule-1 (CD54) in the striatum region, which increased following MPTP administration. LMDRT also modulated inflammatory gene expression patterns in the substantia nigra region of the MPTP-treated mice. However, LMDRT had no direct effects on the severe loss of dopaminergic neurons and impaired motor behavior in the rotarod test. These results indicate that LMDRT has anti-inflammatory effects by modulating neuroinflammatory factors, including glial fibrillary acidic protein and intercellular adhesion molecule-1, but showed no behavioral improvements or neuroprotection in the MPTP-induced mouse model of PD.
Collapse
MESH Headings
- Animals
- Mice
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/therapeutic use
- Brain/metabolism
- Brain/radiation effects
- Disease Models, Animal
- Dopaminergic Neurons/pathology
- Glial Fibrillary Acidic Protein/metabolism
- Intercellular Adhesion Molecule-1/metabolism
- Intercellular Adhesion Molecule-1/pharmacology
- Intercellular Adhesion Molecule-1/therapeutic use
- Mice, Inbred C57BL
- Parkinson Disease/metabolism
- Parkinson Disease/radiotherapy
- Substantia Nigra/metabolism
Collapse
Affiliation(s)
- Mijeong Park
- Radiological and Medical Support Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Jimin Ha
- Radiological and Medical Support Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Division of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Yuri Lee
- Radiological and Medical Support Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Division of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Hoon-Seong Choi
- Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Byoung Soo Kim
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Youn Kyoung Jeong
- Radiological and Medical Support Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea.
| |
Collapse
|
12
|
He Y, Zhao J, Dong H, Zhang X, Duan Y, Ma Y, Yu M, Fei J, Huang F. TLR2 deficiency is beneficial at the late phase in MPTP-induced Parkinson' disease mice. Life Sci 2023; 333:122171. [PMID: 37827233 DOI: 10.1016/j.lfs.2023.122171] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 10/07/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
AIMS Parkinson's disease (PD) is a progressive neurodegenerative disorder. The etiology of PD is still elusive but neuroinflammation is proved to be an important contributor. Toll-like receptor 2 (TLR2) involves in the release of several inflammatory cytokines. Whether TLR2 serves as a mediator contributing to the damage of DA system in PD remain unclear. MAIN METHODS Tlr2 knockout (Tlr2-/-) and wild-type (WT) mice were treated with a subacute regimen of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). At 3, 7 and 14 days after MPTP injection, the behavioral performance, including the Pole test, the Rotarod test, the Rearing test and the Wire hang test was evaluated. Moreover, the PD-like phenotypes, including dopaminergic degeneration, the activation of glial cells and the α-Syn expression were systematically analyzed in the nigrostriatal pathway. Finally, the composition of gut microbiota in the MPTP-treated groups were assessed. KEY FINDINGS TLR2 deficiency had no obvious impact on the dopaminergic injury at 3 and 7 days following MPTP administration. On the contrary, at 14 days post injection, TLR2 deficiency not only significantly attenuated motor deficits in the Pole test and the Rotarod test, and the nigrostriatal dopaminergic degeneration, but also mitigated α-Syn abnormality, astrocyte activation and neuroinflammation through the suppressed TLR2/MyD88/TRAF6/NF-κB signaling pathways. Additionally, the alteration of gut microbiota was also detected in the mutant mice. SIGNIFICANCE These findings highlight the neuroprotective effect of TLR2-pathways at the late phase in the MPTP-induced PD mouse model.
Collapse
Affiliation(s)
- Yongtao He
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jiayin Zhao
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Hongtian Dong
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Xiaoshuang Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yufei Duan
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yuanyuan Ma
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Mei Yu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.
| | - Jian Fei
- School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Engineering Research Center for Model Organisms, Shanghai Model Organisms Center, INC., Shanghai 201203, China.
| | - Fang Huang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.
| |
Collapse
|
13
|
Qi Y, Zhang Z, Li Y, Zhao G, Huang J, Zhang Y, Xue J, Tang X. Whether the Subacute MPTP-Treated Mouse is as Suitable as a Classic Model of Parkinsonism. Neuromolecular Med 2023; 25:360-374. [PMID: 36913134 DOI: 10.1007/s12017-023-08740-7] [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: 01/24/2022] [Accepted: 01/08/2023] [Indexed: 03/14/2023]
Abstract
1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mice model is one of the most common animal models for Parkinson's disease (PD). It is classified into three types: acute, subacute, and chronic intoxication models. The subacute model has attracted much attention for its short period and similarity to PD. However, whether subacute MPTP intoxication in mouse mimics the movement and cognitive disorders of PD still remains highly controversial. Therefore, the present study reassessed the behavioral performances of subacute MPTP intoxication in mice using open field, rotarod, Y maze, and gait analysis at different time points (1, 7, 14, and 21 days) after modeling. Results of the current study showed that although MPTP-treated mice using subacute regimen showed severe dopaminergic neuronal loss and evident astrogliosis, they failed to display significant motor and cognitive deficits. Besides, expression of mixed lineage kinase domain-like (MLKL), a marker of necroptosis, was also significantly increased in the ventral midbrain and striatum of MPTP-intoxicated mice. This evidently implies that necroptosis may play an important role in MPTP-induced neurodegeneration. In conclusion, the findings of the present study suggest that subacute MPTP-intoxicated mice may not be a suitable model for studying parkinsonism. However, it can help in revealing the early pathophysiology of PD and studying the compensatory mechanisms which occur in early PD that prevent the emergence of behavioral deficits.
Collapse
Affiliation(s)
- Yue Qi
- Department of Human Anatomy, School of Basic Medical Sciences, Gannan Medical University, Harmonious Avenue, Zhang Gong District, Ganzhou, 341000, China
| | - Ziwei Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Gannan Medical University, Harmonious Avenue, Zhang Gong District, Ganzhou, 341000, China
| | - Yanning Li
- Department of Forensic Medicine, School of Basic Medical Sciences, Gannan Medical University, Ganzhou, 341000, China
| | - Guojian Zhao
- School of Rehabilitation Sciences, Gannan Medical University, Ganzhou, 341000, China
| | - Jinyong Huang
- School of Rehabilitation Sciences, Gannan Medical University, Ganzhou, 341000, China
| | - Yi Zhang
- Department of Forensic Medicine, School of Basic Medical Sciences, Gannan Medical University, Ganzhou, 341000, China
| | - Jinhua Xue
- Department of Pathophysiology, School of Basic Medical Sciences, Gannan Medical University, Harmonious Avenue, Zhang Gong District, Ganzhou, 341000, China.
| | - Xiaolu Tang
- Department of Human Anatomy, School of Basic Medical Sciences, Gannan Medical University, Harmonious Avenue, Zhang Gong District, Ganzhou, 341000, China.
| |
Collapse
|
14
|
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
|
15
|
Mirzac D, Kreis SL, Luhmann HJ, Gonzalez-Escamilla G, Groppa S. Translating Pathological Brain Activity Primers in Parkinson's Disease Research. RESEARCH (WASHINGTON, D.C.) 2023; 6:0183. [PMID: 37383218 PMCID: PMC10298229 DOI: 10.34133/research.0183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/02/2023] [Indexed: 06/30/2023]
Abstract
Translational experimental approaches that help us better trace Parkinson's disease (PD) pathophysiological mechanisms leading to new therapeutic targets are urgently needed. In this article, we review recent experimental and clinical studies addressing abnormal neuronal activity and pathological network oscillations, as well as their underlying mechanisms and modulation. Our aim is to enhance our knowledge about the progression of Parkinson's disease pathology and the timing of its symptom's manifestation. Here, we present mechanistic insights relevant for the generation of aberrant oscillatory activity within the cortico-basal ganglia circuits. We summarize recent achievements extrapolated from available PD animal models, discuss their advantages and limitations, debate on their differential applicability, and suggest approaches for transferring knowledge on disease pathology into future research and clinical applications.
Collapse
Affiliation(s)
- Daniela Mirzac
- Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Svenja L. Kreis
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Heiko J. Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Gabriel Gonzalez-Escamilla
- Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Sergiu Groppa
- Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| |
Collapse
|
16
|
Pfab A, Belikov S, Keuper M, Jastroch M, Mannervik M. Inhibition of mitochondrial transcription by the neurotoxin MPP . Exp Cell Res 2023; 425:113536. [PMID: 36858342 DOI: 10.1016/j.yexcr.2023.113536] [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: 11/23/2022] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/02/2023]
Abstract
The neurotoxin MPP+ triggers cell death of dopamine neurons and induces Parkinson's disease symptoms in mice and men, but the immediate transcriptional response to this neurotoxin has not been studied. We therefore treated human SH-SY5Y cells with a low dose (0.1 mM) of MPP+ and measured the effect on nascent transcription by precision run-on sequencing (PRO-seq). We found that transcription of the mitochondrial genome was significantly reduced already after 30 min, whereas nuclear gene transcription was unaffected. Inhibition of respiratory complex I by MPP+ led to reduced ATP production, that may explain the diminished activity of mitochondrial RNA polymerase. Our results show that MPP+ has a direct effect on mitochondrial function and transcription, and that other gene expression or epigenetic changes induced by this neurotoxin are secondary effects that reflect a cellular adaptation program.
