1
|
Santoro M, Lam RK, Blumenfeld SE, Tan W, Ciari P, Chu EK, Saw NL, Rijsketic DR, Lin JS, Heifets BD, Shamloo M. Mapping of catecholaminergic denervation, neurodegeneration, and inflammation in 6-OHDA-treated Parkinson's disease mice. NPJ Parkinsons Dis 2025; 11:28. [PMID: 39934193 PMCID: PMC11814337 DOI: 10.1038/s41531-025-00872-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Efforts to develop disease-modifying treatments for Parkinson's disease (PD) have been hindered by the lack of animal models replicating all hallmarks of PD and the insufficient attention to extra-nigrostriatal regions pathologically critical for the prodromal appearance of non-motor symptoms. Among PD models, 6-hydroxydopamine (6-OHDA) infusion in mice has gained prominence since 2012, primarily focusing on the nigrostriatal region. This study characterized tyrosine hydroxylase-positive neuron and fiber loss across the brain following a unilateral 6-OHDA (20 µg) infusion into the dorsal striatum. Our analysis integrates immunolabeling, brain clearing (iDISCO+), light sheet microscopy, and computational methods, including fMRI and machine learning tools. We also examined sex differences, disease progression, neuroinflammatory responses, and pro-apoptotic signaling in nigrostriatal regions of C57BL/6 mice exposed to varying 6-OHDA dosages (5, 10, or 20 µg) followed by 1, 7, and 14 days of recovery. This comprehensive, spatiotemporal analysis of 6-OHDA-induced pathology was used to map the time course of neuronal degeneration and the onset of neuroinflammation.
Collapse
Affiliation(s)
- Matteo Santoro
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Rachel K Lam
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Sarah E Blumenfeld
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Weiqi Tan
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Peter Ciari
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Emily K Chu
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Nay L Saw
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Daniel Ryskamp Rijsketic
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jennifer S Lin
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
- Department of Bioengineering, Stanford University School of Medicine and School of Engineering, Stanford, CA, USA
| | - Boris D Heifets
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
2
|
Leser FS, Júnyor FDS, Pagnoncelli IB, Delgado AB, Medeiros I, Nóbrega ACC, Andrade BDS, de Lima MN, da Silva NE, Jacob L, Boyé K, Geraldo LHM, de Souza AMT, Maron-Gutierrez T, Castro-Faria-Neto H, Follmer C, Braga C, Neves GA, Eichmann A, Romão LF, Lima FRS. CCL21-CCR7 blockade prevents neuroinflammation and degeneration in Parkinson's disease models. J Neuroinflammation 2025; 22:31. [PMID: 39894839 PMCID: PMC11789347 DOI: 10.1186/s12974-024-03318-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/02/2024] [Indexed: 02/04/2025] Open
Abstract
Parkinson's disease (PD) is a progressive degenerative disease of the central nervous system associated with neuroinflammation and microglial cell activation. Chemokine signaling regulates neuron-glia communication and triggers a microglial inflammatory profile. Herein, we identified the neuronal chemokine CCL21 as a major cause of microglial cell imbalance through the CCR7 receptor pathway with therapeutic implications for PD. In humans, we found that CCL21 transcript expression was increased in dopaminergic neurons (DANs) of the substantia nigra in PD patients. CCL21 and CCR7 expressions were spatially associated with brain regional vulnerability to synucleinopathies, as well as with the expression of microglial activation, neuroinflammation, and degeneration-related genes. Also, in mouse models of PD, we showed that CCL21 was overexpressed in DANs in vivo and in vitro. Mechanistically, neuronal CCL21 was shown to regulate microglial cell migration, proliferation, and activation in a CCR7-dependent manner through both canonical (PI3K/AKT) and non-canonical (ERK1/2/JNK) signaling pathways. Finally, we demonstrated that navarixin, a clinically relevant chemokine inhibitor with high affinity for the CCR7 receptor, could block CCL21 effects on microglia and prevent neurodegeneration and behavioral deficits in two mouse models of PD induced with either α-synuclein oligomers (αSynO) or 3,4-dihydroxyphenylacetaldehyde (DOPAL). Altogether, our data indicate that navarixin blocks CCL21/CCR7-mediated neuron-microglia communication and could be used as a therapeutic strategy against PD.
Collapse
Affiliation(s)
- Felipe Saceanu Leser
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France
| | - Flavio de Souza Júnyor
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Iohanna Bianca Pagnoncelli
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Anna Beatriz Delgado
- Laboratory of Neurobiology Applied to Biomedicine, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Isabelle Medeiros
- Laboratory of Neurobiology Applied to Biomedicine, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Ana Clara Campanelli Nóbrega
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Brenda da Silva Andrade
- Laboratory of Molecular Pharmacology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Maiara Nascimento de Lima
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Nícolas Emanoel da Silva
- Laboratory Molecular Modeling & QSAR, Pharmaceutical Sciences Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Laurent Jacob
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France
| | - Kevin Boyé
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France
| | - Luiz Henrique Medeiros Geraldo
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France
- Department of Internal Medicine, Department of Cellular and Molecular Physiology, Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, 06510-3221, USA
| | - Alessandra Mendonça Teles de Souza
- Laboratory Molecular Modeling & QSAR, Pharmaceutical Sciences Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Tatiana Maron-Gutierrez
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Hugo Castro-Faria-Neto
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Cristian Follmer
- Laboratory of Physical Chemistry of Proteins and Peptides (Lafipp), Chemistry Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-909, Brazil
| | - Carolina Braga
- Núcleo Multidisciplinar de Pesquisas em Biologia, NUMPEX-Bio, Universidade Federal do Rio de Janeiro, Duque de Caxias, RJ, 25240-005, Brasil
| | - Gilda Angela Neves
- Laboratory of Molecular Pharmacology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Anne Eichmann
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center (PARCC), Paris, 75015, France.
- Department of Internal Medicine, Department of Cellular and Molecular Physiology, Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, 06510-3221, USA.
| | - Luciana Ferreira Romão
- Laboratory of Neurobiology Applied to Biomedicine, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Flavia Regina Souza Lima
- Laboratory of Glial Cell Biology, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21949-590, Brazil.
| |
Collapse
|
3
|
Prates‐Rodrigues M, Schweizer BLA, de Paula Gomes C, Ribeiro ÂM, Padovan‐Neto FE, Masini D, Lopes‐Aguiar C. Challenges and Opportunities in Exploring Non-Motor Symptoms in 6-Hydroxydopamine Models of Parkinson's Disease: A Systematic Review. J Neurochem 2025; 169:e70008. [PMID: 39901598 PMCID: PMC11791392 DOI: 10.1111/jnc.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/18/2024] [Accepted: 01/08/2025] [Indexed: 02/05/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the progressive loss of midbrain dopaminergic neurons, leading to motor symptoms such as tremors, rigidity, and bradykinesia. Non-motor symptoms, including depression, hyposmia, and sleep disturbances, often emerge in the early stages of PD, but their mechanisms remain poorly understood. The 6-hydroxydopamine (6-OHDA) rodent model is a well-established tool for preclinical research, replicating key motor and non-motor symptoms of PD. In this review, we systematically analyzed 135 studies that used 6-OHDA rodent models of PD to investigate non-motor symptoms. The review process adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Our analysis highlights the growing use of 6-OHDA PD models for experimental research of non-motor symptoms. It also reveals significant variability in methodologies, including choices of brain target, toxin dosage, lesion verification strategies, and behavioral assessment reporting. Factors that hinder reproducibility and comparability of findings across studies. We highlight the need for standardization in 6-OHDA-based models with particular emphasis on consistent evaluation of lesion extent and reporting of the co-occurrence of non-motor symptoms. By fostering methodological coherence, this framework aims to enhance the reproducibility, reliability, and translational value of 6-OHDA models in PD non-motor symptom research.
Collapse
Affiliation(s)
- Mateus Prates‐Rodrigues
- Departamento de Fisiologia e Biofísica, Laboratório de Neurociências Comportamental e Molecular (LANEC)Universidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Beatriz Lage Araújo Schweizer
- Departamento de Fisiologia e Biofísica, Laboratório de Neurociências Comportamental e Molecular (LANEC)Universidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Clara de Paula Gomes
- Departamento de Fisiologia e Biofísica, Laboratório de Neurociências Comportamental e Molecular (LANEC)Universidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Ângela Maria Ribeiro
- Departamento de Fisiologia e Biofísica, Laboratório de Neurociências Comportamental e Molecular (LANEC)Universidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Fernando E. Padovan‐Neto
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão PretoUniversity of São PauloRibeirão PretoSPBrazil
| | - Debora Masini
- Department of Biochemistry and BiophysicsStockholm UniversityStockholmSweden
| | - Cleiton Lopes‐Aguiar
- Departamento de Fisiologia e Biofísica, Laboratório de Neurociências Comportamental e Molecular (LANEC)Universidade Federal de Minas GeraisBelo HorizonteBrazil
| |
Collapse
|
4
|
Gimenez GA, Romijn M, van den Herik J, Meijer W, Eggers R, Hobo B, De Zeeuw CI, Canto CB, Verhaagen J, Carulli D. A Study on Potential Sources of Perineuronal Net-Associated Sema3A in Cerebellar Nuclei Reveals Toxicity of Non-Invasive AAV-Mediated Cre Expression in the Central Nervous System. Int J Mol Sci 2025; 26:819. [PMID: 39859534 PMCID: PMC11765860 DOI: 10.3390/ijms26020819] [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: 12/19/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Semaphorin 3A (Sema3A) is an axon guidance molecule, which is also abundant in the adult central nervous system (CNS), particularly in perineuronal nets (PNNs). PNNs are extracellular matrix structures that restrict plasticity. The cellular sources of Sema3A in PNNs are unknown. Most Sema3A-bearing neurons do not express Sema3A mRNA, suggesting that Sema3A may be released from other neurons. Another potential source of Sema3A is the choroid plexus. To identify sources of PNN-associated Sema3A, we focused on the cerebellar nuclei, which contain Sema3A+ PNNs. Cerebellar nuclei neurons receive prominent input from Purkinje cells (PCs), which express high levels of Sema3A mRNA. By using a non-invasive viral vector approach, we overexpressed Cre in PCs, the choroid plexus, or throughout the CNS of Sema3Afl/fl mice. Knocking out Sema3A in PCs or the choroid plexus was not sufficient to decrease the amount of PNN-associated Sema3A. Alternatively, knocking out Sema3A throughout the CNS induced a decrease in PNN-associated Sema3A. However, motor deficits, microgliosis, and neurodegeneration were observed, which were due to Cre toxicity. Our study represents the first attempt to unravel cellular sources of PNN-associated Sema3A and shows that non-invasive viral-mediated Cre expression throughout the CNS could lead to toxicity, complicating the interpretation of Cre-mediated Sema3A knock-out.
Collapse
Affiliation(s)
- Geoffrey-Alexander Gimenez
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
- Department of Cerebellar Coordination & Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (C.I.D.Z.); (C.B.C.)
| | - Maurits Romijn
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Joëlle van den Herik
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Wouter Meijer
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Ruben Eggers
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Barbara Hobo
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Chris I. De Zeeuw
- Department of Cerebellar Coordination & Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (C.I.D.Z.); (C.B.C.)
- Department of Neuroscience, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Cathrin B. Canto
- Department of Cerebellar Coordination & Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (C.I.D.Z.); (C.B.C.)
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Daniela Carulli
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| |
Collapse
|
5
|
Santoro M, Lam RK, Blumenfeld SE, Tan W, Ciari P, Chu EK, Saw NL, Rijsketic DR, Lin JS, Heifets BD, Shamloo M. Mapping of catecholaminergic denervation, neurodegeneration, and inflammation in 6-OHDA-treated Parkinson's disease mice. RESEARCH SQUARE 2024:rs.3.rs-5206046. [PMID: 39483924 PMCID: PMC11527254 DOI: 10.21203/rs.3.rs-5206046/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Efforts to develop disease-modifying treatments for Parkinson's disease (PD) have been hindered by the lack of animal models replicating all hallmarks of PD and the insufficient attention to extra-nigrostriatal regions pathologically critical for the prodromal appearance of non-motor symptoms. Among PD models, 6-hydroxydopamine (6-OHDA) infusion in mice has gained prominence since 2012, primarily focusing on the nigrostriatal region. This study characterized widespread tyrosine hydroxylase-positive neuron and fiber loss across the brain following a unilateral 6-OHDA (20 μg) infusion into the dorsal striatum. Our analysis integrates immunolabeling, brain clearing (iDISCO+), light sheet microscopy, and computational methods, including fMRI and machine learning tools. We also examined sex differences, disease progression, neuroinflammatory responses, and pro-apoptotic signaling in nigrostriatal regions of C57BL/6 mice exposed to varying 6-OHDA dosages (5, 10, or 20 μg). This comprehensive, spatiotemporal analysis of 6-OHDA-induced pathology may guide the future design of experimental PD studies and neurotherapeutic development.