Collapse
Affiliation(s)
- Alexander Pfab
- Dept. Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden
| | - Sergey Belikov
- Dept. Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden
| | - Michaela Keuper
- Dept. Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden
| | - Martin Jastroch
- Dept. Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden
| | - Mattias Mannervik
- Dept. Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden.
| |
Collapse
|
17
|
Santoro M, Fadda P, Klephan KJ, Hull C, Teismann P, Platt B, Riedel G. Neurochemical, histological, and behavioral profiling of the acute, sub-acute, and chronic MPTP mouse model of Parkinson's disease. J Neurochem 2023; 164:121-142. [PMID: 36184945 PMCID: PMC10098710 DOI: 10.1111/jnc.15699] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/25/2022] [Accepted: 09/30/2022] [Indexed: 02/04/2023]
Abstract
Parkinson's disease (PD) is a heterogeneous multi-systemic disorder unique to humans characterized by motor and non-motor symptoms. Preclinical experimental models of PD present limitations and inconsistent neurochemical, histological, and behavioral readouts. The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD is the most common in vivo screening platform for novel drug therapies; nonetheless, behavioral endpoints yielded amongst laboratories are often discordant and inconclusive. In this study, we characterized neurochemically, histologically, and behaviorally three different MPTP mouse models of PD to identify translational traits reminiscent of PD symptomatology. MPTP was intraperitoneally (i.p.) administered in three different regimens: (i) acute-four injections of 20 mg/kg of MPTP every 2 h; (ii) sub-acute-one daily injection of 30 mg/kg of MPTP for 5 consecutive days; and (iii) chronic-one daily injection of 4 mg/kg of MPTP for 28 consecutive days. A series of behavioral tests were conducted to assess motor and non-motor behavioral changes including anxiety, endurance, gait, motor deficits, cognitive impairment, circadian rhythm and food consumption. Impairments in balance and gait were confirmed in the chronic and acute models, respectively, with the latter showing significant correlation with lesion size. The sub-acute model, by contrast, presented with generalized hyperactivity. Both, motor and non-motor changes were identified in the acute and sub-acute regime where habituation to a novel environment was significantly reduced. Moreover, we report increased water and food intake across all three models. Overall, the acute model displayed the most severe lesion size, while across the three models striatal dopamine content (DA) did not correlate with the behavioral performance. The present study demonstrates that detection of behavioral changes following MPTP exposure is challenging and does not correlate with the dopaminergic lesion extent.
Collapse
Affiliation(s)
- Matteo Santoro
- Institute of Medical SciencesUniversity of AberdeenAberdeenUK
- Present address:
Department of Neurosurgery, School of MedicineStanford UniversityPalo AltoCaliforniaUSA
| | - Paola Fadda
- Department of NeuroscienceUniversity of CagliariCagliariItaly
| | - Katie J. Klephan
- Newcastle UniversitySchool of Biomedical, Nutritional, and Sport SciencesNewcastle upon TyneUK
- Present address:
AccuRXLondonLondonUK
| | - Claire Hull
- Institute of Medical SciencesUniversity of AberdeenAberdeenUK
| | - Peter Teismann
- Institute of Medical SciencesUniversity of AberdeenAberdeenUK
| | - Bettina Platt
- Institute of Medical SciencesUniversity of AberdeenAberdeenUK
| | - Gernot Riedel
- Institute of Medical SciencesUniversity of AberdeenAberdeenUK
| |
Collapse
|
18
|
Kolacheva A, Bannikova A, Pavlova E, Bogdanov V, Ugrumov M. Modeling of the Progressive Degradation of the Nigrostriatal Dopaminergic System in Mice to Study the Mechanisms of Neurodegeneration and Neuroplasticity in Parkinson's Disease. Int J Mol Sci 2022; 24:ijms24010683. [PMID: 36614126 PMCID: PMC9820573 DOI: 10.3390/ijms24010683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/25/2022] [Accepted: 12/26/2022] [Indexed: 01/03/2023] Open
Abstract
The fight against neurodegenerative diseases, including Parkinson's disease (PD), is among the global challenges of the 21st century. The low efficiency of therapy is due to the late diagnosis and treatment of PD, which take place when there is already significant degradation of the nigrostriatal dopaminergic system, a key link in the regulation of motor function. We have developed a subchronic mouse model of PD by repeatedly administering 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) at gradually increasing doses with a 24 h interval between injections, a period comparable to the time of MPTP metabolism and elimination from the body. This model reproduces the main hallmarks of PD: progressive degeneration of dopaminergic neurons; the appearance of motor disorders with a 70-80% decrease in the level of dopamine in the striatum; an increase in dopamine turnover in the striatum to compensate for dopamine deficiency. When comparing the degradation of the nigrostriatal dopaminergic system and motor disorders in mice in the acute and subchronic models of PD, it has turned out that the resistance of dopaminergic neurons to MPTP increases with its repeated administration. Our subchronic model of PD opens up broad prospects for studying the molecular mechanisms of PD pathogenesis and developing technologies for early diagnosis and preventive treatment.
Collapse
|
19
|
Zhong Z, He X, Ge J, Zhu J, Yao C, Cai H, Ye XY, Xie T, Bai R. Discovery of small-molecule compounds and natural products against Parkinson's disease: Pathological mechanism and structural modification. Eur J Med Chem 2022; 237:114378. [DOI: 10.1016/j.ejmech.2022.114378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/08/2021] [Accepted: 04/09/2022] [Indexed: 11/24/2022]
|
20
|
Tong W, Zhang K, Yao H, Li L, Hu Y, Zhang J, Song Y, Guan Q, Li S, Sun YE, Jin L. Transcriptional Profiling Reveals Brain Region-Specific Gene Networks Regulated in Exercise in a Mouse Model of Parkinson’s Disease. Front Aging Neurosci 2022; 14:891644. [PMID: 35813950 PMCID: PMC9260255 DOI: 10.3389/fnagi.2022.891644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/12/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundExercise plays an essential role in improving motor symptoms in Parkinson’s disease (PD), but the underlying mechanism in the central nervous system remains unclear.MethodsMotor ability was observed after 12-week treadmill exercise on a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. RNA-sequencing on four brain regions (cerebellum, cortex, substantia nigra (SN), and striatum) from control animals, MPTP-induced PD, and MPTP-induced PD model treated with exercise for 12 weeks were performed. Transcriptional networks on the four regions were further identified by an integrative network biology approach.ResultsThe 12-week treadmill exercise significantly improved the motor ability of an MPTP-induced mouse model of PD. RNA-seq analysis showed SN and striatum were remarkably different among individual region’s response to exercise in the PD model. Especially, synaptic regulation pathways about axon guidance, synapse assembly, neurogenesis, synaptogenesis, transmitter transport-related pathway, and synaptic regulation genes, including Neurod2, Rtn4rl2, and Cd5, were upregulated in SN and striatum. Lastly, immunofluorescence staining revealed that exercise rescued the loss of TH+ synapses in the striatal region in PD mice, which validates the key role of synaptic regulation pathways in exercise-induced protective effects in vivo.ConclusionSN and striatum are important brain regions in which critical transcriptional changes, such as in synaptic regulation pathways, occur after the exercise intervention on the PD model.
Collapse
Affiliation(s)
- Weifang Tong
- Department of Neurology, Tongji Hospital, School of Medicine, Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji University, Shanghai, China
- Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kunshan Zhang
- Department of Neurology, Tongji Hospital, School of Medicine, Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji University, Shanghai, China
| | - Hongkai Yao
- Department of Neurology, Tongji Hospital, School of Medicine, Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji University, Shanghai, China
| | - Lixi Li
- Department of Neurology, Tongji Hospital, School of Medicine, Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji University, Shanghai, China
| | - Yong Hu
- The Marlene and Paolo Fresco Institute for Parkinson’s and Movement Disorders, Department of Neurology, NYU Langone Health, NYU School of Medicine, New York, NY, United States
| | - Jingxing Zhang
- Department of Neurology, Tongji Hospital, School of Medicine, Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji University, Shanghai, China
| | - Yunping Song
- Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qiang Guan
- Department of Neurology, Tongji Hospital, School of Medicine, Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji University, Shanghai, China
| | - Siguang Li
- Department of Neurology, Tongji Hospital, School of Medicine, Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji University, Shanghai, China
- *Correspondence: Siguang Li,
| | - Yi E. Sun
- Department of Neurology, Tongji Hospital, School of Medicine, Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji University, Shanghai, China
- Yi E. Sun,
| | - Lingjing Jin
- Department of Neurology, Tongji Hospital, School of Medicine, Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji University, Shanghai, China
- Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital, School of Medicine, Tongji University, Shanghai, China
- Lingjing Jin,
| |
Collapse
|
21
|
UPLC-MS/MS assay for the simultaneous determination of catecholamines and their metabolites at low pg/mg in rat/mouse striatum. J Pharm Biomed Anal 2022; 213:114697. [DOI: 10.1016/j.jpba.2022.114697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 02/17/2022] [Accepted: 02/26/2022] [Indexed: 12/23/2022]
|
22
|
Doyle JM, Croll RP. A Critical Review of Zebrafish Models of Parkinson's Disease. Front Pharmacol 2022; 13:835827. [PMID: 35370740 PMCID: PMC8965100 DOI: 10.3389/fphar.2022.835827] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/08/2022] [Indexed: 11/17/2022] Open
Abstract
A wide variety of human diseases have been modelled in zebrafish, including various types of cancer, cardiovascular diseases and neurodegenerative diseases like Alzheimer’s and Parkinson’s. Recent reviews have summarized the currently available zebrafish models of Parkinson’s Disease, which include gene-based, chemically induced and chemogenetic ablation models. The present review updates the literature, critically evaluates each of the available models of Parkinson’s Disease in zebrafish and compares them with similar models in invertebrates and mammals to determine their advantages and disadvantages. We examine gene-based models, including ones linked to Early-Onset Parkinson’s Disease: PARKIN, PINK1, DJ-1, and SNCA; but we also examine LRRK2, which is linked to Late-Onset Parkinson’s Disease. We evaluate chemically induced models like MPTP, 6-OHDA, rotenone and paraquat, as well as chemogenetic ablation models like metronidazole-nitroreductase. The article also reviews the unique advantages of zebrafish, including the abundance of behavioural assays available to researchers and the efficiency of high-throughput screens. This offers a rare opportunity for assessing the potential therapeutic efficacy of pharmacological interventions. Zebrafish also are very amenable to genetic manipulation using a wide variety of techniques, which can be combined with an array of advanced microscopic imaging methods to enable in vivo visualization of cells and tissue. Taken together, these factors place zebrafish on the forefront of research as a versatile model for investigating disease states. The end goal of this review is to determine the benefits of using zebrafish in comparison to utilising other animals and to consider the limitations of zebrafish for investigating human disease.
Collapse
Affiliation(s)
- Jillian M Doyle
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Roger P Croll
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
23
|
Sinitsyn D, Garcia-Reyero N, Watanabe KH. From Qualitative to Quantitative AOP: A Case Study of Neurodegeneration. FRONTIERS IN TOXICOLOGY 2022; 4:838729. [PMID: 35434701 PMCID: PMC9006165 DOI: 10.3389/ftox.2022.838729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/24/2022] [Indexed: 12/17/2022] Open
Abstract
Adverse outcome pathways (AOPs) include a sequence of events that connect a molecular-level initiating event with an adverse outcome at the cellular level for human health endpoints, or at the population level for ecological endpoints. When there is enough quantitative understanding of the relationships between key events in an AOP, a mathematical model may be developed to connect key events in a quantitative AOP (qAOP). Ideally, a qAOP will reduce the time and resources spent for chemical toxicity testing and risk assessment and enable the extrapolation of data collected at the molecular-level by in vitro assays, for example, to predict whether an adverse outcome may occur. Here, we review AOPs in the AOPWiki, an AOP repository, to determine best practices that would facilitate conversion from AOP to qAOP. Then, focusing on a particular case study, acetylcholinesterase inhibition leading to neurodegeneration, we describe specific methods and challenges. Examples of challenges include the availability and collection of quantitative data amenable to model development, the lack of studies that measure multiple key events, and model accessibility or transferability across platforms. We conclude with recommendations for improving key event and key event relationship descriptions in the AOPWiki that facilitate the transition of qualitative AOPs to qAOPs.
Collapse
Affiliation(s)
- Dennis Sinitsyn
- Arizona State University, School of Mathematical and Natural Sciences, Glendale, AZ, United States
- Oak Ridge Institute for Science and Education, Environmental Laboratory, US Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - Natàlia Garcia-Reyero
- US Army Engineer Research and Development Center, Environmental Laboratory, Vicksburg, MS, United States
| | - Karen H. Watanabe
- Arizona State University, School of Mathematical and Natural Sciences, Glendale, AZ, United States
- *Correspondence: Karen H. Watanabe,
| |
Collapse
|
24
|
Ginsenoside Rg1 Plays a Neuroprotective Role in Regulating the Iron-Regulated Proteins and Against Lipid Peroxidation in Oligodendrocytes. Neurochem Res 2022; 47:1721-1735. [PMID: 35229270 DOI: 10.1007/s11064-022-03564-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/20/2022] [Accepted: 02/21/2022] [Indexed: 01/18/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder. Progressive loss of dopaminergic neurons in the substantia nigra (SN) is one of the major pathological changes. However, the reasons for the dopaminergic neuron loss are still ambiguous and further studies are needed to evaluate the in-depth mechanisms of neuron death. Oxidative stress is a significant factor causing neuronal damage. Dopaminergic neurons in the SN are susceptible to oxidative stress, which is closely associated with iron dyshomeostasis in the brain. Ginsenoside Rg1 from ginseng plays a crucial role in neuroprotective effects through anti-inflammation and attenuating the aggregation of abnormal α-synuclein. In our study, we established a chronic PD mouse model by 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine combined with probenecid and explored the effect of Rg1 on the oxidative stress and brain iron homeostasis. Rg1 was verified to improve the level of tyrosine hydroxylase and anti-oxidant stress. In addition, Rg1 maintained the iron-regulated protein homeostasis by increasing the expression of ferritin heavy chain and decreasing ferritin light chain in oligodendrocytes, especially the mature oligodendrocytes (OLs). Furthermore, Rg1 had a positive effect on the myelin sheath protection and increased the number of mature oligodendrocytes, proved by the increased staining of myelin basic protein and CC-1. In conclusion, Rg1 could play a neuroprotective role through remitting the iron-regulated protein dyshomeostasis by ferritin and against lipid peroxidation stress in oligodendrocytes.
Collapse
|
25
|
Zhang Z, Li M, Zuo Y, Chen S, Zhuo Y, Lu M, Shi G, Gu H. In Vivo Monitoring of pH in Subacute PD Mouse Brains with a Ratiometric Electrochemical Microsensor Based on Poly(melamine) Films. ACS Sens 2022; 7:235-244. [PMID: 34936337 DOI: 10.1021/acssensors.1c02051] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In vivo monitoring of cerebral pH is of great significance because its disturbance is related to some pathological processes such as neurodegenerative diseases, for example, Parkinson's disease (PD). In this study, we developed an electrochemical microsensor based on poly(melamine) (PMel) films for ratiometric monitoring of pH in subacute PD mouse brains. In this microsensor, PMel films were prepared from a simple electropolymerization approach in a melamine-containing solution, serving as the selective pH recognition membrane undergoing a 2H+/2e- process. Meanwhile, electrochemically oxidized graphene oxide (EOGO) produced a built-in correction signal which helped avoid the environmental interference of the complicated brain systems. The potential difference between the peaks generated from EOGO and PMel gradually decreased with the aqueous pH increasing from 4.0 to 9.0, constituting the detection foundation of the ratiometric electrochemical microsensor (REM). The in vitro studies demonstrated that this proposed method exhibited a high sensitivity (a Nernstian response of -61.35 mV/pH) and remarkable selectivity against amino acids, anions, cations, and biochemical and reactive oxygen species coexisting in the brain. Coupled with its excellent stability and reproducibility and good antibiofouling based on short-term detection, the developed REM could serve as a disposable sensor for the determination of cerebral pH in vivo. Its following successful application in the real-time measurement of pH in the striatum, hippocampus, and cortex of rat brains in the events of global cerebral ischemia/reperfusion verified the reliability of this method. Finally, we adopted this robust REM to systematically analyze and compare the average pH in different regions of normal and subacute PD mouse brains.
Collapse
Affiliation(s)
- Ziyi Zhang
- A Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan 411201, P. R. China
| | - Mengyin Li
- A Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan 411201, P. R. China
| | - Yimei Zuo
- A Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan 411201, P. R. China
| | - Shu Chen
- A Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan 411201, P. R. China
| | - Yi Zhuo
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, Hunan Provincial Key Laboratory of Neurorestoratology, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410006, P.R. China
| | - Ming Lu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, Hunan Provincial Key Laboratory of Neurorestoratology, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410006, P.R. China
| | - Guoyue Shi
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory for Urban Ecological Processes and Eco-Restoration, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| | - Hui Gu
- A Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan 411201, P. R. China
| |
Collapse
|
26
|
Hunt J, Coulson EJ, Rajnarayanan R, Oster H, Videnovic A, Rawashdeh O. Sleep and circadian rhythms in Parkinson's disease and preclinical models. Mol Neurodegener 2022; 17:2. [PMID: 35000606 PMCID: PMC8744293 DOI: 10.1186/s13024-021-00504-w] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022] Open
Abstract
The use of animals as models of human physiology is, and has been for many years, an indispensable tool for understanding the mechanisms of human disease. In Parkinson's disease, various mouse models form the cornerstone of these investigations. Early models were developed to reflect the traditional histological features and motor symptoms of Parkinson's disease. However, it is important that models accurately encompass important facets of the disease to allow for comprehensive mechanistic understanding and translational significance. Circadian rhythm and sleep issues are tightly correlated to Parkinson's disease, and often arise prior to the presentation of typical motor deficits. It is essential that models used to understand Parkinson's disease reflect these dysfunctions in circadian rhythms and sleep, both to facilitate investigations into mechanistic interplay between sleep and disease, and to assist in the development of circadian rhythm-facing therapeutic treatments. This review describes the extent to which various genetically- and neurotoxically-induced murine models of Parkinson's reflect the sleep and circadian abnormalities of Parkinson's disease observed in the clinic.
Collapse
Affiliation(s)
- Jeremy Hunt
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Elizabeth J. Coulson
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | | | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | - Aleksandar Videnovic
- Movement Disorders Unit and Division of Sleep Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Oliver Rawashdeh
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Australia
| |
Collapse
|
27
|
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]
|
28
|
Martín-Montañez E, Valverde N, Ladrón de Guevara-Miranda D, Lara E, Romero-Zerbo YS, Millon C, Boraldi F, Ávila-Gámiz F, Pérez-Cano AM, Garrido-Gil P, Labandeira-Garcia JL, Santin LJ, Pavia J, Garcia-Fernandez M. Insulin-like growth factor II prevents oxidative and neuronal damage in cellular and mice models of Parkinson's disease. Redox Biol 2021; 46:102095. [PMID: 34418603 PMCID: PMC8379511 DOI: 10.1016/j.redox.2021.102095] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/05/2021] [Accepted: 08/05/2021] [Indexed: 01/03/2023] Open
Abstract
Oxidative distress and mitochondrial dysfunction, are key factors involved in the pathophysiology of Parkinson's disease (PD). The pleiotropic hormone insulin-like growth factor II (IGF-II) has shown neuroprotective and antioxidant effects in some neurodegenerative diseases. In this work, we demonstrate the protective effect of IGF-II against the damage induced by 1-methyl-4-phenylpyridinium (MPP+) in neuronal dopaminergic cell cultures and a mouse model of progressive PD. In the neuronal model, IGF-II counteracts the oxidative distress produced by MPP + protecting dopaminergic neurons. Improved mitochondrial function, increased nuclear factor (erythroid-derived 2)-like2 (NRF2) nuclear translocation along with NRF2-dependent upregulation of antioxidative enzymes, and modulation of mammalian target of rapamycin (mTOR) signalling pathway were identified as mechanisms leading to neuroprotection and the survival of dopaminergic cells. The neuroprotective effect of IGF-II against MPP + -neurotoxicity on dopaminergic neurons depends on the specific IGF-II receptor (IGF-IIr). In the mouse model, IGF-II prevents behavioural dysfunction and dopaminergic nigrostriatal pathway degeneration and mitigates neuroinflammation induced by MPP+. Our work demonstrates that hampering oxidative stress and normalising mitochondrial function through the interaction of IGF-II with its specific IGF-IIr are neuroprotective in both neuronal and mouse models. Thus, the modulation of the IGF-II/IGF-IIr signalling pathway may be a useful therapeutic approach for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Elisa Martín-Montañez
- Departamento de Farmacología y Pediatría, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Malaga, 29010, Spain
| | - Nadia Valverde
- Departamento de Farmacología y Pediatría, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Malaga, 29010, Spain; Departamento de Fisiología Humana, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Malaga, 29010, Spain
| | - David Ladrón de Guevara-Miranda
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Malaga, 29010, Spain
| | - Estrella Lara
- Departamento de Fisiología Humana, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Malaga, 29010, Spain
| | - Yanina S Romero-Zerbo
- Departamento de Fisiología Humana, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Malaga, 29010, Spain
| | - Carmelo Millon
- Departamento de Fisiología Humana, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Malaga, 29010, Spain
| | - Federica Boraldi
- Dipartimento di Scienze della Vita. Patologia Generale.Universita di Modena e Reggio Emilia. 41125, Italy
| | - Fabiola Ávila-Gámiz
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Malaga, 29010, Spain
| | - Ana M Pérez-Cano
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Malaga, 29010, Spain
| | - Pablo Garrido-Gil
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS) y Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED-Madrid). Universidad de Santiago de Compostela, 15782 Spain
| | - Jose Luis Labandeira-Garcia
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS) y Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED-Madrid). Universidad de Santiago de Compostela, 15782 Spain
| | - Luis J Santin
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Malaga, 29010, Spain
| | - Jose Pavia
- Departamento de Farmacología y Pediatría, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Malaga, 29010, Spain.