Collapse
Affiliation(s)
| | | | | | - Weiqi Tan
- Stanford University School of Medicine
| | | | | | - Nay L Saw
- Stanford University School of Medicine
| | | | | | | | | |
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
|
Ferreira AFF, Ulrich H, Feng ZP, Sun HS, Britto LR. Neurodegeneration and glial morphological changes are both prevented by TRPM2 inhibition during the progression of a Parkinson's disease mouse model. Exp Neurol 2024; 377:114780. [PMID: 38649091 DOI: 10.1016/j.expneurol.2024.114780] [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: 01/17/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by dopaminergic neuron death and neuroinflammation. Emerging evidence points to the involvement of the transient receptor potential melastatin 2 (TRPM2) channel in neuron death and glial activation in several neurodegenerative diseases. However, the involvement of TRPM2 in PD and specifically its relation to the neuroinflammation aspect of the disease remains poorly understood. Here, we hypothesized that AG490, a TRPM2 inhibitor, can be used as a treatment in a mouse model of PD. Mice underwent stereotaxic surgery for 6-hydroxydopamine (6-OHDA) administration in the right striatum. Motor behavioral tests (apomorphine, cylinder, and rotarod) were performed on day 3 post-injection to confirm the PD model induction. AG490 was then daily injected i.p. between days 3 to 6 after surgery. On day 6, motor behavior was assessed again. Substantia nigra (SNc) and striatum (CPu) were collected for immunohistochemistry, immunoblotting, and RT-qPCR analysis on day 7. Our results revealed that AG490 post-treatment reduced motor behavior impairment and nigrostriatal neurodegeneration. In addition, the compound prevented TRPM2 upregulation and changes of the Akt/GSK-3β/caspase-3 signaling pathway. The TRPM2 inhibition also avoids the glial morphology changes observed in the PD group. Remarkably, the morphometrical analysis revealed that the ameboid-shaped microglia, found in 6-OHDA-injected animals, were no longer present in the AG490-treated group. These results indicate that AG490 treatment can reduce dopaminergic neuronal death and suppress neuroinflammation in a PD mouse model. Inhibition of TRPM2 by AG490 could then represent a potential therapeutical strategy to be evaluated for PD treatment.
Collapse
Affiliation(s)
- Ana Flavia F Ferreira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada.
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Luiz Roberto Britto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
8
|
Zhang X, Shen ZL, Ji YW, Yin C, Xiao C, Zhou C. Activation and polarization of striatal microglia and astrocytes are involved in bradykinesia and allodynia in early-stage parkinsonian mice. FUNDAMENTAL RESEARCH 2024; 4:806-819. [PMID: 39156564 PMCID: PMC11330119 DOI: 10.1016/j.fmre.2023.05.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/13/2023] [Accepted: 05/17/2023] [Indexed: 08/20/2024] Open
Abstract
In addition to the cardinal motor symptoms, pain is a major non-motor symptom of Parkinson's disease (PD). Neuroinflammation in the substantia nigra pars compacta and dorsal striatum is involved in neurodegeneration in PD. But the polarization of microglia and astrocytes in the dorsal striatum and their contribution to motor deficits and hyperalgesia in PD have not been characterized. In the present study, we observed that hemiparkinsonian mice established by unilateral 6-OHDA injection in the medial forebrain bundle exhibited motor deficits and mechanical allodynia. In these mice, both microglia and astrocytes in the dorsal striatum were activated and polarized to M1/M2 microglia and A1/A2 astrocytes as genes specific to these cells were upregulated. These effects peaked 7 days after 6-OHDA injection. Meanwhile, striatal astrocytes in parkinsonian mice also displayed hyperpolarized membrane potentials, enhanced voltage-gated potassium currents, and dysfunction in inwardly rectifying potassium channels and glutamate transporters. Systemic administration of minocycline, a microglia inhibitor, attenuated the expression of genes specific to M1 microglia and A1 astrocytes in the dorsal striatum (but not those specific to M2 microglia and A2 astrocytes), attenuated the damage in the nigrostriatal dopaminergic system, and alleviated the motor deficits and mechanical allodynia in parkinsonian mice. By contrast, local administration of minocycline into the dorsal striatum of parkinsonian mice mitigated only hyperalgesia. This study suggests that M1 microglia and A1 astrocytes in the dorsal striatum may play important roles in the development of pathophysiology underlying hyperalgesia in the early stages of PD.
Collapse
Affiliation(s)
- Xue Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Zi-Lin Shen
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Ya-Wei Ji
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Cui Yin
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Cheng Xiao
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Chunyi Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
9
|
Rodríguez-Pérez AI, Garrido-Gil P, García-Garrote M, Muñoz A, Parga JA, Labandeira-García JL, Rodríguez-Pallares J. Non-HLA angiotensin-type-1 receptor autoantibodies mediate the long-term loss of grafted neurons in Parkinson's disease models. Stem Cell Res Ther 2024; 15:138. [PMID: 38735991 PMCID: PMC11089721 DOI: 10.1186/s13287-024-03751-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/02/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND Clinical trials have provided evidence that transplants of dopaminergic precursors, which may be replaced by new in vitro stem cell sources, can integrate into the host tissue, and alleviate motor symptoms in Parkinson´s disease (PD). In some patients, deterioration of graft function occurred several months after observing a graft-derived functional improvement. Rejection of peripheral organs was initially related to HLA-specific antibodies. However, the role of non-HLA antibodies is now considered also relevant for rejection. Angiotensin-II type-1 receptor autoantibodies (AT1-AA) act as agonists of the AT1 receptors. AT1-AA are the non-HLA antibodies most widely associated with graft dysfunction or rejection after transplantation of different solid organs and hematopoietic stem cells. However, it is not known about the presence and possible functional effects of AT1-AA in dopaminergic grafts, and the effects of treatment with AT1 receptor blockers (ARBs) such as candesartan on graft survival. METHODS In a 6-hydroxydopamine PD rat model, we studied the short-term (10 days)- and long-term (3 months) effects of chronic treatment with the ARB candesartan on survival of grafted dopaminergic neurons and microglial graft infiltration, as well as the effects of dopaminergic denervation and grafting on serum and CSF AT1-AA levels. The expression of AT1 receptors in grafted neurons was determined by laser capture microdissection. RESULTS At the early period post-grafting, the number of grafted dopaminergic neurons that survived was not significantly different between treated and untreated hosts (i.e., control rats and rats treated with candesartan), probably because, just after grafting, other deleterious factors are predominant for dopaminergic cell death, such as mechanical trauma, lack of growth factors/nutrients and ischemia. However, several months post-grafting, we observed a significantly higher number of surviving dopaminergic neurons and a higher density of striatal dopaminergic terminals in the candesartan-treated group. For several months, grafted rats showed blood and cerebrospinal fluid levels of AT1-AA higher than normal controls, and also higher AT1-AA levels than non-grafted parkinsonian rats. CONCLUSIONS The results suggest the use of ARBs such as candesartan in PD patients, particularly before and after dopaminergic grafts, and the need to monitor AT1-AA levels in PD patients, particularly in those candidates for dopaminergic grafting.
Collapse
Affiliation(s)
- Ana I Rodríguez-Pérez
- Research Center for Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Pablo Garrido-Gil
- Research Center for Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Maria García-Garrote
- Research Center for Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ana Muñoz
- Research Center for Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Juan A Parga
- Research Center for Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jose Luis Labandeira-García
- Research Center for Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain.
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| | - Jannette Rodríguez-Pallares
- Research Center for Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain.
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| |
Collapse
|
10
|
Mendonça MD, da Silva JA, Hernandez LF, Castela I, Obeso J, Costa RM. Dopamine neuron activity encodes the length of upcoming contralateral movement sequences. Curr Biol 2024; 34:1034-1047.e4. [PMID: 38377999 PMCID: PMC10931818 DOI: 10.1016/j.cub.2024.01.067] [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: 02/13/2022] [Revised: 12/01/2023] [Accepted: 01/26/2024] [Indexed: 02/22/2024]
Abstract
Dopaminergic neurons (DANs) in the substantia nigra pars compacta (SNc) have been related to movement speed, and loss of these neurons leads to bradykinesia in Parkinson's disease (PD). However, other aspects of movement vigor are also affected in PD; for example, movement sequences are typically shorter. However, the relationship between the activity of DANs and the length of movement sequences is unknown. We imaged activity of SNc DANs in mice trained in a freely moving operant task, which relies on individual forelimb sequences. We uncovered a similar proportion of SNc DANs increasing their activity before either ipsilateral or contralateral sequences. However, the magnitude of this activity was higher for contralateral actions and was related to contralateral but not ipsilateral sequence length. In contrast, the activity of reward-modulated DANs, largely distinct from those modulated by movement, was not lateralized. Finally, unilateral dopamine depletion impaired contralateral, but not ipsilateral, sequence length. These results indicate that movement-initiation DANs encode more than a general motivation signal and invigorate aspects of contralateral movements.
Collapse
Affiliation(s)
- Marcelo D Mendonça
- Champalimaud Research, Champalimaud Foundation, 1400 038 Lisbon, Portugal; Champalimaud Clinical Centre, Champalimaud Foundation, 1400 038 Lisbon, Portugal; NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon 1169 056, Portugal
| | - Joaquim Alves da Silva
- Champalimaud Research, Champalimaud Foundation, 1400 038 Lisbon, Portugal; NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon 1169 056, Portugal
| | - Ledia F Hernandez
- HM CINAC, Centro Integral de Neurociencias AC, Fundación de Investigación HM Hospitales, Madrid 28938, Spain; Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Carlos III Institute of Health, Madrid 28029, Spain; Universidad CEU San Pablo, Madrid 28003, Spain
| | - Ivan Castela
- HM CINAC, Centro Integral de Neurociencias AC, Fundación de Investigación HM Hospitales, Madrid 28938, Spain; Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Carlos III Institute of Health, Madrid 28029, Spain; PhD Program in Neuroscience, Autonoma de Madrid University, Madrid 28029, Spain
| | - José Obeso
- HM CINAC, Centro Integral de Neurociencias AC, Fundación de Investigación HM Hospitales, Madrid 28938, Spain; Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Carlos III Institute of Health, Madrid 28029, Spain; Universidad CEU San Pablo, Madrid 28003, Spain; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Rui M Costa
- Champalimaud Research, Champalimaud Foundation, 1400 038 Lisbon, Portugal; Departments of Neuroscience and Neurology, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Allen Institute, Seattle, WA 98109, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
11
|
Athari SZ, Farajdokht F, Keyhanmanesh R, Mohaddes G. AMPK Signaling Pathway as a Potential Therapeutic Target for Parkinson's Disease. Adv Pharm Bull 2024; 14:120-131. [PMID: 38585465 PMCID: PMC10997932 DOI: 10.34172/apb.2024.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 09/30/2023] [Accepted: 10/08/2023] [Indexed: 04/09/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease caused by the loss of dopaminergic neurons. Genetic factors, inflammatory responses, oxidative stress, metabolic disorders, cytotoxic factors, and mitochondrial dysfunction are all involved in neuronal death in neurodegenerative diseases. The risk of PD can be higher in aging individuals due to decreased mitochondrial function, energy metabolism, and AMP-activated protein kinase (AMPK) function. The potential of AMPK to regulate neurodegenerative disorders lies in its ability to enhance antioxidant capacity, reduce oxidative stress, improve mitochondrial function, decrease mitophagy and macroautophagy, and inhibit inflammation. In addition, it has been shown that modulating the catalytic activity of AMPK can protect the nervous system. This article reviews the mechanisms by which AMPK activation can modulate PD.
Collapse
Affiliation(s)
- Seyed Zanyar Athari
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gisou Mohaddes
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biomedical Education, California Health Sciences University, College of Osteopathic Medicine, Clovis, CA, USA
| |
Collapse
|
12
|
Pan Z, Shao M, Zhao C, Yang X, Li H, Cui G, Liang X, Yu CW, Ye Q, Gao C, Di L, Chern JW, Zhou H, Lee SMY. J24335 exerts neuroprotective effects against 6-hydroxydopamine-induced lesions in PC12 cells and mice. Eur J Pharm Sci 2024; 194:106696. [PMID: 38199443 DOI: 10.1016/j.ejps.2024.106696] [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: 10/09/2023] [Revised: 01/06/2024] [Accepted: 01/07/2024] [Indexed: 01/12/2024]
Abstract
Parkinson's disease is the second most prevalent age-related neurodegenerative disease and disrupts the lives of people aged >60 years. Meanwhile, single-target drugs becoming inapplicable as PD pathogenesis diversifies. Mitochondrial dysfunction and neurotoxicity have been shown to be relevant to the pathogenesis of PD. The novel synthetic compound J24335 (11-Hydroxy-1-(8-methoxy-5-(trifluoromethyl)quinolin-2-yl)undecan-1-one oxime), which has been researched similarly to J2326, has the potential to be a multi-targeted drug and alleviate these lesions. Therefore, we investigated the mechanism of action and potential neuroprotective function of J24335 against 6-OHDA-induced neurotoxicity in mice, and in PC12 cell models. The key target of action of J24335 was also screened. MTT assay, LDH assay, flow cytometry, RT-PCR, LC-MS, OCR and ECAR detection, and Western Blot analysis were performed to characterize the neuroprotective effects of J24335 on PC12 cells and its potential mechanism. Behavioral tests and immunohistochemistry were used to evaluate behavioral changes and brain lesions in mice. Moreover, bioinformatics was employed to assess the drug-likeness of J24335 and screen its potential targets. J24335 attenuated the degradation of mitochondrial membrane potential and enhanced glucose metabolism and mitochondrial biosynthesis to ameliorate 6-OHDA-induced mitochondrial dysfunction. Animal behavioral tests demonstrated that J24335 markedly improved motor function and loss of TH-positive neurons and dopaminergic nerve fibers, and contributed to an increase in the levels of dopamine and its metabolites in brain tissue. The activation of both the CREB/PGC-1α/NRF-1/TFAM and PKA/Akt/GSK-3β pathways was a major contributor to the neuroprotective effects of J24335. Furthermore, bioinformatics predictions revealed that J24335 is a low toxicity and highly BBB permeable compound targeting 8 key genes (SRC, EGFR, ERBB2, SYK, MAPK14, LYN, NTRK1 and PTPN1). Molecular docking suggested a strong and stable binding between J24335 and the 8 core targets. Taken together, our results indicated that J24335, as a multi-targeted neuroprotective agent with promising therapeutic potential for PD, could protect against 6-OHDA-induced neurotoxicity via two potential pathways in mice and PC12 cells.