| | - Maria Garcia-Fernandez
- Departamento de Fisiología Humana, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Malaga, 29010, Spain.
| |
Collapse
|
29
|
Biagioni F, Vivacqua G, Lazzeri G, Ferese R, Iannacone S, Onori P, Morini S, D’Este L, Fornai F. Chronic MPTP in Mice Damage-specific Neuronal Phenotypes within Dorsal Laminae of the Spinal Cord. Neurotox Res 2021; 39:156-169. [PMID: 33206341 PMCID: PMC7936970 DOI: 10.1007/s12640-020-00313-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/06/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023]
Abstract
The neurotoxin 1-methyl, 4-phenyl, 1, 2, 3, 6-tetrahydropiridine (MPTP) is widely used to produce experimental parkinsonism. Such a disease is characterized by neuronal damage in multiple regions beyond the nigrostriatal pathway including the spinal cord. The neurotoxin MPTP damages spinal motor neurons. So far, in Parkinson's disease (PD) patients alpha-synuclein aggregates are described in the dorsal horn of the spinal cord. Nonetheless, no experimental investigation was carried out to document whether MPTP affects the sensory compartment of the spinal cord. Thus, in the present study, we investigated whether chronic exposure to small doses of MPTP (5 mg/kg/X2, daily, for 21 days) produces any pathological effect within dorsal spinal cord. This mild neurotoxic protocol produces a damage only to nigrostriatal dopamine (DA) axon terminals with no decrease in DA nigral neurons assessed by quantitative stereology. In these experimental conditions we documented a decrease in enkephalin-, calretinin-, calbindin D28K-, and parvalbumin-positive neurons within lamina I and II and the outer lamina III. Met-Enkephalin and substance P positive fibers are reduced in laminae I and II of chronically MPTP-treated mice. In contrast, as reported in PD patients, alpha-synuclein is markedly increased within spared neurons and fibers of lamina I and II after MPTP exposure. This is the first evidence that experimental parkinsonism produces the loss of specific neurons of the dorsal spinal cord, which are likely to be involved in sensory transmission and in pain modulation providing an experimental correlate for sensory and pain alterations in PD.
Collapse
Affiliation(s)
| | - Giorgio Vivacqua
- Integrated Research Center (PRAAB), Campus Biomedico University of Roma, Via Alvaro del Portillo 21, 00125 Roma, Italy
- Department of Anatomic, Histologic, Forensic and Locomotor Apparatus Sciences, Sapienza University of Roma, Via Alfonso Borelli 50, 00161 Roma, Italy
| | - Gloria Lazzeri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | | | - Simone Iannacone
- Department of Anatomic, Histologic, Forensic and Locomotor Apparatus Sciences, Sapienza University of Roma, Via Alfonso Borelli 50, 00161 Roma, Italy
| | - Paolo Onori
- Department of Anatomic, Histologic, Forensic and Locomotor Apparatus Sciences, Sapienza University of Roma, Via Alfonso Borelli 50, 00161 Roma, Italy
| | - Sergio Morini
- Integrated Research Center (PRAAB), Campus Biomedico University of Roma, Via Alvaro del Portillo 21, 00125 Roma, Italy
| | - Loredana D’Este
- Department of Anatomic, Histologic, Forensic and Locomotor Apparatus Sciences, Sapienza University of Roma, Via Alfonso Borelli 50, 00161 Roma, Italy
| | - Francesco Fornai
- I.R.C.C.S. Neuromed, via dell’Elettronica, Pozzilli, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| |
Collapse
|
30
|
El-Gamal M, Salama M, Collins-Praino LE, Baetu I, Fathalla AM, Soliman AM, Mohamed W, Moustafa AA. Neurotoxin-Induced Rodent Models of Parkinson's Disease: Benefits and Drawbacks. Neurotox Res 2021; 39:897-923. [PMID: 33765237 DOI: 10.1007/s12640-021-00356-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disorder, is characterized by cardinal motor impairments, including akinesia and tremor, as well as by a host of non-motor symptoms, including both autonomic and cognitive dysfunction. PD is associated with a death of nigral dopaminergic neurons, as well as the pathological spread of Lewy bodies, consisting predominantly of the misfolded protein alpha-synuclein. To date, only symptomatic treatments, such as levodopa, are available, and trials aiming to cure the disease, or at least halt its progression, have not been successful. Wong et al. (2019) suggested that the lack of effective therapy against neurodegeneration in PD might be attributed to the fact that the molecular mechanisms standing behind the dopaminergic neuronal vulnerability are still a major scientific challenge. Understanding these molecular mechanisms is critical for developing effective therapy. Thirty-five years ago, Calne and William Langston (1983) raised the question of whether biological or environmental factors precipitate the development of PD. In spite of great advances in technology and medicine, this question still lacks a clear answer. Only 5-15% of PD cases are attributed to a genetic mutation, with the majority of cases classified as idiopathic, which could be linked to exposure to environmental contaminants. Rodent models play a crucial role in understanding the risk factors and pathogenesis of PD. Additionally, well-validated rodent models are critical for driving the preclinical development of clinically translatable treatment options. In this review, we discuss the mechanisms, similarities and differences, as well as advantages and limitations of different neurotoxin-induced rat models of PD. In the second part of this review, we will discuss the potential future of neurotoxin-induced models of PD. Finally, we will briefly demonstrate the crucial role of gene-environment interactions in PD and discuss fusion or dual PD models. We argue that these models have the potential to significantly further our understanding of PD.
Collapse
Affiliation(s)
- Mohamed El-Gamal
- Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt. .,Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Mohamed Salama
- Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Global Brain Health Institute (GBHI), Trinity College Dublin (TCD), Dublin, Ireland
| | | | | | - Ahmed M Fathalla
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Amira M Soliman
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Wael Mohamed
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Mansoura, Egypt.,Department of Basic Medical Science, Kulliyyah of Medicine, International Islamic University, Kuantan, Pahang, Malaysia
| | - Ahmed A Moustafa
- School of Social Sciences and Psychology and Marcs Institute for Brain and Behaviour, Western Sydney University, Sydney, NSW, Australia.,Department of Human Anatomy and Physiology, the Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
31
|
Oda W, Fujita Y, Baba K, Mochizuki H, Niwa H, Yamashita T. Inhibition of repulsive guidance molecule-a protects dopaminergic neurons in a mouse model of Parkinson's disease. Cell Death Dis 2021; 12:181. [PMID: 33589594 PMCID: PMC7884441 DOI: 10.1038/s41419-021-03469-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 01/21/2021] [Accepted: 01/26/2021] [Indexed: 11/26/2022]
Abstract
Repulsive guidance molecule-a (RGMa), a glycosylphosphatidylinositol-anchored membrane protein, has diverse functions in axon guidance, cell patterning, and cell survival. Inhibition of RGMa attenuates pathological dysfunction in animal models of central nervous system (CNS) diseases including spinal cord injury, multiple sclerosis, and neuromyelitis optica. Here, we examined whether antibody-based inhibition of RGMa had therapeutic effects in a mouse model of Parkinson's disease (PD). We treated mice with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and found increased RGMa expression in the substantia nigra (SN). Intraventricular, as well as intravenous, administration of anti-RGMa antibodies reduced the loss of tyrosine hydroxylase (TH)-positive neurons and accumulation of Iba1-positive microglia/macrophages in the SN of MPTP-treated mice. Selective expression of RGMa in TH-positive neurons in the SN-induced neuronal loss/degeneration and inflammation, resulting in a progressive movement disorder. The pathogenic effects of RGMa overexpression were attenuated by treatment with minocycline, which inhibits microglia and macrophage activation. Increased RGMa expression upregulated pro-inflammatory cytokine expression in microglia. Our observations suggest that the upregulation of RGMa is associated with the PD pathology; furthermore, inhibitory RGMa antibodies are a potential therapeutic option.
Collapse
Affiliation(s)
- Wakana Oda
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Dental Anesthesiology, Graduate School of Dentistry, Osaka University, 1-8 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- World Premier International, Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kousuke Baba
- Department of Neurology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hitoshi Niwa
- Department of Dental Anesthesiology, Graduate School of Dentistry, Osaka University, 1-8 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- World Premier International, Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Graduate School of Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
32
|
Chaprov KD, Teterina EV, Roman AY, Ivanova TA, Goloborshcheva VV, Kucheryanu VG, Morozov SG, Lysikova EA, Lytkina OA, Koroleva IV, Popova NI, Antohin AI, Ovchinnikov RK, Kukharsky MS. Comparative Analysis of MPTP Neurotoxicity in Mice with a Constitutive Knockout of the α-Synuclein Gene. Mol Biol 2021. [DOI: 10.1134/s0026893321010039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
33
|
Cuevas E, Burks S, Raymick J, Robinson B, Gómez-Crisóstomo NP, Escudero-Lourdes C, Lopez AGG, Chigurupati S, Hanig J, Ferguson SA, Sarkar S. Tauroursodeoxycholic acid (TUDCA) is neuroprotective in a chronic mouse model of Parkinson's disease. Nutr Neurosci 2020; 25:1374-1391. [PMID: 33345721 DOI: 10.1080/1028415x.2020.1859729] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Parkinson's disease (PD) is a progressive motor disease of unknown etiology. Although neuroprotective ability of endogenous bile acid, tauroursodeoxycholic acid (TUDCA), shown in various diseases, including an acute model of PD,the potential therapeutic role of TUDCA in progressive models of PD that exhibit all aspects of PD has not been elucidated. In the present study, mice were assigned to one of four treatment groups: (1) Probenecid (PROB); (2) TUDCA, (3) MPTP + PROB (MPTPp); and (3) TUDCA + MPTPp. Methods: Markers for dopaminergic function, neuroinflammation, oxidative stress and autophagy were assessed using high performance liquid chromatography (HPLC), immunohistochemistry (IHC) and western blot (WB) methods. Locomotion was measured before and after treatments. Results: MPTPp decreased the expression of dopamine transporters (DAT) and tyrosine hydroxylase (TH), indicating dopaminergic damage, and induced microglial and astroglial activation as demonstrated by IHC analysis. MPTPp also decreased DA and its metabolites as demonstrated by HPLC analysis. Further, MPTPp-induced protein oxidation; increased LAMP-1 expression indicated autophagy and the promotion of alpha-synuclein (α-SYN) aggregation.. Discussion: Pretreatment with TUDCA protected against dopaminergic neuronal damage, prevented the microglial and astroglial activation, as well as the DA and DOPAC reductions caused by MPTPp. TUDCA by itself did not produce any significant change, with data similar to the negative control group. Pretreatment with TUDCA prevented protein oxidation and autophagy, in addition to inhibiting α-SYN aggregation. Although TUDCA pretreatment did not significantly affect locomotion, only acute treatment effects were measured, indicating more extensive assessments may be necessary to reveal potential therapeutic effects on behavior. Together, these results suggest that autophagy may be involved in the progression of PD and that TUDCA may attenuate these effects. The efficacy of TUDCA as a novel therapy in patients with PD clearly warrants further study.
Collapse
Affiliation(s)
- Elvis Cuevas
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR, USA
| | - Susan Burks
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR, USA
| | - James Raymick
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR, USA
| | - Bonnie Robinson
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR, USA
| | | | | | | | - Srinivasulu Chigurupati
- Office of Regulatory Affairs, Office of Regulatory Science, Food and Drug Administration, Rockville, MD, USA
| | - Joseph Hanig
- Office of Testing & Research, CDER/FDA, White Oak, MD, USA
| | - Sherry A Ferguson
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR, USA
| | - Sumit Sarkar
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR, USA
| |
Collapse
|
34
|
Park H, Chang KA. Therapeutic Potential of Repeated Intravenous Transplantation of Human Adipose-Derived Stem Cells in Subchronic MPTP-Induced Parkinson's Disease Mouse Model. Int J Mol Sci 2020; 21:ijms21218129. [PMID: 33143234 PMCID: PMC7663651 DOI: 10.3390/ijms21218129] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease, which is clinically and pathologically characterized by motor dysfunction and the loss of dopaminergic neurons in the substantia nigra, respectively. PD treatment with stem cells has long been studied by researchers; however, no adequate treatment strategy has been established. The results of studies so far have suggested that stem cell transplantation can be an effective treatment for PD. However, PD is a progressively deteriorating neurodegenerative disease that requires long-term treatment, and this has been insufficiently studied. Thus, we aimed to investigate the therapeutic potential of human adipose-derived stem cells (hASC) for repeated vein transplantation over long-term in an animal model of PD. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD model mice, hASCs were administered on the tail vein six times at two-week intervals. After the last injection of hASCs, motor function significantly improved. The number of dopaminergic neurons present in the nigrostriatal pathway was recovered using hASC transplantation. Moreover, the administration of hASC restored altered dopamine transporter expression and increased neurotrophic factors, such as brain-derived neurotrophic factor (BDNF) and glial cell-derived neurotrophic factor (GDNF), in the striatum. Overall, this study suggests that repeated intravenous transplantation of hASC may exert therapeutic effects on PD by restoring BDNF and GDNF expressions, protecting dopaminergic neurons, and maintaining the nigrostriatal pathway.
Collapse
Affiliation(s)
- Hyunjun Park
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Incheon 21936, Korea;
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea
| | - Keun-A Chang
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Incheon 21936, Korea;
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea
- Department of Pharmacology, College of Medicine, Gachon University, Incheon 21936, Korea
- Correspondence: ; Tel.: +82-32-899-6411
| |
Collapse
|
35
|
Tang H, Xu Y, Liu L, He L, Huang J, Pan J, He W, Wang Y, Yang X, Hou X, Xu K. Nogo-A/S1PR2 Signaling Pathway Inactivation Decreases Microvascular Damage and Enhances Microvascular Regeneration in PDMCI Mice. Neuroscience 2020; 449:21-34. [PMID: 33039527 DOI: 10.1016/j.neuroscience.2020.09.057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 09/05/2020] [Accepted: 09/28/2020] [Indexed: 11/19/2022]
Abstract
The incidence of mild cognitive impairment in Parkinson's disease (PDMCI) is as high as 18-55%. However, the pathological mechanism of PDMCI is not yet clear. Our previous research showed that microvascular pathology and chronic cerebral hypoperfusion participated in the occurrence and development of PDMCI. Nogo-A has been suggested to be a negative regulator of microvascular regeneration in the central nervous system. Moreover, few insights have illuminated the mechanisms of Nogo-A and microvascular pathology in PDMCI. Therefore, we hypothesized that Nogo-A might be involved in the negative regulation of PDMCI angiogenesis. In this study, C57BL/6J mice were injected with Nogo-A-specific short hairpin RNA (shRNA-Nogo-A) in the lateral ventricle and intraperitoneally injected with a combination of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and probenecid. Subjects were classified into the following five groups for the Morris water maze test: control (CON), CON + shRNA-GFP, CON + shRNA-Nogo-A, PDMCI, and PDMCI + shRNA-Nogo-A. Furthermore, blood-brain barrier (BBB) permeability, fluorescein isothiocyanate (FITC)-conjugated dextran, transmission electron microscopy (TEM), immunofluorescence and Western blot analyses were performed. The results showed that MPTP could cause spatial memory and behavioral impairment, significant microvascular impairment and increased Nogo-A expression. When Nogo-A expression was downregulated, the cognitive and microvascular impairments were alleviated, and the expression of sphingosine-1-phosphate receptor 2 (S1PR2) and the RhoA/ROCK signaling pathway were inhibited. These findings suggested that Nogo-A could bind to S1PR2, activate related signaling pathways, and lead to the inhibition of vascular remodeling in PDMCI mice. This study indicated that Nogo-A downregulation could mediate microvascular remodeling and provide further insights into the pathogenesis of PDMCI.
Collapse
Affiliation(s)
- Hongmei Tang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Yunxian Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China; Department of Sports and Health, Guangzhou Sport University, Guangzhou 510500, China
| | - Liru Liu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Lu He
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Jingyu Huang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Jing Pan
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China; Department of Sports and Health, Guangzhou Sport University, Guangzhou 510500, China
| | - Wenjie He
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Yuxin Wang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Xubo Yang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China; School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Xiaohui Hou
- Department of Sports and Health, Guangzhou Sport University, Guangzhou 510500, China; School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China.
| | - Kaishou Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510120, China.
| |
Collapse
|
36
|
Airavaara M, Parkkinen I, Konovalova J, Albert K, Chmielarz P, Domanskyi A. Back and to the Future: From Neurotoxin-Induced to Human Parkinson's Disease Models. ACTA ACUST UNITED AC 2020; 91:e88. [PMID: 32049438 DOI: 10.1002/cpns.88] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is an age-related neurodegenerative disorder characterized by motor symptoms such as tremor, slowness of movement, rigidity, and postural instability, as well as non-motor features like sleep disturbances, loss of ability to smell, depression, constipation, and pain. Motor symptoms are caused by depletion of dopamine in the striatum due to the progressive loss of dopamine neurons in the substantia nigra pars compacta. Approximately 10% of PD cases are familial arising from genetic mutations in α-synuclein, LRRK2, DJ-1, PINK1, parkin, and several other proteins. The majority of PD cases are, however, idiopathic, i.e., having no clear etiology. PD is characterized by progressive accumulation of insoluble inclusions, known as Lewy bodies, mostly composed of α-synuclein and membrane components. The cause of PD is currently attributed to cellular proteostasis deregulation and mitochondrial dysfunction, which are likely interdependent. In addition, neuroinflammation is present in brains of PD patients, but whether it is the cause or consequence of neurodegeneration remains to be studied. Rodents do not develop PD or PD-like motor symptoms spontaneously; however, neurotoxins, genetic mutations, viral vector-mediated transgene expression and, recently, injections of misfolded α-synuclein have been successfully utilized to model certain aspects of the disease. Here, we critically review the advantages and drawbacks of rodent PD models and discuss approaches to advance pre-clinical PD research towards successful disease-modifying therapy. © 2020 The Authors.