Collapse
Affiliation(s)
- Zhijian Pan
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Min Shao
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Chen Zhao
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Xuanjun Yang
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Haitao Li
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Guozhen Cui
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Xiaonan Liang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Chao-Wu Yu
- School of Pharmacy, National Taiwan University, Taipei 10050, Taiwan, China
| | - Qingqing Ye
- School of Pharmacy, National Taiwan University, Taipei 10050, Taiwan, China
| | - Cheng Gao
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Lijun Di
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Ji-Wang Chern
- School of Pharmacy, National Taiwan University, Taipei 10050, Taiwan, China
| | - Hefeng Zhou
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China.
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau; Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China.
| |
Collapse
|
13
|
Cui J, Zhao D, Xu M, Li Z, Qian J, Song N, Wang J, Xie J. Characterization of graded 6-Hydroxydopamine unilateral lesion in medial forebrain bundle of mice. Sci Rep 2024; 14:3721. [PMID: 38355892 PMCID: PMC10866897 DOI: 10.1038/s41598-024-54066-0] [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: 11/05/2023] [Accepted: 02/08/2024] [Indexed: 02/16/2024] Open
Abstract
Parkinson's disease (PD) is the second most common age-related neurodegenerative disease, with a progressive loss of dopaminergic cells and fibers. The purpose of this study was to use different doses of 6-hydroxydopamine (6-OHDA) injection into the medial forebrain bundle (MFB) of mice to mimic the different stages of the disease and to characterize in detail their motor and non-motor behavior, as well as neuropathological features in the nigrostriatal pathway. MFB were injected with 0.5 μg, 1 μg, 2 μg of 6-OHDA using a brain stereotaxic technique. 6-OHDA induced mitochondrial damage dose-dependently, as well as substantia nigra pars compacta (SNpc) tyrosine hydroxylase-positive (TH+) cell loss and striatal TH fiber loss. Activation of astrocytes and microglia in the SNpc and striatum were consistently observed at 7 weeks, suggesting a long-term glial response in the nigrostriatal system. Even with a partial or complete denervation of the nigrostriatal pathway, 6-OHDA did not cause anxiety, although depression-like behavior appeared. Certain gait disturbances were observed in 0.5 μg 6-OHDA lesioned mice, and more extensive in 1 μg group. Despite the loss of more neurons from 2 μg 6-OHDA, there was no further impairment in behaviors compared to 1 μg 6-OHDA. Our data have implications that 1 μg 6-OHDA was necessary and sufficient to induce motor and non-motor symptoms in mice, thus a valuable mouse tool to explore disease progression and new treatment in PD.
Collapse
Affiliation(s)
- Juntao Cui
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Di Zhao
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
| | - Manman Xu
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Zheheng Li
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Junliang Qian
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Ning Song
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China.
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China.
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Jun Wang
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China.
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China.
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Junxia Xie
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China.
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
14
|
Chernyshova EV, Potanina DV, Sadovnikova IS, Krutskikh EP, Volodina DE, Samoylova NA, Gureev AP. The study of the protective effect of mitochondrial uncouplers during acute toxicity of the fungicide difenoconazole in different organs of mice. BIOMEDITSINSKAIA KHIMIIA 2024; 70:41-51. [PMID: 38450680 DOI: 10.18097/pbmc20247001041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Pesticides represent a serious problem for agricultural workers due to their neurotoxic effects. The aim of this study was to evaluate the ability of pharmacological oxidative phosphorylation uncouplers to reduce the effect of the difenoconazole fungicide on mitochondrial DNA (mtDNA) of various organs in mice. Injections of difenoconazole caused cognitive deficits in mice, and the protonophore 2,4-dinitrophenol (2,4-DNP) and Azur I (AzI), a demethylated metabolite of methylene blue (MB), prevented the deterioration of cognitive abilities in mice induced by difenoconazole. Difenoconazole increased the rate of reactive oxygen species (ROS) production, likely through inhibition of complex I of the mitochondrial respiratory chain. After intraperitoneal administration of difenoconazole lungs, testes and midbrain were most sensitive to the accumulation of mtDNA damage. In contrast, the cerebral cortex and hippocampus were not tolerant to the effects of difenoconazole. The protonophore 2,4-DNP reduced the rate of ROS formation and significantly reduced the amount of mtDNA damage caused by difenoconazole in the midbrain, and partially, in the lungs and testes. MB, an alternative electron carrier capable of bypassing inhibited complex I, had no effect on the effect of difenoconazole on mtDNA, while its metabolite AzI, a demethylated metabolite of MB, was able to protect the mtDNA of the midbrain and testes. Thus, mitochondria-targeted therapy is a promising approach to reduce pesticide toxicity for agricultural workers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - A P Gureev
- Voronezh State University, Voronezh, Russia; Voronezh State University of Engineering Technologies, Voronezh, Russia
| |
Collapse
|
15
|
Shin W, Lee Y, Lim J, Lee Y, Lah JD, Lee S, Lee JU, Yu R, Lee PH, Lee JH, Kwak M, Cheon J. Nanoscale Magneto-mechanical-genetics of Deep Brain Neurons Reversing Motor Deficits in Parkinsonian Mice. NANO LETTERS 2024; 24:270-278. [PMID: 38157214 DOI: 10.1021/acs.nanolett.3c03899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Here, we introduce the magneto-mechanical-genetic (MMG)-driven wireless deep brain stimulation (DBS) using magnetic nanostructures for therapeutic benefits in the mouse model of Parkinson's disease (PD). Electrical DBS of the subthalamic nucleus (STN) is an effective therapy for mitigating Parkinson's motor symptoms. However, its broader application is hampered by the requirement for implanted electrodes and the lack of anatomical and cellular specificity. Using the nanoscale magnetic force actuators (m-Torquer), which deliver torque force under rotating magnetic fields to activate pre-encoded Piezo1 ion channels on target neurons, our system enables wireless and STN-specific DBS without implants, addressing key unmet challenges in the DBS field. In both late- and early-stage PD mice, MMG-DBS significantly improved locomotor activity and motor balance by 2-fold compared to untreated PD mice. Moreover, MMG-DBS enabled sustained therapeutic effects. This approach provides a non-invasive and implant-free DBS with cellular targeting capability for the effective treatment of Parkinsonian symptoms.
Collapse
Affiliation(s)
- Wookjin Shin
- Center for Nanomedicine, Institute for Basic Science, Seoul 03722, Republic of Korea
| | - Yeongdo Lee
- Center for Nanomedicine, Institute for Basic Science, Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering, Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Jueun Lim
- Center for Nanomedicine, Institute for Basic Science, Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering, Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Youbin Lee
- Center for Nanomedicine, Institute for Basic Science, Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering, Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Jungsu David Lah
- Center for Nanomedicine, Institute for Basic Science, Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering, Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Somin Lee
- Center for Nanomedicine, Institute for Basic Science, Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering, Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Jung-Uk Lee
- Center for Nanomedicine, Institute for Basic Science, Seoul 03722, Republic of Korea
| | - Ri Yu
- Center for Nanomedicine, Institute for Basic Science, Seoul 03722, Republic of Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul 03722, South Korea
- Department of Biomedical Science, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Jae-Hyun Lee
- Center for Nanomedicine, Institute for Basic Science, Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering, Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Minsuk Kwak
- Center for Nanomedicine, Institute for Basic Science, Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering, Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Jinwoo Cheon
- Center for Nanomedicine, Institute for Basic Science, Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering, Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
16
|
Athari SZ, Farajdokht F, Sadigh-Eteghad S, Mohajeri D, Nourazar MA, Mohaddes G. Hydroxychloroquine attenuated motor impairment and oxidative stress in a rat 6-hydroxydopamine model of Parkinson's disease. Int J Neurosci 2023; 133:1252-1261. [PMID: 35522252 DOI: 10.1080/00207454.2022.2074848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/27/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Parkinson's disease (PD) is associated with the destruction of dopaminergic neurons in the substantia nigra (SN). Hydroxychloroquine (HCQ) has the capability to cross the blood-brain barrier and promote a neuroprotective potential. This study evaluated the effects of HCQ on the 6-hydroxydopamine (6-OHDA)-induced PD model in rats. METHODS Wistar rats were randomly divided into sham, PD, PD + levodopa and PD + HCQ groups. The PD model was induced by a stereotactic administration of 6-OHDA into the left SN pars compacta (SNpc) and confirmed by rotation and the Murprogo's tests. HCQ (100 mg/kg, p.o.) and levodopa (12 mg/kg, p.o.) were administered once a day for 21 days. Three weeks after surgery, the behavioral tests were performed. Brain lipid peroxidation index (MDA), glutathione peroxidase activity (GPx), total antioxidant capacity (TAC) levels and α-synuclein protein expression in the SN were also measured. RESULTS The behavioral tests demonstrated that induction of PD increased the muscle rigidity and the number of rotations, which were reversed by HCQ treatment. Also, induction of PD was associated with an increase in α-synuclein protein levels and MDA and decreased TAC levels and GPx activity. However, HCQ decreased α-synuclein and MDA levels while increased TAC levels and GPx activity. In addition, histopathological data showed that HCQ protects dopaminergic neurons against 6-OHDA-induced toxicity. CONCLUSION According to the results, HCQ has a beneficial effect in improving PD-related pathophysiology, in part, by mitigating oxidative stress and protecting the dopaminergic neurons in the SN.
Collapse
Affiliation(s)
- Seyed Zanyar Athari
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Daryoush Mohajeri
- Department of Pathobiology, Faculty of Veterinary Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Mir Alireza Nourazar
- Department of Pathobiology, Faculty of Veterinary Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
- Department of Basic Sciences, Faculty of Veterinary Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Gisou Mohaddes
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Sharma R, Neupane C, Pham TL, Lee M, Lee S, Lee SY, Nam MH, Kim CS, Park JB. Tonic Activation of NR2D-Containing NMDARs Exacerbates Dopaminergic Neuronal Loss in MPTP-Injected Parkinsonian Mice. J Neurosci 2023; 43:7730-7744. [PMID: 37726169 PMCID: PMC10648527 DOI: 10.1523/jneurosci.1955-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 09/01/2023] [Accepted: 09/08/2023] [Indexed: 09/21/2023] Open
Abstract
NR2D subunit-containing NMDA receptors (NMDARs) gradually disappear during brain maturation but can be recruited by pathophysiological stimuli in the adult brain. Here, we report that 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) intoxication recruited NR2D subunit-containing NMDARs that generated an Mg2+-resistant tonic NMDA current (INMDA) in dopaminergic (DA) neurons in the midbrain of mature male mice. MPTP selectively generated an Mg2+-resistant tonic INMDA in DA neurons in the substantia nigra pars compacta (SNpc) and ventral tegmental area (VTA). Consistently, MPTP increased NR2D but not NR2B expression in the midbrain regions. Pharmacological or genetic NR2D interventions abolished the generation of Mg2+-resistant tonic INMDA in SNpc DA neurons, and thus attenuated subsequent DA neuronal loss and gait deficits in MPTP-treated mice. These results show that extrasynaptic NR2D recruitment generates Mg2+-resistant tonic INMDA and exacerbates DA neuronal loss, thus contributing to MPTP-induced Parkinsonism. The state-dependent NR2D recruitment could be a novel therapeutic target for mitigating cell type-specific neuronal death in neurodegenerative diseases.SIGNIFICANCE STATEMENT NR2D subunit-containing NMDA receptors (NMDARs) are widely expressed in the brain during late embryonic and early postnatal development, and then downregulated during brain maturation and preserved at low levels in a few regions of the adult brain. Certain stimuli can recruit NR2D subunits to generate tonic persistent NMDAR currents in nondepolarized neurons in the mature brain. Our results show that MPTP intoxication recruits NR2D subunits in midbrain dopaminergic (DA) neurons, which leads to tonic NMDAR current-promoting dopaminergic neuronal death and consequent abnormal gait behavior in the MPTP mouse model of Parkinson's disease (PD). This is the first study to indicate that extrasynaptic NR2D recruitment could be a target for preventing neuronal death in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ramesh Sharma
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - Chiranjivi Neupane
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - Thuy Linh Pham
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Miae Lee
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Sanghoon Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - So Yeong Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Cuk-Seong Kim
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Jin Bong Park
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| |
Collapse
|
18
|
Mendes-Pinheiro B, Campos J, Marote A, Soares-Cunha C, Nickels SL, Monzel AS, Cibrão JR, Loureiro-Campos E, Serra SC, Barata-Antunes S, Duarte-Silva S, Pinto L, Schwamborn JC, Salgado AJ. Treating Parkinson's Disease with Human Bone Marrow Mesenchymal Stem Cell Secretome: A Translational Investigation Using Human Brain Organoids and Different Routes of In Vivo Administration. Cells 2023; 12:2565. [PMID: 37947643 PMCID: PMC10650433 DOI: 10.3390/cells12212565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Parkinson's disease (PD) is the most common movement disorder, characterized by the progressive loss of dopaminergic neurons from the nigrostriatal system. Currently, there is no treatment that retards disease progression or reverses damage prior to the time of clinical diagnosis. Mesenchymal stem cells (MSCs) are one of the most extensively studied cell sources for regenerative medicine applications, particularly due to the release of soluble factors and vesicles, known as secretome. The main goal of this work was to address the therapeutic potential of the secretome collected from bone-marrow-derived MSCs (BM-MSCs) using different models of the disease. Firstly, we took advantage of an optimized human midbrain-specific organoid system to model PD in vitro using a neurotoxin-induced model through 6-hydroxydopamine (6-OHDA) exposure. In vivo, we evaluated the effects of BM-MSC secretome comparing two different routes of secretome administration: intracerebral injections (a two-site single administration) against multiple systemic administration. The secretome of BM-MSCs was able to protect from dopaminergic neuronal loss, these effects being more evident in vivo. The BM-MSC secretome led to motor function recovery and dopaminergic loss protection; however, multiple systemic administrations resulted in larger therapeutic effects, making this result extremely relevant for potential future clinical applications.