Collapse
Affiliation(s)
- Mikko Airavaara
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ilmari Parkkinen
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Julia Konovalova
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Katrina Albert
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Piotr Chmielarz
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Andrii Domanskyi
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
37
|
Surkov SA, Mingazov ER, Blokhin VE, Sturova AI, Gretskaya NM, Zinchenko GN, Bezuglov VV, Ugrumov MV. The Neuroprotective Effect of N-Docosahexaenoyldopamine on Degenerating Dopaminergic Neurons of the Mesencephalon. BIOL BULL+ 2020. [DOI: 10.1134/s1062359020050106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
38
|
Wang X, Sun X, Niu M, Zhang X, Wang J, Zhou C, Xie A. RAGE Silencing Ameliorates Neuroinflammation by Inhibition of p38-NF-κB Signaling Pathway in Mouse Model of Parkinson's Disease. Front Neurosci 2020; 14:353. [PMID: 32410941 PMCID: PMC7201072 DOI: 10.3389/fnins.2020.00353] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 03/24/2020] [Indexed: 12/14/2022] Open
Abstract
Accumulating evidence suggested that neuroinflammation played a crucial role in dopaminergic neuronal death in Parkinson's disease (PD). The receptor for advanced glycation end products (RAGE), a multi-ligand receptor of the immunoglobulin superfamily, has been proposed as a key molecule in the onset and sustainment of the inflammatory response. Engagement of RAGE contributed to neuroinflammation by upregulating nuclear factor-κB (NF-κB) as well as cytokines. The aim of the present study was to investigate the expression of RAGE in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice and elucidate the RAGE signal pathway involved in the inflammation. Results showed that RAGE protein and pro-inflammatory cytokines cyclooxygenase type 2 (COX-2) were upregulated in MPTP-treated mice. Further experiments showed that RAGE ablation inhibited phosphorylation of IκB and p38 and protected nigral dopaminergic neurons against cell death in the substantia nigra (SN). These results suggested that RAGE participated in the pathogenesis of PD by neuroinflammation and p38MAPK-NFκB signal pathway may be involved in the process. Moreover, interfering with RAGE signaling pathway may be a reasonable therapeutic option in slowing PD development and progression.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoxuan Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mengyue Niu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaona Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chang Zhou
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Anmu Xie
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
39
|
Tonelli Gombalová Z, Košuth J, Alexovič Matiašová A, Zrubáková J, Žežula I, Giallongo T, Di Giulio AM, Carelli S, Tomašková L, Daxnerová Z, Ševc J. Majority of cerebrospinal fluid‐contacting neurons in the spinal cord of
C57Bl/6N
mice is present in ectopic position unlike in other studied experimental mice strains and mammalian species. J Comp Neurol 2020; 528:2523-2550. [DOI: 10.1002/cne.24909] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Zuzana Tonelli Gombalová
- Institute of Biology and Ecology, Faculty of ScienceP.J. Šafárik University in Košice Košice Slovak Republic
| | - Ján Košuth
- Institute of Biology and Ecology, Faculty of ScienceP.J. Šafárik University in Košice Košice Slovak Republic
| | - Anna Alexovič Matiašová
- Institute of Biology and Ecology, Faculty of ScienceP.J. Šafárik University in Košice Košice Slovak Republic
| | - Jarmila Zrubáková
- Institute of Biology and Ecology, Faculty of ScienceP.J. Šafárik University in Košice Košice Slovak Republic
| | - Ivan Žežula
- Institute of Mathematics, Faculty of ScienceP.J. Šafárik University in Košice Košice Slovak Republic
| | - Toniella Giallongo
- Laboratories of Pharmacology, Department of Health SciencesUniversity of Milan Milan Italy
- Pediatric Clinical Research Center "Fondazione Romeo e Enrica Invernizzi", L. Sacco Department of Biomedical and Clinical ScienceUniversity of Milan Milan Italy
| | - Anna Maria Di Giulio
- Laboratories of Pharmacology, Department of Health SciencesUniversity of Milan Milan Italy
- Pediatric Clinical Research Center "Fondazione Romeo e Enrica Invernizzi", L. Sacco Department of Biomedical and Clinical ScienceUniversity of Milan Milan Italy
| | - Stephana Carelli
- Laboratories of Pharmacology, Department of Health SciencesUniversity of Milan Milan Italy
- Pediatric Clinical Research Center "Fondazione Romeo e Enrica Invernizzi", L. Sacco Department of Biomedical and Clinical ScienceUniversity of Milan Milan Italy
| | - Lenka Tomašková
- Institute of Biology and Ecology, Faculty of ScienceP.J. Šafárik University in Košice Košice Slovak Republic
| | - Zuzana Daxnerová
- Institute of Biology and Ecology, Faculty of ScienceP.J. Šafárik University in Košice Košice Slovak Republic
| | - Juraj Ševc
- Institute of Biology and Ecology, Faculty of ScienceP.J. Šafárik University in Košice Košice Slovak Republic
| |
Collapse
|
40
|
Historical Perspective: Models of Parkinson's Disease. Int J Mol Sci 2020; 21:ijms21072464. [PMID: 32252301 PMCID: PMC7177377 DOI: 10.3390/ijms21072464] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 01/02/2023] Open
Abstract
Parkinson’s disease (PD) is the most common movement disorder with motor and nonmotor signs. The current therapeutic regimen for PD is mainly symptomatic as the etio-pathophysiology has not been fully elucidated. A variety of animal models has been generated to study different aspects of the disease for understanding the pathogenesis and therapeutic development. The disease model can be generated through neurotoxin-based or genetic-based approaches in a wide range of animals such as non-human primates (NHP), rodents, zebrafish, Caenorhabditis (C.) elegans, and drosophila. Cellular-based disease model is frequently used because of the ease of manipulation and suitability for large-screen assays. In neurotoxin-induced models, chemicals such as 6-hydroxydopamine (6-OHDA), 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), rotenone, and paraquat are used to recapitulate the disease. Genetic manipulation of PD-related genes, such as α-Synuclein(SNCA), Leucine-rich repeat kinase 2 (LRRK2), Pten-Induced Kinase 1 (PINK1), Parkin(PRKN), and Protein deglycase (DJ-1) Are used in the transgenic models. An emerging model that combines both genetic- and neurotoxin-based methods has been generated to study the role of the immune system in the pathogenesis of PD. Here, we discuss the advantages and limitations of the different PD models and their utility for different research purposes.
Collapse
|
41
|
TLR4 deficiency has a protective effect in the MPTP/probenecid mouse model of Parkinson's disease. Acta Pharmacol Sin 2019; 40:1503-1512. [PMID: 31388087 PMCID: PMC7471440 DOI: 10.1038/s41401-019-0280-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 06/23/2019] [Indexed: 02/05/2023]
Abstract
Parkinson’s disease (PD) is a multifactorial disorder characterized by progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) and the presence of Lewy bodies (LBs) consisting of misfolded α-synuclein protein. The etiology of PD is still not clear but systemic inflammation is proved to trigger and exacerbate DA neurons degeneration. Toll-like receptor 4 (TLR4) is a pattern-recognition receptor (PRR) and plays a major role in promoting the host immune. TLR4-mediated signal pathways induce the release of many inflammatory cytokines. It is reasonable to hypothesize that TLR4 is the mediator in microglia contributing to the damage of DA neurons in the SNpc. In this study, we evaluated the role of TLR4 in the chronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/probenecid mouse model. Both TLR4-deficient and wild-type (WT) mice were injected with probenecid (250 mg/kg, i.p.) followed by injection of MPTP (25 mg/kg, s.c.) every 4 days for 10 times. From D43 to D47, the behavioral performance in pole test and wire hang test was assessed. Then the mice were euthanized, and SN and striatum were dissected out for biochemical tests. We showed that compared with MPTP-treated WT mice, TLR4 deficiency significantly attenuated MPTP-induced motor deficits and TH-protein expression reduction in SNpc and striatum, suppressed MPTP-induced α-synuclein abnormality and neuroinflammation mediated through oxidative stress, glial activation, NF-κB and the NLRP3 inflammasome signaling pathways. These findings highlight the neuroprotective effect of TLR4-pathways in the chronic MPTP-induced PD mouse model.
Collapse
|
42
|
Xu M, Bohlen JK, Moore C, Nipper MA, Finn DA, Jones CE, Lim MM, Meshul CK. Effects of sleep disruption on stress, nigrostriatal markers, and behavior in a chronic/progressive MPTP male mouse model of parkinsonism. J Neurosci Res 2019; 97:1706-1719. [PMID: 31535395 PMCID: PMC6801095 DOI: 10.1002/jnr.24520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/14/2019] [Accepted: 08/19/2019] [Indexed: 12/16/2022]
Abstract
Sleep complaints are an early clinical symptom of neurodegenerative disorders. Patients with Parkinson's disease (PD) experience sleep disruption (SD). The objective of this study was to determine if preexisting, chronic SD leads to a greater loss of tyrosine hydroxylase (TH) within the striatum and the substantia nigra following chronic/progressive exposure with the neurotoxin, 1-methyl-2-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Male mice underwent chronic SD for 4 weeks, then injected with vehicle (VEH) or increasing doses of MPTP for 4 weeks. There was a significant decrease in the plasma corticosterone levels in the MPTP group, an increase in the SD group, and a return to the VEH levels in the SD+MPTP group. Protein expression levels for TH in the striatum (terminals) and substantia nigra pars compacta (dopamine [DA] cell counts) revealed up to a 78% and 38% decrease, respectively, in the MPTP and SD+MPTP groups compared to their relevant VEH and SD groups. DA transporter protein expression increased in the striatum in the MPTP versus VEH group and in the SN/midbrain between the SD+MPTP and the VEH group. There was a main effect of MPTP on various gait measures (e.g., braking) relative to the SD or VEH groups. In the SD+MPTP group, there were no differences compared to the VEH group. Thus, SD, prior to administration of MPTP, has effects on serum corticosterone and gait but more importantly does not potentiate greater loss of TH within the nigrostriatal pathway compared to the MPTP group, suggesting that in PD patients with SD, there is no exacerbation of the DA cell loss.
Collapse
MESH Headings
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine
- Animals
- Corpus Striatum/enzymology
- Corpus Striatum/pathology
- Corticosterone/blood
- Disease Models, Animal
- Dopamine Plasma Membrane Transport Proteins/analysis
- Gait Disorders, Neurologic/etiology
- Gait Disorders, Neurologic/physiopathology
- Male
- Mice
- Mice, Inbred C57BL
- Nerve Tissue Proteins/analysis
- Oxidopamine/toxicity
- Parkinsonian Disorders/complications
- Parkinsonian Disorders/metabolism
- Single-Blind Method
- Sleep Disorders, Intrinsic/blood
- Sleep Disorders, Intrinsic/etiology
- Sleep Disorders, Intrinsic/physiopathology
- Stress, Physiological
- Substantia Nigra/enzymology
- Substantia Nigra/pathology
- Tyrosine 3-Monooxygenase/analysis
- Vesicular Monoamine Transport Proteins/analysis
Collapse
Affiliation(s)
- Mo Xu
- Research Services, VA Medical Center/Portland, OR
| | | | | | | | - Deborah A. Finn
- Research Services, VA Medical Center/Portland, OR
- Department of Behavioral Neuroscience, Oregon Heath & Science University
| | - Carolyn E. Jones
- Research Services, VA Medical Center/Portland, OR
- Department of Behavioral Neuroscience, Oregon Heath & Science University
| | - Miranda M. Lim
- Research Services, VA Medical Center/Portland, OR
- Department of Behavioral Neuroscience, Oregon Heath & Science University
- Department of Neurology, Oregon Health & Science University
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University
| | - Charles K. Meshul
- Research Services, VA Medical Center/Portland, OR
- Department of Behavioral Neuroscience, Oregon Heath & Science University
- Department of Pathology, Oregon Health & Science University
| |
Collapse
|
43
|
Wang LY, Yu X, Li XX, Zhao YN, Wang CY, Wang ZY, He ZY. Catalpol Exerts a Neuroprotective Effect in the MPTP Mouse Model of Parkinson's Disease. Front Aging Neurosci 2019; 11:316. [PMID: 31849636 PMCID: PMC6889905 DOI: 10.3389/fnagi.2019.00316] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/01/2019] [Indexed: 12/11/2022] Open
Abstract
The degeneration of dopaminergic (DA) neurons in Parkinson’s disease (PD) is related to inflammation and oxidative stress. Anti-inflammatory agents could reduce the risk or slow the progression of PD. Catalpol, an iridoid glycoside extracted from the roots of Rehmannia radix, has been reported to reduce the release of inflammatory factors and exert neuroprotective effects. 1-methyl-4-phenyl-1,2,3, 6-tetrahydropyridine (MPTP)-treated mice were used as the PD model and the roles of catalpol on DA neurons and its potential mechanism were investigated in this study. We found that catalpol administration mitigated the loss of DA neurons induced by MPTP and increased exploratory behavior along with tyrosine hydroxylase (TH) expression, which was accompanied by astrocyte and microglia activation. Importantly, catalpol administration significantly inhibited MPTP-triggered oxidative stress, restored growth-associated protein 43 (GAP43) and vascular endothelial growth factor (VEGF) levels. Further, we found that catalpol suppressed the activation of MKK4/JNK/c-Jun signaling, and reduced the pro-inflammatory factors and inflammasome in the mouse model of PD. Our results suggest that catalpol relieves MPTP-triggered oxidative stress, which may benefit to avoid the occurrence of chronic inflammatory reaction. Catalpol alleviates MPTP-triggered oxidative stress and thereby prevents neurodegenerative diseases-related inflammatory reaction, highlighting its therapeutic potential for the management of PD symptoms.
Collapse
Affiliation(s)
- Li-Yuan Wang
- Department of Neurology, the First Hospital of China Medical University, Shenyang, China
| | - Xin Yu
- Institute of Health Science, China Medical University, Shenyang, China
| | - Xiao-Xi Li
- Department of Neurology, the First Hospital of China Medical University, Shenyang, China
| | - Yi-Nan Zhao
- Department of Neurology, the First Hospital of China Medical University, Shenyang, China
| | - Chun-Yan Wang
- Institute of Health Science, China Medical University, Shenyang, China
| | - Zhan-You Wang
- Institute of Health Science, China Medical University, Shenyang, China
| | - Zhi-Yi He
- Department of Neurology, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
44
|
Vivacqua G, Biagioni F, Busceti CL, Ferrucci M, Madonna M, Ryskalin L, Yu S, D'Este L, Fornai F. Motor Neurons Pathology After Chronic Exposure to MPTP in Mice. Neurotox Res 2019; 37:298-313. [PMID: 31721049 DOI: 10.1007/s12640-019-00121-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/18/2019] [Accepted: 09/25/2019] [Indexed: 12/12/2022]
Abstract
The neurotoxin 1-methyl,4-phenyl-1,2,3,6-tetrahydropiridine (MPTP) is widely used to produce experimental parkinsonism in rodents and primates. Among different administration protocols, continuous or chronic exposure to small amounts of MPTP is reported to better mimic cell pathology reminiscent of Parkinson's disease (PD). Catecholamine neurons are the most sensitive to MPTP neurotoxicity; however, recent studies have found that MPTP alters the fine anatomy of the spinal cord including motor neurons, thus overlapping again with the spinal cord involvement documented in PD. In the present study, we demonstrate that chronic exposure to low amounts of MPTP (10 mg/kg daily, × 21 days) significantly reduces motor neurons in the ventral lumbar spinal cord while increasing α-synuclein immune-staining within the ventral horn. Spinal cord involvement in MPTP-treated mice extends to Calbindin D28 KDa immune-reactive neurons other than motor neurons within lamina VII. These results were obtained in the absence of significant reduction of dopaminergic cell bodies in the Substantia Nigra pars compacta, while a slight decrease was documented in striatal tyrosine hydroxylase immune-staining. Thus, the present study highlights neuropathological similarities between dopaminergic neurons and spinal motor neurons and supports the pathological involvement of spinal cord in PD and experimental MPTP-induced parkinsonism. Remarkably, the toxic threshold for motor neurons appears to be lower compared with nigral dopaminergic neurons following a chronic pattern of MPTP intoxication. This sharply contrasts with previous studies showing that MPTP intoxication produces comparable neuronal loss within spinal cord and Substantia Nigra.
Collapse
Affiliation(s)
- Giorgio Vivacqua
- Department of Anatomy, Histology, Forensic Medicine and Locomotor Sciences, Via A. Borelli 50, 00161, Rome, Italy
- Department of Neurobiology, Xuan Wu Hospital, Capital University of Medical Sciences, 45 Changchun St, Beijing, 100053, China
| | | | | | - Michela Ferrucci
- Department of Traslational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126, Pisa, Italy
| | | | - Larisa Ryskalin
- Department of Traslational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126, Pisa, Italy
| | - Shun Yu
- Department of Neurobiology, Xuan Wu Hospital, Capital University of Medical Sciences, 45 Changchun St, Beijing, 100053, China
| | - Loredana D'Este
- Department of Anatomy, Histology, Forensic Medicine and Locomotor Sciences, Via A. Borelli 50, 00161, Rome, Italy
| | - Francesco Fornai
- I.R.C.C.S. Neuromed, Via Atinense, 18, Pozzilli, Italy.
- Department of Traslational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126, Pisa, Italy.
| |
Collapse
|
45
|
Ahn S, Liu QF, Jang JH, Park J, Jeong HJ, Kim Y, Kim DH, Jeong G, Oh ST, Park SU, Cho SY, Park HJ, Jeon S. Gami-Chunggan Formula Prevents Motor Dysfunction in MPTP/p-Induced and A53T α-Synuclein Overexpressed Parkinson's Disease Mouse Model Though DJ-1 and BDNF Expression. Front Aging Neurosci 2019; 11:230. [PMID: 31555122 PMCID: PMC6724569 DOI: 10.3389/fnagi.2019.00230] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 08/13/2019] [Indexed: 12/25/2022] Open
Abstract
The Gami–Chunggan formula (GCF) is a modification of the Chunggan (CG) decoction, which has been used to treat movement disorders such as Parkinson’s disease (PD) in Traditional East Asian Medicine. To evaluate the neuroprotective effects of GCF in chronic PD animal models, we used either a 5-week treatment of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine with probenecid (MPTP/p) or the α-synuclein A53T overexpressed PD mouse model. C57BL/6 mice were treated with MPTP, in combination with probenecid, for 5 weeks. GCF was administered simultaneously with MPTP injection for 38 days. The A53T α-synuclein overexpressed mice were also fed with GCF for 60 days. Using behavioral readouts and western blot analyses, it was observed that GCF prevents motor dysfunction in the MPTP/p-induced and A53T α-synuclein overexpressed mice. Moreover, GCF inhibited the reduction of dopaminergic neurons in the substantia nigra (SN) and fibers in the striatum (ST) against MPTP/p challenge. The expression of DJ-1 was increased but that of α-synuclein was decreased in the SN of PD-like brains by GCF administration. In vitro experiments also showed that GCF inhibited 6-OHDA-induced neurotoxicity in SH-SY5Y neuroblastoma cell lines and that it did so to a greater degree than CG. Furthermore, GCF induced BDNF expression through phosphorylation of Akt, ERK, CREB, and AMPK in the SN of PD-like brains. Therefore, use of the herbal medicine GCF offers a potential remedy for neurodegenerative disorders, including Parkinson’s disease.