Collapse
Affiliation(s)
- Bárbara Mendes-Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Jonas Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Ana Marote
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Carina Soares-Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Sarah L. Nickels
- Luxembourg Centre for Systems and Biomedicine (LCSB), University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Anna S. Monzel
- Luxembourg Centre for Systems and Biomedicine (LCSB), University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Jorge R. Cibrão
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Eduardo Loureiro-Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Sofia C. Serra
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Sandra Barata-Antunes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Jens C. Schwamborn
- Luxembourg Centre for Systems and Biomedicine (LCSB), University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - António J. Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| |
Collapse
|
19
|
Muksuris K, Scarisbrick DM, Mahoney JJ, Cherkasova MV. Noninvasive Neuromodulation in Parkinson's Disease: Insights from Animal Models. J Clin Med 2023; 12:5448. [PMID: 37685514 PMCID: PMC10487610 DOI: 10.3390/jcm12175448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
The mainstay treatments for Parkinson's Disease (PD) have been limited to pharmacotherapy and deep brain stimulation. While these interventions are helpful, a new wave of research is investigating noninvasive neuromodulation methods as potential treatments. Some promising avenues have included transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), electroconvulsive therapy (ECT), and focused ultrasound (FUS). While these methods are being tested in PD patients, investigations in animal models of PD have sought to elucidate their therapeutic mechanisms. In this rapid review, we assess the available animal literature on these noninvasive techniques and discuss the possible mechanisms mediating their therapeutic effects based on these findings.
Collapse
Affiliation(s)
- Katherine Muksuris
- Department of Psychology, West Virginia University, Morgantown, WV 26506, USA
| | - David M. Scarisbrick
- Department of Behavioral Medicine and Psychiatry, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - James J. Mahoney
- Department of Behavioral Medicine and Psychiatry, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Mariya V. Cherkasova
- Department of Psychology, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
20
|
Dovonou A, Bolduc C, Soto Linan V, Gora C, Peralta Iii MR, Lévesque M. Animal models of Parkinson's disease: bridging the gap between disease hallmarks and research questions. Transl Neurodegener 2023; 12:36. [PMID: 37468944 PMCID: PMC10354932 DOI: 10.1186/s40035-023-00368-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by motor and non-motor symptoms. More than 200 years after its first clinical description, PD remains a serious affliction that affects a growing proportion of the population. Prevailing treatments only alleviate symptoms; there is still neither a cure that targets the neurodegenerative processes nor therapies that modify the course of the disease. Over the past decades, several animal models have been developed to study PD. Although no model precisely recapitulates the pathology, they still provide valuable information that contributes to our understanding of the disease and the limitations of our treatment options. This review comprehensively summarizes the different animal models available for Parkinson's research, with a focus on those induced by drugs, neurotoxins, pesticides, genetic alterations, α-synuclein inoculation, and viral vector injections. We highlight their characteristics and ability to reproduce PD-like phenotypes. It is essential to realize that the strengths and weaknesses of each model and the induction technique at our disposal are determined by the research question being asked. Our review, therefore, seeks to better aid researchers by ensuring a concrete discernment of classical and novel animal models in PD research.
Collapse
Affiliation(s)
- Axelle Dovonou
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Cyril Bolduc
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Victoria Soto Linan
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Charles Gora
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Modesto R Peralta Iii
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Martin Lévesque
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada.
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
21
|
Khan E, Hasan I, Haque ME. Parkinson's Disease: Exploring Different Animal Model Systems. Int J Mol Sci 2023; 24:ijms24109088. [PMID: 37240432 DOI: 10.3390/ijms24109088] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/30/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
Disease modeling in non-human subjects is an essential part of any clinical research. To gain proper understanding of the etiology and pathophysiology of any disease, experimental models are required to replicate the disease process. Due to the huge diversity in pathophysiology and prognosis in different diseases, animal modeling is customized and specific accordingly. As in other neurodegenerative diseases, Parkinson's disease is a progressive disorder coupled with varying forms of physical and mental disabilities. The pathological hallmarks of Parkinson's disease are associated with the accumulation of misfolded protein called α-synuclein as Lewy body, and degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNc) area affecting the patient's motor activity. Extensive research has already been conducted regarding animal modeling of Parkinson's diseases. These include animal systems with induction of Parkinson's, either pharmacologically or via genetic manipulation. In this review, we will be summarizing and discussing some of the commonly employed Parkinson's disease animal model systems and their applications and limitations.
Collapse
Affiliation(s)
- Engila Khan
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Ikramul Hasan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - M Emdadul Haque
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
22
|
Ye P, Bi L, Yang M, Qiu Y, Huang G, Liu Y, Hou Y, Li Z, Tong HHY, Cui M, Jin H. Activated Microglia in the Early Stage of a Rat Model of Parkinson's Disease: Revealed by PET-MRI Imaging by [ 18F]DPA-714 Targeting TSPO. ACS Chem Neurosci 2023. [PMID: 37146429 DOI: 10.1021/acschemneuro.3c00080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023] Open
Abstract
In the past decades, translocator protein (TSPO) has been considered as an in vivo biomarker to measure the presence of neuroinflammatory reactions. In this study, expression of TSPO was quantified via [18F]DPA-714 positron emission tomography-magnetic resonance imaging (PET-MRI) imaging to investigate the effects of microglial activation associated with motor behavioral impairments in the 6-hydroxydopamine (6-OHDA)-treated rodent model of Parkinson's disease (PD). [18F]FDG PET-MRI (for non-specific inflammation), [18F]D6-FP-(+)-DTBZ PET-MRI (for damaged dopaminergic neurons), post-PET immunofluorescence, and Pearson's correlation analyses were also performed. The time course of the striatal [18F]DPA-714 binding ratio elevated in 6-OHDA-treated rats during 1-3 weeks post-treatment, with the peak TSPO binding in the 1st week. No differences between bilateral striatum in [18F]FDG PET imaging were found. Moreover, an obvious correlation between [18F]DPA-714 SUVRR/L and rotation numbers was found (r = 0.434, *p = 0.049). No correlation between [18F]FDG SUVRR/L and rotation behavior was found. [18F]DPA-714 appeared to be a potential PET tracer for imaging the microglia-mediated neuroinflammation in the early stage of PD.
Collapse
Affiliation(s)
- Peizhen Ye
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Lei Bi
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Min Yang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Yifan Qiu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Guolong Huang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Yongshan Liu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Yuyi Hou
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Zhijun Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Henry Hoi Yee Tong
- Faculty of Health Sciences and Sports, Macao Polytechnic University, Macao SAR 999078, China
| | - Mengchao Cui
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Hongjun Jin
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| |
Collapse
|
23
|
Ye P, Bi L, Yang M, Qiu Y, Huang G, Liu Y, Hou Y, Li Z, Yee Tong HH, Cui M, Jin H. Activated Microglia in the Early Stage of a Rat Model of Parkinson's Disease: Revealed by PET-MRI Imaging by [ 18F]DPA-714 Targeting TSPO. ACS Chem Neurosci 2023. [PMID: 37134001 DOI: 10.1021/acschemneuro.3c00202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023] Open
Abstract
In the past decades, translocator protein (TSPO) has been considered as an in vivo biomarker to measure the presence of neuroinflammatory reactions. In this study, expression of TSPO was quantified via [18F]DPA-714 positron emission tomography-magnetic resonance imaging (PET-MRI) to investigate the effects of microglial activation associated with motor behavioral impairments in the 6-hydroxydopamine (6-OHDA)-treated rodent model of Parkinson's disease (PD). [18F]FDG PET-MRI (for non-specific inflammation), [18F]D6-FP-(+)-DTBZ PET-MRI (for damaged dopaminergic (DA) neurons), post-PET immunofluorescence, and Pearson's correlation analyses were also performed. The time course of striatal [18F]DPA-714 binding ratio was elevated in 6-OHDA-treated rats during 1-3 weeks post-treatments, with peak TSPO binding in the 1st week. No difference between the bilateral striatum in [18F]FDG PET imaging were found. Moreover, an obvious correlation between [18F]DPA-714 SUVRR/L and rotation numbers was found (r = 0.434, *p = 0.049). No correlation between [18F]FDG SUVRR/L and rotation behavior was found. [18F]DPA-714 appeared to be a potential PET tracer for imaging the microglia-mediated neuroinflammation in the early stage of PD.
Collapse
Affiliation(s)
- Peizhen Ye
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai City 519000, Guangdong Province, China
| | - Lei Bi
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai City 519000, Guangdong Province, China
| | - Min Yang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai City 519000, Guangdong Province, China
| | - Yifan Qiu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai City 519000, Guangdong Province, China
| | - Guolong Huang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai City 519000, Guangdong Province, China
| | - Yongshan Liu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai City 519000, Guangdong Province, China
| | - Yuyi Hou
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai City 519000, Guangdong Province, China
| | - Zhijun Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai City 519000, Guangdong Province, China
| | - Henry Hoi Yee Tong
- Faculty of Health Sciences and Sports, Macao Polytechnic University, Macao SAR 999078, China
| | - Mengchao Cui
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Hongjun Jin
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai City 519000, Guangdong Province, China
| |
Collapse
|
24
|
Scott KM, Chong YT, Park S, Wijeyekoon RS, Hayat S, Mathews RJ, Fitzpatrick Z, Tyers P, Wright G, Whitby J, Barker RA, Hu MT, Williams-Gray CH, Clatworthy MR. B lymphocyte responses in Parkinson's disease and their possible significance in disease progression. Brain Commun 2023; 5:fcad060. [PMID: 36993946 PMCID: PMC10042276 DOI: 10.1093/braincomms/fcad060] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 09/27/2022] [Accepted: 03/08/2023] [Indexed: 03/11/2023] Open
Abstract
Inflammation contributes to Parkinson's disease pathogenesis. We hypothesized that B lymphocytes are involved in Parkinson's disease progression. We measured antibodies to alpha-synuclein and tau in serum from patients with rapid eye movement sleep behaviour disorder (n = 79), early Parkinson's disease (n = 50) and matched controls (n = 50). Rapid eye movement sleep behaviour disorder cases were stratified by risk of progression to Parkinson's disease (low risk = 30, high risk = 49). We also measured B-cell activating factor of the tumour necrosis factor receptor family, C-reactive protein and total immunoglobulin G. We found elevated levels of antibodies to alpha-synuclein fibrils in rapid eye movement sleep behaviour disorder patients at high risk of Parkinson's disease conversion (ANOVA, P < 0.001) and lower S129D peptide-specific antibodies in those at low risk (ANOVA, P < 0.001). An early humoral response to alpha-synuclein is therefore detectable prior to the development of Parkinson's disease. Peripheral B lymphocyte phenotyping using flow cytometry in early Parkinson's disease patients and matched controls (n = 41 per group) revealed reduced B cells in Parkinson's disease, particularly in those at higher risk of developing an early dementia [t(3) = 2.87, P = 0.01]. Patients with a greater proportion of regulatory B cells had better motor scores [F(4,24) = 3.612, P = 0.019], suggesting they have a protective role in Parkinson's disease. In contrast, B cells isolated from Parkinson's disease patients at higher risk of dementia had greater cytokine (interleukin 6 and interleukin 10) responses following in vitro stimulation. We assessed peripheral blood lymphocytes in alpha-synuclein transgenic mouse models of Parkinson's disease: they also had reduced B cells, suggesting this is related to alpha-synuclein pathology. In a toxin-based mouse model of Parkinson's disease, B-cell deficiency or depletion resulted in worse pathological and behavioural outcomes, supporting the conclusion that B cells play an early protective role in dopaminergic cell loss. In conclusion, we found changes in the B-cell compartment associated with risk of disease progression in rapid eye movement sleep behaviour disorder (higher alpha-synuclein antibodies) and early Parkinson's disease (lower levels of B lymphocytes that were more reactive to stimulation). Regulatory B cells play a protective role in a mouse model, potentially by attenuating inflammation and dopaminergic cell loss. B cells are therefore likely to be involved in the pathogenesis of Parkinson's disease, albeit in a complex way, and thus warrant consideration as a therapeutic target.
Collapse
Affiliation(s)
- Kirsten M Scott
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0QH, UK
| | - Yen Ting Chong
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Seoyoung Park
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Ruwani S Wijeyekoon
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Shaista Hayat
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Rebeccah J Mathews
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0QH, UK
| | - Zachary Fitzpatrick
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0QH, UK
| | - Pam Tyers
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Georgia Wright
- University of Cambridge Clinical School of Medicine, Cambridge CB2 OQQ, UK
| | - Jennifer Whitby
- University of Cambridge Clinical School of Medicine, Cambridge CB2 OQQ, UK
| | - Roger A Barker
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Michele T Hu
- Division of Neurology, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Caroline H Williams-Gray
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Menna R Clatworthy
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0QH, UK
- Cellular Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| |
Collapse
|
25
|
Mousa HH, Sharawy MH, Nader MA. Empagliflozin enhances neuroplasticity in rotenone-induced parkinsonism: Role of BDNF, CREB and Npas4. Life Sci 2022; 312:121258. [PMID: 36462721 DOI: 10.1016/j.lfs.2022.121258] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022]
Abstract
AIMS Parkinsonism is characterized by degeneration of dopaminergic neurons and impairment in neuroplasticity. Empagliflozin (EMPA) is an anti-diabetic drug that has been shown to improve cognitive dysfunctions and exerted antioxidant and anti-inflammatory effects in different models. This study aimed to determine the neuroprotective effects of EMPA against rotenone (ROT)-induced parkinsonism. MAIN METHODS ROT (1.5 mg/kg) was injected subcutaneously three times per week for two successive weeks. Mice were treated with EMPA (3 and 10 mg/kg, orally) for one week prior ROT administration and for another two weeks along with ROT. After that, motor functions and histopathological changes were assessed, and brains were isolated for biochemical analyses and immunohistochemical investigation. KEY FINDINGS Results indicated that, in a dose dependent manner, EMPA improved motor functions and histopathological changes induced by ROT, increased brain content of reduced glutathione (GSH), dopamine (DA), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), nuclear factor erythroid 2-related factor 2 (Nrf2), inositol trisphosphate (IP3), calcium (Ca2+), calcium/calmodulin-dependent protein kinase type IV (CaMKIV) and phospho-Protein kinase B (p-Akt) levels compared to ROT group. Additionally, EMPA decreased the levels of malondialdehyde (MDA), and tumor necrosis factor-α (TNF-α), and inactivated glycogen synthase kinase-3 beta (GSK-3β). Improvement in neuroplasticity was also observed indicated by elevation in brain derived neurotrophic factor (BDNF), cAMP response element-binding protein (CREB), and neuronal PAS domain Protein 4 (Npas4). SIGNIFICANCE EMPA improved motor functions possibly through improving neuroplasticity markers and antioxidant, anti-inflammatory, and neuroprotective effects in a dose dependent manner.
Collapse
Affiliation(s)
- Hager H Mousa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Maha H Sharawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Manar A Nader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
26
|
Vijayanathan Y, Hamzah NM, Lim SM, Lim FT, Tan MP, Majeed ABA, Ramasamy K. Newly regenerated dopaminergic neurons in 6-OHDA-lesioned adult zebrafish brain proliferate in the Olfactory bulb and telencephalon, but migrate to, differentiate and mature in the diencephalon. Brain Res Bull 2022; 190:218-233. [PMID: 36228872 DOI: 10.1016/j.brainresbull.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 09/20/2022] [Accepted: 10/04/2022] [Indexed: 12/24/2022]
Abstract
In order to understand the biological processes underlying dopaminergic neurons (DpN) regeneration in a 6-hydroxydopamine(6-OHDA)-induced adult zebrafish-based Parkinson's disease model, this study investigated the specific phases of neuroregeneration in a time-based manner. Bromodeoxyuridine (BrdU) was administered 24 h before the harvest of brain tissues at day three, five, seven, nine, 12 and 14 postlesion. Potential migration of proliferative cells was tracked over 14 days postlesion through double-pulse tracking [BrdU and 5-ethynyl-2'-deoxyuridine (EdU)] of cells and immunohistostaining of astrocytes [glial fibrillary acidic protein (GFAP)]. Gene expression of foxa2 and nurr1 (nr4a2a) at day three, nine, 14, 18, 22 and 30 postlesion was quantified using qPCR. Protein expression of foxa2 at day three, seven, 14 and 22 postlesion was validated using the western blot technique. Double labelling [EdU and tyrosine hydroxylase (TH)] of proliferative cells was performed to ascertain their fate after the neuroregeneration processes. It was found that whilst cell proliferation remained unchanged in the area of substantial DpN loss, the ventral diencephalon (vDn), there was a transient increase of cell proliferation in the olfactory bulb (OB) and telencephalon (Tel) seven days postlesion. BrdU-immunoreactive (ir)/ EdU-ir cells and activated astrocytes were later found to be significantly increased in the vDn and its nearby area (Tel) 14 days postlesion. There was a significant but transient downregulation of foxa2 at day three and nine postlesion, and nr4a2a at day three, nine and 14 postlesion. The expression of both genes remained unchanged in the OB and Tel. There was a transient downregulation of foxa2 protein expression at day three and seven postlesion. The significant increase of EdU-ir/ TH-ir cells in the vDn 30 days postlesion indicates maturation of proliferative cells (formed between day five-seven postlesion) into DpN. The present findings warrant future investigation of critical factors that govern the distinctive phases of DpN regeneration.
Collapse
Affiliation(s)
- Yuganthini Vijayanathan
- Collaborative Drug Discovery Research (CDDR) Group and Brain Degeneration and Therapeutics Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia; Department of Medicine, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Naemah Md Hamzah
- Collaborative Drug Discovery Research (CDDR) Group and Brain Degeneration and Therapeutics Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Siong Meng Lim
- Collaborative Drug Discovery Research (CDDR) Group and Brain Degeneration and Therapeutics Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Fei Ting Lim
- Collaborative Drug Discovery Research (CDDR) Group and Brain Degeneration and Therapeutics Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Maw Pin Tan
- Department of Medicine, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Abu Bakar Abdul Majeed
- Collaborative Drug Discovery Research (CDDR) Group and Brain Degeneration and Therapeutics Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.
| | - Kalavathy Ramasamy
- Collaborative Drug Discovery Research (CDDR) Group and Brain Degeneration and Therapeutics Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
27
|
Liu J, Yang L, Li H, Cai Y, Feng J, Hu Z. Conditional ablation of protein tyrosine phosphatase receptor U in midbrain dopaminergic neurons results in reduced neuronal size. J Chem Neuroanat 2022; 124:102135. [DOI: 10.1016/j.jchemneu.2022.102135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/17/2022] [Accepted: 07/01/2022] [Indexed: 11/30/2022]
|
28
|
Gopinath A, Mackie P, Hashimi B, Buchanan AM, Smith AR, Bouchard R, Shaw G, Badov M, Saadatpour L, Gittis A, Ramirez-Zamora A, Okun MS, Streit WJ, Hashemi P, Khoshbouei H. DAT and TH expression marks human Parkinson's disease in peripheral immune cells. NPJ Parkinsons Dis 2022; 8:72. [PMID: 35672374 PMCID: PMC9174333 DOI: 10.1038/s41531-022-00333-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/11/2022] [Indexed: 12/26/2022] Open
Abstract
Parkinson's disease (PD) is marked by a loss of dopamine neurons, decreased dopamine transporter (DAT) and tyrosine hydroxylase (TH) expression. However, this validation approach cannot be used for diagnostic, drug effectiveness or investigational purposes in human patients because midbrain tissue is accessible postmortem. PD pathology affects both the central nervous and peripheral immune systems. Therefore, we immunophenotyped blood samples of PD patients for the presence of myeloid derived suppressor cells (MDSCs) and discovered that DAT+/TH+ monocytic MDSCs, but not granulocytic MDSCs are increased, suggesting a targeted immune response to PD. Because in peripheral immune cells DAT activity underlies an immune suppressive mechanism, we investigated whether expression levels of DAT and TH in the peripheral immune cells marks PD. We found drug naïve PD patients exhibit differential DAT+/TH+ expression in peripheral blood mononuclear cells (PBMCs) compared to aged/sex matched healthy subjects. While total PBMCs are not different between the groups, the percentage of DAT+/TH+ PBMCs was significantly higher in drug naïve PD patients compared to healthy controls irrespective of age, gender, disease duration, disease severity or treatment type. Importantly, treatment for PD negatively modulates DAT+/TH+ expressing PBMCs. Neither total nor the percentage of DAT+/TH+ PBMCs were altered in the Alzheimer's disease cohort. The mechanistic underpinning of this discovery in human PD was revealed when these findings were recapitulated in animal models of PD. The reverse translational experimental strategy revealed that alterations in dopaminergic markers in peripheral immune cells are due to the disease associated changes in the CNS. Our study demonstrates that the dopaminergic machinery on peripheral immune cells displays an association with human PD, with exciting implications in facilitating diagnosis and investigation of human PD pathophysiology.
Collapse
Affiliation(s)
- Adithya Gopinath
- Department of Neuroscience, University of Florida, Gainesville, FL, USA.
| | - Phillip Mackie
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Basil Hashimi
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | | | - Aidan R Smith
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | | | - Gerry Shaw
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
- EnCor Biotechnology, Inc, Gainesville, FL, USA
| | - Martin Badov
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Leila Saadatpour
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
- Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Aryn Gittis
- Carnegie Mellon University, Pittsburgh, PA, USA
| | - Adolfo Ramirez-Zamora
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, UF Health, Gainesville, FL, USA
| | - Michael S Okun
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, UF Health, Gainesville, FL, USA
| | - Wolfgang J Streit
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Parastoo Hashemi
- University of South Carolina, Columbia, SC, USA
- Department of Bioengineering, Imperial College, London, UK
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
29
|
Thomasi BBDM, Valdetaro L, Ricciardi MCG, Hayashide L, Fernandes ACMN, Mussauer A, da Silva ML, da Cunha Faria-Melibeu A, Ribeiro MGL, de Mattos Coelho-Aguiar J, Campello-Costa P, Moura-Neto V, Tavares-Gomes AL. Enteric glial cell reactivity in colonic layers and mucosal modulation in a mouse model of Parkinson’s disease induced by 6-hydroxydopamine. Brain Res Bull 2022; 187:111-121. [DOI: 10.1016/j.brainresbull.2022.06.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/20/2022] [Accepted: 06/25/2022] [Indexed: 11/02/2022]
|
30
|
Fieblinger T, Li C, Espa E, Cenci MA. Non-Apoptotic Caspase-3 Activation Mediates Early Synaptic Dysfunction of Indirect Pathway Neurons in the Parkinsonian Striatum. Int J Mol Sci 2022; 23:ijms23105470. [PMID: 35628278 PMCID: PMC9141690 DOI: 10.3390/ijms23105470] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/09/2022] [Accepted: 05/12/2022] [Indexed: 11/27/2022] Open
Abstract
Non-apoptotic caspase-3 activation is critically involved in dendritic spine loss and synaptic dysfunction in Alzheimer’s disease. It is, however, not known whether caspase-3 plays similar roles in other pathologies. Using a mouse model of clinically manifest Parkinson’s disease, we provide the first evidence that caspase-3 is transiently activated in the striatum shortly after the degeneration of nigrostriatal dopaminergic projections. This caspase-3 activation concurs with a rapid loss of dendritic spines and deficits in synaptic long-term depression (LTD) in striatal projection neurons forming the indirect pathway. Interestingly, systemic treatment with a caspase inhibitor prevents both the spine pruning and the deficit of indirect pathway LTD without interfering with the ongoing dopaminergic degeneration. Taken together, our data identify transient and non-apoptotic caspase activation as a critical event in the early plastic changes of indirect pathway neurons following dopamine denervation.
Collapse
Affiliation(s)
- Tim Fieblinger
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, 223 62 Lund, Sweden; (C.L.); (E.E.)
- University Medical Center Hamburg-Eppendorf, Institute for Synaptic Physiology, 20251 Hamburg, Germany
- Correspondence: (T.F.); (M.A.C.)
| | - Chang Li
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, 223 62 Lund, Sweden; (C.L.); (E.E.)
| | - Elena Espa
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, 223 62 Lund, Sweden; (C.L.); (E.E.)
| | - M. Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, 223 62 Lund, Sweden; (C.L.); (E.E.)
- Correspondence: (T.F.); (M.A.C.)
| |
Collapse
|
31
|
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
|
32
|
Gramage E, Sáiz J, Fernández-Calle R, Martín YB, Uribarri M, Ferrer-Alcón M, Barbas C, Herradón G. Metabolomics and biochemical alterations caused by pleiotrophin in the 6-hydroxydopamine mouse model of Parkinson's disease. Sci Rep 2022; 12:3577. [PMID: 35246557 PMCID: PMC8897456 DOI: 10.1038/s41598-022-07419-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/18/2022] [Indexed: 12/23/2022] Open
Abstract
Pleiotrophin (PTN) is a cytokine involved in nerve tissue repair processes, neuroinflammation and neuronal survival. PTN expression levels are upregulated in the nigrostriatal pathway of Parkinson's Disease (PD) patients. We aimed to characterize the dopaminergic injury and glial responses in the nigrostriatal pathway of mice with transgenic Ptn overexpression in the brain (Ptn-Tg) after intrastriatal injection of the catecholaminergic toxic 6-hydroxydopamine (6-OHDA) at a low dose (5 µg). Ten days after surgery, the injection of 6-OHDA induced a significant decrease of the number of tyrosine hydroxylase (TH)-positive neurons in the substantia nigra and of the striatal TH contents in Wild type (Wt) mice. In contrast, these effects of 6-OHDA were absent in Ptn-Tg mice. When the striatal Iba1 and GFAP immunoreactivity was studied, no statistical differences were found between vehicle-injected Wt and Ptn-Tg mice. Furthermore, 6-OHDA did not cause robust glial responses neither on Wt or Ptn-Tg mice 10 days after injections. In metabolomics studies, we detected interesting metabolites that significantly discriminate the more injured 6-OHDA-injected Wt striatum and the more protected 6-OHDA-injected Ptn-Tg striatum. Particularly, we detected groups of metabolites, mostly corresponding to phospholipids, whose trends were opposite in both groups. In summary, the data confirm lower 6-OHDA-induced decreases of TH contents in the nigrostriatal pathway of Ptn-Tg mice, suggesting a neuroprotective effect of brain PTN overexpression in this mouse model of PD. New lipid-related PD drug candidates emerge from this study and the data presented here support the increasingly recognized "lipid cascade" in PD.