Collapse
Affiliation(s)
- Sora Ahn
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Quan Feng Liu
- Department of Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, Gyeongju, South Korea
| | - Jae-Hwan Jang
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Jeonghun Park
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Ha Jin Jeong
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists, Chonnam National University, Gwangju, South Korea
| | - Youngman Kim
- Research Institute, Dong Kwang Pharmaceutical Co., Ltd., Seoul, South Korea
| | - Dong-Hee Kim
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Guwon Jeong
- Research Institute, Dong Kwang Pharmaceutical Co., Ltd., Seoul, South Korea
| | - Seung Tack Oh
- Research Institute, Dong Kwang Pharmaceutical Co., Ltd., Seoul, South Korea
| | - Seong-Uk Park
- Stroke and Neurological Disorders Center, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, South Korea
| | - Seung-Yeon Cho
- Stroke and Neurological Disorders Center, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, South Korea
| | - Hi-Joon Park
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, Seoul, South Korea.,Studies of Translational Acupuncture Research (STAR), Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, Seoul, South Korea
| | - Songhee Jeon
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
46
|
Ahmadian E, Eftekhari A, Samiei M, Maleki Dizaj S, Vinken M. The role and therapeutic potential of connexins, pannexins and their channels in Parkinson's disease. Cell Signal 2019; 58:111-118. [DOI: 10.1016/j.cellsig.2019.03.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/10/2019] [Accepted: 03/10/2019] [Indexed: 02/07/2023]
|
47
|
Su Y, Deng MF, Xiong W, Xie AJ, Guo J, Liang ZH, Hu B, Chen JG, Zhu X, Man HY, Lu Y, Liu D, Tang B, Zhu LQ. MicroRNA-26a/Death-Associated Protein Kinase 1 Signaling Induces Synucleinopathy and Dopaminergic Neuron Degeneration in Parkinson's Disease. Biol Psychiatry 2019; 85:769-781. [PMID: 30718039 PMCID: PMC8861874 DOI: 10.1016/j.biopsych.2018.12.008] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/06/2018] [Accepted: 12/07/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND Death-associated protein kinase 1 (DAPK1) is a widely distributed serine/threonine kinase that is critical for cell death in multiple neurological disorders, including Alzheimer's disease and stroke. However, little is known about the role of DAPK1 in the pathogenesis of Parkinson's disease (PD), the second most common neurodegenerative disorder. METHODS We used Western blot and immunohistochemistry to evaluate the alteration of DAPK1. Quantitative polymerase chain reaction and fluorescence in situ hybridization were used to analyze the expression of microRNAs in PD mice and patients with PD. Rotarod, open field, and pole tests were used to evaluate the locomotor ability. Immunofluorescence, Western blot, and filter traps were used to evaluate synucleinopathy in PD mice. RESULTS We found that DAPK1 is posttranscriptionally upregulated by a reduction in microRNA-26a (miR-26a) caused by a loss of the transcription factor CCAAT enhancer-binding protein alpha. The overexpression of DAPK1 in PD mice is positively correlated with neuronal synucleinopathy. Suppressing miR-26a or upregulating DAPK1 results in synucleinopathy, dopaminergic neuron cell death, and motor disabilities in wild-type mice. In contrast, genetic deletion of DAPK1 in dopaminergic neurons by crossing DAT-Cre mice with DAPK1 floxed mice effectively rescues the abnormalities in mice with chronic MPTP treatment. We further showed that DAPK1 overexpression promotes PD-like phenotypes by direct phosphorylation of α-synuclein at the serine 129 site. Correspondingly, a cell-permeable competing peptide that blocks the phosphorylation of α-synuclein prevents motor disorders, synucleinopathy, and dopaminergic neuron loss in the MPTP mice. CONCLUSIONS miR-26a/DAPK1 signaling cascades are essential in the formation of the molecular and cellular pathologies in PD.
Collapse
Affiliation(s)
- Ying Su
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, P.R.China
| | - Man-Fei Deng
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, P.R.China,Department of Pathophysiology, Key lab of neurological disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P. R. China
| | - Wan Xiong
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, P.R.China,Department of Pathophysiology, Key lab of neurological disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P. R. China
| | - Ao-Ji Xie
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, P.R.China,Department of Pathophysiology, Key lab of neurological disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P. R. China
| | - Jifeng Guo
- Center for Medical Genetics, School of Life Science, Central South University; National Research Center for Geriatric Diseases, Xiangya Hospital, Changsha, Hunan 410078, China
| | - Zhi-Hou Liang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jian-Guo Chen
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, P.R.China
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, 02215, USA
| | - Youming Lu
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, P.R.China
| | - Dan Liu
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, P.R.China,Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P. R. China
| | - Beisha Tang
- National Research Center for Geriatric Diseases, Xiangya Hospital, and Center for Medical Genetics, School of Life Science, Central South University, Changsha, Hunan, China.
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China; National Research Center for Geriatric Diseases, Xiangya Hospital, and Center for Medical Genetics, School of Life Science, Central South University, Changsha, Hunan, China.
| |
Collapse
|
48
|
Neuroprotective effects of acetyl-l-carnitine (ALC) in a chronic MPTP-induced Parkinson's disease mouse model: Endothelial and microglial effects. Neurosci Lett 2019; 703:86-95. [PMID: 30890473 DOI: 10.1016/j.neulet.2019.03.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/26/2019] [Accepted: 03/11/2019] [Indexed: 01/27/2023]
Abstract
Parkinson's disease (PD) is a progressive motor disease with clinical features emerging due to degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNc), which project to the caudate putamen (CPu) where they release dopamine (DA). The current study investigated whether acetyl-l-carnitine (ALC) could ameliorate the pathology seen in an in vivoin vivo chronic 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced mouse model of PD. Four treatment groups were included: 1) CONTROL receiving probenecid (PROB; 250 mg/kg) only, 2) MPTP (25 mg/kg) + PROB, 3) MPTP + ALC (100 mg/kg), and 4) ALC alone. MPTP-induced losses in tyrosine hydroxylase and DA transporter immunoreactivity in the SNc and CPu were significantly reduced by ALC. HPLC data further suggests that decreases in CPu DA levels produced by MPTP were also attenuated by ALC. Additionally, microglial activation and astrocytic reactivity induced by MPTP were greatly reduced by ALC, indicating protection against neuroinflammation. Glucose transporter-1 and the tight junction proteins occludin and zonula occludins-1 were also protected from MPTP-induced down-regulation by ALC. Together, data suggest that in this model, ALC protects against MPTP-induced damage to endothelial cells and loss of DA neurons in the SNc and CPu, suggesting that ALC therapy may have the potential to slow or ameliorate the progression of PD pathology in a clinical setting.
Collapse
|
49
|
Lasbleiz C, Mestre-Francés N, Devau G, Luquin MR, Tenenbaum L, Kremer EJ, Verdier JM. Combining Gene Transfer and Nonhuman Primates to Better Understand and Treat Parkinson's Disease. Front Mol Neurosci 2019; 12:10. [PMID: 30804750 PMCID: PMC6378268 DOI: 10.3389/fnmol.2019.00010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/14/2019] [Indexed: 01/27/2023] Open
Abstract
Parkinson’s disease (PD) is a progressive CNS disorder that is primarily associated with impaired movement. PD develops over decades and is linked to the gradual loss of dopamine delivery to the striatum, via the loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). While the administration of L-dopa and deep brain stimulation are potent therapies, their costs, side effects and gradual loss of efficacy underlines the need to develop other approaches. Unfortunately, the lack of pertinent animal models that reproduce DA neuron loss and behavior deficits—in a timeline that mimics PD progression—has hindered the identification of alternative therapies. A complementary approach to transgenic animals is the use of nonhuman primates (NHPs) combined with the overexpression of disease-related genes using viral vectors. This approach may induce phenotypes that are not influenced by developmental compensation mechanisms, and that take into account the personality of animals. In this review article, we discuss the combination of gene transfer and NHPs to develop “genetic” models of PD that are suitable for testing therapeutic approaches.
Collapse
Affiliation(s)
- Christelle Lasbleiz
- MMDN, University of Montpellier, EPHE, INSERM, U1198, PSL University, Montpellier, France
| | - Nadine Mestre-Francés
- MMDN, University of Montpellier, EPHE, INSERM, U1198, PSL University, Montpellier, France
| | - Gina Devau
- MMDN, University of Montpellier, EPHE, INSERM, U1198, PSL University, Montpellier, France
| | | | - Liliane Tenenbaum
- Laboratory of Molecular Neurotherapies and NeuroModulation, Clinical Neuroscience Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Jean-Michel Verdier
- MMDN, University of Montpellier, EPHE, INSERM, U1198, PSL University, Montpellier, France
| |
Collapse
|
50
|
He X, Yang S, Zhang R, Hou L, Xu J, Hu Y, Xu R, Wang H, Zhang Y. Smilagenin Protects Dopaminergic Neurons in Chronic MPTP/Probenecid-Lesioned Parkinson's Disease Models. Front Cell Neurosci 2019; 13:18. [PMID: 30804756 PMCID: PMC6371654 DOI: 10.3389/fncel.2019.00018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/16/2019] [Indexed: 01/22/2023] Open
Abstract
Current therapies for Parkinson’s disease (PD) only offer limited symptomatic alleviation but fail to hamper the progress of the disease. Thus, it is imperative to establish new approaches aiming at protecting or reversing neurodegeneration in PD. Recent work elucidates whether smilagenin (abbreviated SMI), a steroidal sapogenin from traditional Chinese medicinal herbs, can take neuroprotective effect on dopaminergic neurons in a chronic model of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) conjuncted with probenecid mice. We reported for the first time that SMI significantly improved the locomotor ability of chronic MPTP/probenecid–lesioned mice. SMI increased the tyrosine hydroxylase (TH) positive and Nissl positive neuron number in the substantia nigra pars compacta (SNpc), augmented striatal DA and its metabolites concentration and elevated striatal dopamine transporter density (DAT). In addition, dopamine receptor D2R not D1R was down-regulated by MPTP/probenecid and slightly raised by SMI prevention. What’s more, we discovered that SMI markedly elevated striatal glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) protein levels in SMI prevented mice. And we found that SMI increased GDNF and BDNF mRNA level by promoting CREB phosphorylation in 1-methyl-4-phenylpyridimium (MPP+) treated SH-SY5Y cells. The results illustrated that SMI could prevent the impairment of dopaminergic neurons in chronic MPTP/probenecid-induced mouse model.
Collapse
Affiliation(s)
- Xuan He
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Shuangshuang Yang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Rui Zhang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Lina Hou
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Jianrong Xu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Yaer Hu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Rang Xu
- Scientific Research Center, Xinhua Hospital, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Hao Wang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Yongfang Zhang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| |
Collapse
|