Collapse
Affiliation(s)
- Esther Gramage
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Madrid, Spain
| | - Jorge Sáiz
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Madrid, Spain
| | - Rosalía Fernández-Calle
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Madrid, Spain
| | - Yasmina B Martín
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Madrid, Spain.,Departamento de Anatomía, Facultad de Medicina, Universidad Francisco de Vitoria, Ctra. Pozuelo-Majadahonda KM 1.800, 28223, Pozuelo de Alarcón, Madrid, Spain
| | - María Uribarri
- BRAINco Biopharma, S.L., Bizkaia Technology Park, Zamudio, Spain
| | | | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Madrid, Spain
| | - Gonzalo Herradón
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Madrid, Spain.
| |
Collapse
|
33
|
Masini D, Kiehn O. Targeted activation of midbrain neurons restores locomotor function in mouse models of parkinsonism. Nat Commun 2022; 13:504. [PMID: 35082287 PMCID: PMC8791953 DOI: 10.1038/s41467-022-28075-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 01/07/2022] [Indexed: 12/26/2022] Open
Abstract
The pedunculopontine nucleus (PPN) is a locomotor command area containing glutamatergic neurons that control locomotor initiation and maintenance. These motor actions are deficient in Parkinson’s disease (PD), where dopaminergic neurodegeneration alters basal ganglia activity. Being downstream of the basal ganglia, the PPN may be a suitable target for ameliorating parkinsonian motor symptoms. Here, we use in vivo cell-type specific PPN activation to restore motor function in two mouse models of parkinsonism made by acute pharmacological blockage of dopamine transmission. With a combination of chemo- and opto-genetics, we show that excitation of caudal glutamatergic PPN neurons can normalize the otherwise severe locomotor deficit in PD, whereas targeting the local GABAergic population only leads to recovery of slow locomotion. The motor rescue driven by glutamatergic PPN activation is independent of activity in nearby locomotor promoting glutamatergic Cuneiform neurons. Our observations point to caudal glutamatergic PPN neurons as a potential target for neuromodulatory restoration of locomotor function in PD. Here, the authors use cell-type specific stimulation of brainstem neurons within the caudal pedunculopontine nucleus to show that activation of excitatory neurons can normalize severe locomotor deficit in mouse models of parkinsonism. The study defines a potential target for neuromodulatory restoration of locomotor function in Parkinson’s disease.
Collapse
Affiliation(s)
- Débora Masini
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Ole Kiehn
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark. .,Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
34
|
Alberts T, Antipova V, Holzmann C, Hawlitschka A, Schmitt O, Kurth J, Stenzel J, Lindner T, Krause BJ, Wree A, Witt M. Olfactory Bulb D 2/D 3 Receptor Availability after Intrastriatal Botulinum Neurotoxin-A Injection in a Unilateral 6-OHDA Rat Model of Parkinson's Disease. Toxins (Basel) 2022; 14:94. [PMID: 35202123 PMCID: PMC8879205 DOI: 10.3390/toxins14020094] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 11/16/2022] Open
Abstract
Olfactory deficits occur as early non-motor symptoms of idiopathic Parkinson's disease (PD) in humans. The first central relay of the olfactory pathway, the olfactory bulb (OB), depends, among other things, on an intact, functional crosstalk between dopaminergic interneurons and dopamine receptors (D2/D3R). In rats, hemiparkinsonism (hemi-PD) can be induced by unilateral injection of 6-hydroxydopamine (6-OHDA) into the medial forebrain bundle (MFB), disrupting dopaminergic neurons of the substantia nigra pars compacta (SNpc). In a previous study, we showed that subsequent injection of botulinum neurotoxin-A (BoNT-A) into the striatum can reverse most of the pathological motor symptoms and normalize the D2/D3R availability. To determine whether this rat model is suitable to explain olfactory deficits that occur in humans with PD, we examined the availability of D2/D3R by longitudinal [18F]fallypride-PET/CT, the density of tyrosine hydroxylase immunoreactivity in the OB, olfactory performance by an orienting odor identification test adapted for rats, and a connectome analysis. PET/CT and immunohistochemical data remained largely unchanged after 6-OHDA lesion in experimental animals, suggesting that outcomes of the 6-OHDA hemi-PD rat model do not completely explain olfactory deficits in humans. However, after subsequent ipsilateral BoNT-A injection into the striatum, a significant 8.5% increase of the D2/D3R availability in the ipsilateral OB and concomitant improvement of olfactory performance were detectable. Based on tract-tracing meta-analysis, we speculate that this may be due to indirect connections between the striatum and the OB.
Collapse
Affiliation(s)
- Teresa Alberts
- Department of Anatomy, Rostock University Medical Center, D-18057 Rostock, Germany
| | - Veronica Antipova
- Department of Anatomy, Rostock University Medical Center, D-18057 Rostock, Germany
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Macroscopic and Clinical Anatomy, Medical University of Graz, A-8010 Graz, Austria
| | - Carsten Holzmann
- Department of Medical Genetics, Rostock University Medical Center, D-18057 Rostock, Germany
- Center of Transdisciplinary Neuroscience Rostock, D-18147 Rostock, Germany
| | | | - Oliver Schmitt
- Department of Anatomy, Rostock University Medical Center, D-18057 Rostock, Germany
| | - Jens Kurth
- Department of Nuclear Medicine, Rostock University Medical Center, D-18057 Rostock, Germany
| | - Jan Stenzel
- Core Facility Small Animal Imaging, Rostock University Medical Center, D-18057 Rostock, Germany
| | - Tobias Lindner
- Core Facility Small Animal Imaging, Rostock University Medical Center, D-18057 Rostock, Germany
| | - Bernd J Krause
- Center of Transdisciplinary Neuroscience Rostock, D-18147 Rostock, Germany
- Department of Nuclear Medicine, Rostock University Medical Center, D-18057 Rostock, Germany
| | - Andreas Wree
- Department of Anatomy, Rostock University Medical Center, D-18057 Rostock, Germany
- Center of Transdisciplinary Neuroscience Rostock, D-18147 Rostock, Germany
| | - Martin Witt
- Department of Anatomy, Rostock University Medical Center, D-18057 Rostock, Germany
- Center of Transdisciplinary Neuroscience Rostock, D-18147 Rostock, Germany
| |
Collapse
|
35
|
Ferreira AFF, Singulani MP, Ulrich H, Feng ZP, Sun HS, Britto LR. Inhibition of TRPM2 by AG490 Is Neuroprotective in a Parkinson's Disease Animal Model. Mol Neurobiol 2022; 59:1543-1559. [PMID: 35000153 DOI: 10.1007/s12035-022-02723-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/30/2021] [Indexed: 10/19/2022]
Abstract
Parkinson's disease (PD) is characterized by motor impairment and dopaminergic neuronal loss. There is no cure for the disease, and treatments have several limitations. The transient receptor potential melastatin 2 (TRPM2), a calcium-permeable non-selective cation channel, has been reported to be upregulated in neuronal death. However, there are no in vivo studies evaluating TRPM2's role and neuroprotective effects in PD. Here, we test the hypothesis that TRPM2 is upregulated in the 6-hydroxydopamine (6-OHDA) mouse model of PD and that its inhibition, by the AG490, is neuroprotective. For that, AG490 or vehicle were intraperitoneally administered into C57BL/6 mice. Mice then received 6-OHDA into the right striatum. Motor behavior assessments were evaluated 6, 13, and 20 days after surgery using the cylinder and apomorphine-induced rotational testes, and 7, 14, and 21 days after surgery using rotarod test. Brain samples of substantia nigra (SNc) and striatum (CPu) were collected for immunohistochemistry and immunoblotting on days 7 and 21. We showed that TRPM2 protein expression was upregulated in 6-OHDA-treated animals. In addition, AG490 prevented dopaminergic neuron loss, microglial activation, and astrocyte reactivity in 6-OHDA-treated animals. The compound improved motor behaviors and Akt/GSK-3β/caspase-3 signaling. We conclude that TRPM2 inhibition by AG490 is neuroprotective in the 6-OHDA model and that the TRPM2 channel may represent a potential therapeutic target for PD.
Collapse
Affiliation(s)
- Ana Flávia Fernandes Ferreira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Monique Patricio Singulani
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Laboratory of Neurosciences - LIM27, Department & Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada.,Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Luiz Roberto Britto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
36
|
Thamaraikani T, Karnam M, Velapandian C. In Silico Docking of Novel Phytoalkaloid Camalexin in the Management of Benomyl Induced Parkinson's Disease and its In Vivo Evaluation by Zebrafish Model. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 21:343-353. [PMID: 34477539 DOI: 10.2174/1871527320666210903091447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/29/2021] [Accepted: 07/12/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Parkinson's Disease (PD) exhibits the extrapyramidal symptoms caused due to the dopaminergic neuronal degeneration in the substantia nigra of the brain and depletion of Aldehyde Dehydrogenase (ALDH) enzyme. OBJECTIVE This study was designed to enlighten the importance of the Aldehyde dehydrogenase enzyme in protecting the dopamine levels in a living system. Camalexin, a potentially active compound, has been evaluated for its dopamine enhancing and aldehyde dehydrogenase protecting role in pesticide-induced Parkinson's disease. METHODS AutoDock 4.2 software was employed to perform the docking simulations between the ligand camalexin and standard drugs Alda-1, Ropirinole with three proteins 4WJR, 3INL, 5AER. Consequently, the compound was evaluated for its in vivo neuroprotective role in the zebrafish model by attaining Institutional Animal Ethical Committee permission. The behavioral assessments and catecholamine analysis in zebrafish were performed. RESULTS The Autodock result shows that the ligand camalexin has a lower binding energy (-3.84) that indicates a higher affinity with the proteins when compared to the standard drug of proteins (-3.42). In the zebrafish model, behavioral studies provided evidence that camalexin helps in the improvement of motor functions and cognition. The catecholamine assay has proved that there is an enhancement in dopamine levels, as well as an improvement in aldehyde dehydrogenase enzyme. CONCLUSION The novel compound, camalexin, offers a protective role in Parkinson's disease model by its interaction with neurochemical proteins and also in alternative in vivo model.
Collapse
Affiliation(s)
- Tamilanban Thamaraikani
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur 603203, Tamilnadu, India
| | - Manasa Karnam
- Department of Pharmacology, SRM College of Pharmacy,SRMIST, Kattankulathur-603203,Tamilnadu, India
| | - Chitra Velapandian
- Department of Pharmacology, SRM College of Pharmacy,SRMIST, Kattankulathur-603203,Tamilnadu, India
| |
Collapse
|
37
|
Pereira Luppi M, Azcorra M, Caronia-Brown G, Poulin JF, Gaertner Z, Gatica S, Moreno-Ramos OA, Nouri N, Dubois M, Ma YC, Ramakrishnan C, Fenno L, Kim YS, Deisseroth K, Cicchetti F, Dombeck DA, Awatramani R. Sox6 expression distinguishes dorsally and ventrally biased dopamine neurons in the substantia nigra with distinctive properties and embryonic origins. Cell Rep 2021; 37:109975. [PMID: 34758317 PMCID: PMC8607753 DOI: 10.1016/j.celrep.2021.109975] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 09/15/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Dopamine (DA) neurons in the ventral tier of the substantia nigra pars compacta (SNc) degenerate prominently in Parkinson's disease, while those in the dorsal tier are relatively spared. Defining the molecular, functional, and developmental characteristics of each SNc tier is crucial to understand their distinct susceptibility. We demonstrate that Sox6 expression distinguishes ventrally and dorsally biased DA neuron populations in the SNc. The Sox6+ population in the ventral SNc includes an Aldh1a1+ subset and is enriched in gene pathways that underpin vulnerability. Sox6+ neurons project to the dorsal striatum and show activity correlated with acceleration. Sox6- neurons project to the medial, ventral, and caudal striatum and respond to rewards. Moreover, we show that this adult division is encoded early in development. Overall, our work demonstrates a dual origin of the SNc that results in DA neuron cohorts with distinct molecular profiles, projections, and functions.
Collapse
Affiliation(s)
- Milagros Pereira Luppi
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maite Azcorra
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Giuliana Caronia-Brown
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jean-Francois Poulin
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Zachary Gaertner
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Serafin Gatica
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Navid Nouri
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Marilyn Dubois
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Yongchao C Ma
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Lief Fenno
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Yoon Seok Kim
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Francesca Cicchetti
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Daniel A Dombeck
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
| | - Rajeshwar Awatramani
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
38
|
Mendes-Pinheiro B, Soares-Cunha C, Marote A, Loureiro-Campos E, Campos J, Barata-Antunes S, Monteiro-Fernandes D, Santos D, Duarte-Silva S, Pinto L, José Salgado A. Unilateral Intrastriatal 6-Hydroxydopamine Lesion in Mice: A Closer Look into Non-Motor Phenotype and Glial Response. Int J Mol Sci 2021; 22:ijms222111530. [PMID: 34768962 PMCID: PMC8584172 DOI: 10.3390/ijms222111530] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/20/2022] Open
Abstract
Parkinson’s disease (PD) is a prevalent movement disorder characterized by the progressive loss of dopaminergic neurons in substantia nigra pars compacta (SNpc). The 6-hydroxydopamine (6-OHDA) lesion is still one of the most widely used techniques for modeling Parkinson’s disease (PD) in rodents. Despite commonly used in rats, it can be challenging to reproduce a similar lesion in mice. Moreover, there is a lack of characterization of the extent of behavioral deficits and of the neuronal loss/neurotransmitter system in unilateral lesion mouse models. In this study, we present an extensive behavioral and histological characterization of a unilateral intrastriatal 6-OHDA mouse model. Our results indicate significant alterations in balance and fine motor coordination, voluntary locomotion, and in the asymmetry’s degree of forelimb use in 6-OHDA lesioned animals, accompanied by a decrease in self-care and motivational behavior, common features of depressive-like symptomatology. These results were accompanied by a decrease in tyrosine hydroxylase (TH)-labelling and dopamine levels within the nigrostriatal pathway. Additionally, we also identify a marked astrocytic reaction, as well as proliferative and reactive microglia in lesioned areas. These results confirm the use of unilateral intrastriatal 6-OHDA mice for the generation of a mild model of nigrostriatal degeneration and further evidences the recapitulation of key aspects of PD, thereby being suitable for future studies beholding new therapeutical interventions for this disease.
Collapse
Affiliation(s)
- Bárbara Mendes-Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Carina Soares-Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Ana Marote
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Eduardo Loureiro-Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Jonas Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Sandra Barata-Antunes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Daniela Monteiro-Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Diogo Santos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - António José Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
- Correspondence: ; Tel.: +351-253-60-49-47
| |
Collapse
|
39
|
Kuter KZ, Olech Ł, Głowacka U, Paleczna M. Increased Beta-Hydroxybutyrate Level Is Not Sufficient for the Neuroprotective Effect of Long-Term Ketogenic Diet in an Animal Model of Early Parkinson's Disease. Exploration of Brain and Liver Energy Metabolism Markers. Int J Mol Sci 2021; 22:ijms22147556. [PMID: 34299176 PMCID: PMC8307513 DOI: 10.3390/ijms22147556] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 12/25/2022] Open
Abstract
The benefits of a ketogenic diet in childhood epilepsy steered up hope for neuroprotective effects of hyperketonemia in Parkinson’s disease (PD). There are multiple theoretical reasons but very little actual experimental proof or clinical trials. We examined the long-term effects of the ketogenic diet in an animal model of early PD. A progressive, selective dopaminergic medium size lesion was induced by 6-OHDA injection into the medial forebrain bundle. Animals were kept on the stringent ketogenic diet (1% carbohydrates, 8% protein, 70% fat) for 3 weeks prior and 4 weeks after the brain operation. Locomotor activity, neuron count, dopaminergic terminal density, dopamine level, and turnover were analyzed at three time-points post-lesion, up to 4 weeks after the operation. Energy metabolism parameters (glycogen, mitochondrial complex I and IV, lactate, beta-hydroxybutyrate, glucose) were analyzed in the brain and liver or plasma. Protein expression of enzymes essential for gluconeogenesis (PEPCK, G6PC) and glucose utilization (GCK) was analyzed in the liver. Despite long-term hyperketonemia pre- and post-lesion, the ketogenic diet did not protect against 6-OHDA-induced dopaminergic neuron lesions. The ketogenic diet only tended to improve locomotor activity and normalize DA turnover in the striatum. Rats fed 7 weeks in total with a restrictive ketogenic diet maintained normoglycemia, and neither gluconeogenesis nor glycogenolysis in the liver was responsible for this effect. Therefore, potentially, the ketogenic diet could be therapeutically helpful to support the late compensatory mechanisms active via glial cells but does not necessarily act against the oxidative stress-induced parkinsonian neurodegeneration itself. A word of caution is required as the stringent ketogenic diet itself also carries the risk of unwanted side effects, so it is important to study the long-term effects of such treatments. More detailed metabolic long-term studies using unified diet parameters are required, and human vs. animal differences should be taken under consideration.
Collapse
|
40
|
Lashuel HA, Novello S. Lewy body-associated proteins: victims, instigators, or innocent bystanders? The case of AIMP2 and alpha-synuclein. Neurobiol Dis 2021; 156:105417. [PMID: 34102275 DOI: 10.1016/j.nbd.2021.105417] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 01/21/2023] Open
Abstract
Lewy bodies (LBs), one of the neuropathological defining hallmarks of Parkinson's disease (PD), are composed of a complex mixture of alpha-synuclein (aSyn) filaments and hundreds of proteins, lipids, and membranous organelles. However, these proteins' role in aSyn aggregation and the biogenesis of LBs remains poorly understood. Previous studies have focused on investigating the role of these proteins as modifiers of aSyn aggregation, inclusion formation, and toxicity; very often, one protein at a time. In a recent study, Ham et al. suggest that one of these proteins, aminoacyl tRNA synthase complex-interacting multifunctional protein 2 (AIMP2), plays a primary role in the initiation of aSyn aggregation and is essential for aSyn inclusion formation and toxicity in cells and several models of synucleinopathies (Ham et al., 2020). Based on in vitro aggregation studies, they proposed a model in which AIMP2 self-associates to form amyloid-like aggregates that interact with monomeric aSyn and catalyze/seed the formation of aSyn fibrils and, eventually, LB-like inclusions. Herein, we present a critical analysis of their results and conclusions, review previous studies on AIMP2 aggregation, and reexamine the role of AIMP2 in regulating aSyn inclusion formation and clearance and aSyn-induced neurodegeneration in Parkinson's disease. We conclude by presenting lesson learned and recommendations on experimental factors and approaches that should be considered in future studies aimed at investigating the potential of targeting LBs-associated proteins, including AIMP2, for developing therapies to treat PD and other synucleinopathies.
Collapse
Affiliation(s)
- Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| | - Salvatore Novello
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, School of Life Sciences, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
41
|
Kahana M, Weizman A, Gabay M, Loboda Y, Segal-Gavish H, Gavish A, Barhum Y, Offen D, Finberg J, Allon N, Gavish M. Liposome-based targeting of dopamine to the brain: a novel approach for the treatment of Parkinson's disease. Mol Psychiatry 2021; 26:2626-2632. [PMID: 32372010 DOI: 10.1038/s41380-020-0742-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 03/08/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023]
Abstract
Delivery of drugs into the brain is poor due to the blood brain barrier (BBB). This study describes the development of a novel liposome-based brain-targeting drug delivery system. The liposomes incorporate a diacylglycerol moiety coupled through a linker to a peptide of 5 amino acids selected from amyloid precursor protein (APP), which is recognized by specific transporter(s)/receptor(s) in the BBB. This liposomal system enables the delivery of drugs across the BBB into the brain. The brain-directed liposomal system was used in a mouse model of Parkinson's disease (PD). Intra-peritoneal (IP) administration of liposomes loaded with dopamine (DA) demonstrated a good correlation between liposomal DA dose and the behavioral effects in hemiparkinsonian amphetamine-treated mice, with an optimal DA dose of 60 µg/kg. This is significantly lower dose than commonly used doses of the DA precursor levodopa (in the mg/kg range). IP injection of the APP-targeted liposomes loaded with a DA dose of 800 µg/kg, resulted in a significant increase in striatal DA within 5 min (6.9-fold, p < 0.05), in amphetamine-treated mice. The increase in striatal DA content persisted for at least 3 h after administration, which indicates a slow DA release from the delivery system. No elevation in DA content was detected in the heart or the liver. Similar increases in striatal DA were observed also in rats and mini-pigs. The liposomal delivery system enables penetration of compounds through the BBB and may be a candidate for the treatment of PD and other brain diseases.
Collapse
Affiliation(s)
- Meygal Kahana
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, 31096, Haifa, Israel
| | - Abraham Weizman
- Research Unit at Geha Mental Health Center and the Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, 4910002, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel-Aviv University, 6997801, Tel Aviv, Israel
| | - Martin Gabay
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, 31096, Haifa, Israel
| | - Yelena Loboda
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, 31096, Haifa, Israel
| | - Hadar Segal-Gavish
- Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.,Laboratory of Neuroscience, Felsenstein Medical Research Center, 4910002, Petah Tikva, Israel.,The Child Psychiatry Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, 5262000, Israel
| | - Avishai Gavish
- Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.,Goldschleger Eye Institute, Sheba Medical Center, Tel Hashomer, 5262000, Israel
| | - Yael Barhum
- Laboratory of Neuroscience, Felsenstein Medical Research Center, 4910002, Petah Tikva, Israel
| | - Dani Offen
- Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel-Aviv University, 6997801, Tel Aviv, Israel.,Laboratory of Neuroscience, Felsenstein Medical Research Center, 4910002, Petah Tikva, Israel
| | - John Finberg
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, 31096, Haifa, Israel
| | - Nahum Allon
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, 31096, Haifa, Israel
| | - Moshe Gavish
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, 31096, Haifa, Israel.
| |
Collapse
|
42
|
Masini D, Plewnia C, Bertho M, Scalbert N, Caggiano V, Fisone G. A Guide to the Generation of a 6-Hydroxydopamine Mouse Model of Parkinson's Disease for the Study of Non-Motor Symptoms. Biomedicines 2021; 9:biomedicines9060598. [PMID: 34070345 PMCID: PMC8227396 DOI: 10.3390/biomedicines9060598] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/12/2021] [Accepted: 05/19/2021] [Indexed: 11/16/2022] Open
Abstract
In Parkinson’s disease (PD), a large number of symptoms affecting the peripheral and central nervous system precede, develop in parallel to, the cardinal motor symptoms of the disease. The study of these conditions, which are often refractory to and may even be exacerbated by standard dopamine replacement therapies, relies on the availability of appropriate animal models. Previous work in rodents showed that injection of the neurotoxin 6-hydroxydopamine (6-OHDA) in discrete brain regions reproduces several non-motor comorbidities commonly associated with PD, including cognitive deficits, depression, anxiety, as well as disruption of olfactory discrimination and circadian rhythm. However, the use of 6-OHDA is frequently associated with significant post-surgical mortality. Here, we describe the generation of a mouse model of PD based on bilateral injection of 6-OHDA in the dorsal striatum. We show that the survival rates of males and females subjected to this lesion differ significantly, with a much higher mortality among males, and provide a protocol of enhanced pre- and post-operative care, which nearly eliminates animal loss. We also briefly discuss the utility of this model for the study of non-motor comorbidities of PD.
Collapse
Affiliation(s)
- Débora Masini
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden; (D.M.); (C.P.); (M.B.); (N.S.); (V.C.)
- Department of Neuroscience Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej, 3B, 2200 Copenhagen, Denmark
| | - Carina Plewnia
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden; (D.M.); (C.P.); (M.B.); (N.S.); (V.C.)
| | - Maëlle Bertho
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden; (D.M.); (C.P.); (M.B.); (N.S.); (V.C.)
- Department of Neuroscience Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej, 3B, 2200 Copenhagen, Denmark
| | - Nicolas Scalbert
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden; (D.M.); (C.P.); (M.B.); (N.S.); (V.C.)
| | - Vittorio Caggiano
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden; (D.M.); (C.P.); (M.B.); (N.S.); (V.C.)
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden; (D.M.); (C.P.); (M.B.); (N.S.); (V.C.)
- Correspondence:
| |
Collapse
|
43
|
Ferrari DP, Bortolanza M, Del Bel EA. Interferon-γ Involvement in the Neuroinflammation Associated with Parkinson's Disease and L-DOPA-Induced Dyskinesia. Neurotox Res 2021; 39:705-719. [PMID: 33687725 DOI: 10.1007/s12640-021-00345-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/05/2021] [Accepted: 02/23/2021] [Indexed: 02/08/2023]
Abstract
Interferon-γ (IFN-γ) is a proinflammatory cytokine that activates glial cells. IFN-γ is increased in the plasma and brain of Parkinson's disease patients, suggesting its potential role in the disease. We investigated whether the IFN-γ deficiency could interfere with nigrostriatal degeneration induced by the neurotoxin 6-hydroxydopamine, L-DOPA-induced dyskinesia, and the neuroinflammatory features as astrogliosis, microgliosis, and induced nitric oxide synthase (iNOS) immunoreactivity induced by L-DOPA treatment. Wild type (WT) and IFN-γ knockout (IFN-γ/KO) mice received unilateral striatal microinjections of 6-hydroxydopamine. Animals were sacrificed 1, 3, 7, and 21 days after lesions. Additional group of WT and IFN-γ/KO parkinsonian mice, after 3 weeks of neurotoxin injection, received L-DOPA (intraperitoneally, for 21 days) resulting in dyskinetic-like behavior. Tyrosine hydroxylase immunostaining indicated the starting of dopaminergic lesion since the first day past toxin administration, progressively increased until the third day when it stabilized. There was no difference in the lesion and L-DOPA-induced dyskinesia intensity between WT and IFN-γ/KO mice. Remarkably, IFN-γ/KO mice treated with L-DOPA presented in the lesioned striatum an increase of iNOS and glial fibrilary acid protein (GFAP) density, compared with the WT group. Morphological analysis revealed the rise of astrocytes and microglia reactivity in IFN-γ/KO mice exibiting dyskinesia. In conclusion, IFN-γ/KO mice presented an intensification of the inflammatory reaction accompanying L-DOPA treatment and suggest that iNOS and GFAP increase, and the activation of astrocytes and microglia induced afterward L-DOPA treatment was IFN-γ independent events. Intriguingly, IFN-γ absence did not affect the degeneration of dopaminergic neurons or LID development.
Collapse
Affiliation(s)
- D P Ferrari
- Department of Neuroscience, School of Medicine of Ribeirão Preto, University of São Paulo, SP, 14040-900, Brazil.,Department of Morphology, Physiology and Basic Pathology, School of Dentistry of Ribeirão Preto, University of São Paulo, SP, 14040-904, Brazil
| | - M Bortolanza
- Department of Morphology, Physiology and Basic Pathology, School of Dentistry of Ribeirão Preto, University of São Paulo, SP, 14040-904, Brazil
| | - E A Del Bel
- Department of Neuroscience, School of Medicine of Ribeirão Preto, University of São Paulo, SP, 14040-900, Brazil. .,Department of Morphology, Physiology and Basic Pathology, School of Dentistry of Ribeirão Preto, University of São Paulo, SP, 14040-904, Brazil.
| |
Collapse
|
44
|
Wu Y, Jiao Z, Wan Z, Qu S. Role of autophagy and oxidative stress to astrocytes in fenpropathrin-induced Parkinson-like damage. Neurochem Int 2021; 145:105000. [PMID: 33617931 DOI: 10.1016/j.neuint.2021.105000] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 01/31/2021] [Accepted: 02/15/2021] [Indexed: 11/17/2022]
Abstract
Fenpropathrin is an insecticide that is widely used in agriculture. It remains unknown whether fenpropathrin exposure increases the risk of Parkinson's disease. We found that fenpropathrin increased oxidative stress both in vitro and in vivo. Additionally, fenpropathrin increased production of ROS, NOS2, and HO-1, and decreased SOD and GSH in astrocytes. We further found that fenpropathrin-mediated oxidative stress might inhibit autophagic flow, including decreased expression of LC3A/B and enhanced expression of SQSTM1 via down-regulation of CDK-5, an upstream marker of autophagy. In mice, autophagy was slightly different from that found in astrocytes, as reflected in the increased expressions of LC3A/B and SQSTM1. Our findings elucidate the toxicological phenomena and pathogenic mechanisms of fenpropathrin and may provide guidance for improved pesticide control and environmental protection.
Collapse
Affiliation(s)
- Yixuan Wu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China; Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, Guangdong, 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong, 510515, China; School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhigang Jiao
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China; Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, Guangdong, 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong, 510515, China; School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhiting Wan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China; Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, Guangdong, 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Shaogang Qu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China; Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, Guangdong, 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
45
|
Dos Santos Pereira M, Abreu GHD, Rocca J, Hamadat S, Raisman-Vozari R, Michel PP, Del Bel E. Contributive Role of TNF-α to L-DOPA-Induced Dyskinesia in a Unilateral 6-OHDA Lesion Model of Parkinson's Disease. Front Pharmacol 2021; 11:617085. [PMID: 33510643 PMCID: PMC7836015 DOI: 10.3389/fphar.2020.617085] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/27/2020] [Indexed: 12/19/2022] Open
Abstract
Our present objective was to better characterize the mechanisms that regulate striatal neuroinflammation in mice developing L-DOPA-induced dyskinesia (LID). For that, we used 6-hydroxydopamine (6-OHDA)-lesioned mice rendered dyskinetic by repeated intraperitoneal injections of 3,4-dihydroxyphenyl-L-alanine (L-DOPA) and quantified ensuing neuroinflammatory changes in the dopamine-denervated dorsal striatum. LID development was associated with a prominent astrocytic response, and a more moderate microglial cell reaction restricted to this striatal area. The glial response was associated with elevations in two pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α) and interleukin-1β. Treatment with the phytocannabinoid cannabidiol and the transient receptor potential vanilloid-1 (TRPV-1) channel antagonist capsazepine diminished LID intensity and decreased TNF-α levels without impacting other inflammation markers. To possibly reproduce the neuroinflammatory component of LID, we exposed astrocyte and microglial cells in culture to candidate molecules that might operate as inflammatory cues during LID development, i.e., L-DOPA, dopamine, or glutamate. Neither L-DOPA nor dopamine produced an inflammatory response in glial cell cultures. However, glutamate enhanced TNF-α secretion and GFAP expression in astrocyte cultures and promoted Iba-1 expression in microglial cultures. Of interest, the antidyskinetic treatment with cannabidiol + capsazepine reduced TNF-α release in glutamate-activated astrocytes. TNF-α, on its own, promoted the synaptic release of glutamate in cortical neuronal cultures, whereas cannabidiol + capsazepine prevented this effect. Therefore, we may assume that the release of TNF-α by glutamate-activated astrocytes may contribute to LID by exacerbating corticostriatal glutamatergic inputs excitability and maintaining astrocytes in an activated state through a self-reinforcing mechanism.
Collapse
Affiliation(s)
- Maurício Dos Santos Pereira
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil.,Department of Physiology, FMRP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil.,USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Brazil.,Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France
| | - Gabriel Henrique Dias Abreu
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil.,USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Brazil
| | - Jeremy Rocca
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France
| | - Sabah Hamadat
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France
| | - Rita Raisman-Vozari
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France
| | - Patrick Pierre Michel
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne Université UM75, Paris, France
| | - Elaine Del Bel
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil.,Department of Physiology, FMRP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil.,USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Brazil
| |
Collapse
|
46
|
Carstens M, Haq I, Martinez-Cerrato J, Dos-Anjos S, Bertram K, Correa D. Sustained clinical improvement of Parkinson's disease in two patients with facially-transplanted adipose-derived stromal vascular fraction cells. J Clin Neurosci 2020; 81:47-51. [PMID: 33222965 DOI: 10.1016/j.jocn.2020.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/06/2020] [Indexed: 12/20/2022]
Abstract
Cell-based therapy has been studied as an alternative for Parkinson's Disease (PD), with different routes of administration. The superficial fascia and facial muscles possess a rich blood supply, while venous and lymphatic access via the orbit and the cribriform plate provide a route to cerebral circulation. We here document positive clinical effects in two patients with PD treated with autologous adipose-derived stromal vascular fraction (SVF) cell preparation, implanted into the face and nasal cavity. Two patients with PD were transplanted with 60 million total nucleated cells in processed SVF into the facial muscles and nose. Serial evaluations were carried out up to 5 years (patient 1) and 1 year (patient 2), using the PDQ-39, the UPDRS, and serial videos. Video scoring was reviewed in a blinded fashion. Both patients reported qualitative improvement in motor and nonmotor symptoms following injection. Quantitatively, PDQ-39 scores decreased in all categories for both. On-medication UPDRS motor scores decreased in both (20 to 4 in patient 1, 18 to 3 in patient 2) despite taking the same or less medication (LEDD 350 to 350 in patient 1, LEDD 1175 to 400 in pt2). Both subjects had off-medication UPDRS scores similar to their pretreatment on-medication scores (20 to 14 in patient 1, 18 to 23 in patient 2). These preliminary findings describe local facial and nasal injections of SVF preparation followed by prolonged clinical benefit in two patients. Despite an unknown mechanism of action, this potential therapy warrants careful verification and investigation.
Collapse
Affiliation(s)
- Michael Carstens
- Wake Forest University Institute of Regenerative Medicine, Winston-Salem, NC, USA; Department of Plastic Surgery, Hospital Escuela Oscar Danilo Rosales Argüello, Leon, Nicaragua.
| | - Ihtsham Haq
- Department of Neurology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | | | - Ken Bertram
- Wake Forest University Institute of Regenerative Medicine, Winston-Salem, NC, USA
| | - Diego Correa
- Department of Orthopaedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, Miami, FL, USA; Diabetes Research Institute & Cell Transplant Center, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
47
|
Kadnikov IA, Verbovaya ER, Voronkov DN, Voronin MV, Seredenin SB. Deferred Administration of Afobazole Induces Sigma1R-Dependent Restoration of Striatal Dopamine Content in a Mouse Model of Parkinson's Disease. Int J Mol Sci 2020; 21:E7620. [PMID: 33076300 PMCID: PMC7593947 DOI: 10.3390/ijms21207620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/28/2020] [Accepted: 10/13/2020] [Indexed: 12/22/2022] Open
Abstract
Previously, we demonstrated that the immediate administration of multitarget anxiolytic afobazole slows down the progression of neuronal damage in a 6-hydroxidodamine (6-OHDA) model of Parkinson's disease due to the activation of chaperone Sigma1R. The aim of the present study is to evaluate the therapeutic potential of deferred afobazole administration in this model. Male ICR mice received a unilateral 6-OHDA lesion of the striatum. Fourteen days after the surgery, mice were treated with afobazole, selective Sigma1R agonist PRE-084, selective Sigma1R antagonist BD-1047, and a combination of BD-1047 with afobazole or PRE-084 for another 14 days. The deferred administration of afobazole restored the intrastriatal dopamine content in the 6-OHDA-lesioned striatum and facilitated motor behavior in rotarod tests. The action of afobazole accorded with the effect of Sigma1R selective agonist PRE-084 and was blocked by Sigma1R selective antagonist BD-1047. The present study illustrates the Sigma1R-dependent effects of afobazole in a 6-OHDA model of Parkinson's disease and reveals the therapeutic potential of Sigma1R agonists in treatment of the condition.
Collapse
Affiliation(s)
- Ilya A. Kadnikov
- Department of Pharmacogenetics, FSBI “Zakusov Institute of Pharmacology”, Baltiyskaya street 8, 125315 Moscow, Russia; (E.R.V.); (M.V.V.)
| | - Ekaterina R. Verbovaya
- Department of Pharmacogenetics, FSBI “Zakusov Institute of Pharmacology”, Baltiyskaya street 8, 125315 Moscow, Russia; (E.R.V.); (M.V.V.)
| | - Dmitry N. Voronkov
- Laboratory of neuromorphology, Research Center of Neurology, Volokolamskoe Highway 80, 125367 Moscow, Russia;
| | - Mikhail V. Voronin
- Department of Pharmacogenetics, FSBI “Zakusov Institute of Pharmacology”, Baltiyskaya street 8, 125315 Moscow, Russia; (E.R.V.); (M.V.V.)
| | - Sergei B. Seredenin
- Department of Pharmacogenetics, FSBI “Zakusov Institute of Pharmacology”, Baltiyskaya street 8, 125315 Moscow, Russia; (E.R.V.); (M.V.V.)
| |
Collapse
|
48
|
Li W, Goshima Y, Ohshima T. Loss of Collapsin Response Mediator Protein 4 Attenuates 6-Hydroxydopamine-Induced Impairments in a Mouse Model of Parkinson's Disease. Neurochem Res 2020; 45:2286-2301. [PMID: 32648145 DOI: 10.1007/s11064-020-03086-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 06/11/2020] [Accepted: 07/04/2020] [Indexed: 12/01/2022]
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative disorder characterized by impaired motor symptoms induced by the degeneration of dopaminergic neurons of the substantia nigra pars compacta (SNc). Many factors are speculated to operate in the mechanism of PD, including oxidative stress, mitochondrial dysfunction, abnormal protein handling, and PD induced apoptosis. Besides, researchers have recently shown that inflammatory secretions may engage neighboring cells such as astrocytes, which then induce autocrine and paracrine responses that amplify the inflammation, leading to neurodegeneration. In the present study, we analyzed the neuroprotective and anti-inflammatory effects of collapsin response mediator protein 4 (CRMP4) deletion in 6-hydroxydopamine (6-OHDA)-injected male mice, as well as its effects on motor impairments. Our findings indicated that the deletion of CRMP4 could maintain the TH-positive fibers in the striatum and the TH-positive cells in SNc, attenuate the inflammatory responses, and improve motor coordination and rotational behavior. Furthermore, based on our findings at the early time points, we hypothesized that primary differences between the Crmp4+/+ and Crmp4-/- mice may occur in microglia instead of neurons. Although further work should be carried out to clarify the specific role of CRMP4 in the pathogenesis of PD, our findings suggest that it could be a possible target for the treatment of PD.
Collapse
Affiliation(s)
- Wenting Li
- Department of Life Science and Medical Bio-Science, Waseda University, 2-2 Wakamatsu-cho Shinjukuku, Tokyo, 162-8480, Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Toshio Ohshima
- Department of Life Science and Medical Bio-Science, Waseda University, 2-2 Wakamatsu-cho Shinjukuku, Tokyo, 162-8480, Japan.
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan.
| |
Collapse
|
49
|
Campos ACP, Kikuchi DS, Paschoa AFN, Kuroki MA, Fonoff ET, Hamani C, Pagano RL, Hernandes MS. Unraveling the Role of Astrocytes in Subthalamic Nucleus Deep Brain Stimulation in a Parkinson's Disease Rat Model. Cell Mol Neurobiol 2020; 40:939-954. [PMID: 31939008 PMCID: PMC7295825 DOI: 10.1007/s10571-019-00784-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/31/2019] [Indexed: 12/13/2022]
Abstract
Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is an effective therapeutic strategy for motor symptoms of Parkinson's disease (PD) when L-DOPA therapy induces disabling side effects. Classical inflammatory activation of glial cells is well established in PD, contributing to the progressive neurodegenerative state; however, the role of DBS in regulating the inflammatory response remains largely unknown. To understand the involvement of astrocytes in the mechanisms of action of DBS, we evaluated the effect of STN-DBS in regulating motor symptoms, astrocyte reactivity, and cytokine expression in a 6-OHDA-induced PD rat model. To mimic in vivo DBS, we investigate the effect of high-frequency stimulation (HFS) in cultured astrocytes regulating cytokine induction and NF-κB activation. We found that STN-DBS improved motor impairment, induced astrocytic hyperplasia, and reversed increased IFN-γ and IL-10 levels in the globus pallidus (GP) of lesioned rats. Moreover, HFS activated astrocytes and prevented TNF-α-induced increase of monocyte chemoattractant protein-1 (MCP-1) and NF-κB activation in vitro. Our results indicate that DBS/HFS may act as a regulator of the inflammatory response in PD states, attenuating classical activation of astrocytes and cytokine induction, potentially through its ability to regulate NF-κB activation. These findings may help us understand the role of astrocyte signaling in HFS, highlighting its possible relationship with the effectiveness of DBS in neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | - Mayra Akemi Kuroki
- Division of Neuroscience, Hospital Sírio-Libanês, São Paulo, SP, 01308-060, Brazil
| | - Erich Talamoni Fonoff
- Division of Neurosurgery, Department of Neurology, University of São Paulo Medical School, São Paulo, 01246-903, Brazil
| | - Clement Hamani
- Sunnybrook Health Research Institute, Harquail Centre for Neuromodulation, Toronto, ON, M4N 3M5, Canada
| | - Rosana Lima Pagano
- Division of Neuroscience, Hospital Sírio-Libanês, São Paulo, SP, 01308-060, Brazil.
| | | |
Collapse
|
50
|
Tomov N. Glial cells in intracerebral transplantation for Parkinson's disease. Neural Regen Res 2020; 15:1173-1178. [PMID: 31960796 PMCID: PMC7047789 DOI: 10.4103/1673-5374.270296] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/02/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022] Open
Abstract
In the last few decades, intracerebral transplantation has grown from a dubious neuroscientific topic to a plausible modality for treatment of neurological disorders. The possibility for cell replacement opens a new field of perspectives in the therapy of neurodegenerative disorders, ischemia, and neurotrauma, with the most lessons learned from intracerebral transplantation in Parkinson's disease. Multiple animal studies and a few small-scale clinical trials have proven the concept of intracerebral grafting, but still have to provide a uniform and highly efficient approach to the procedure, suitable for clinical application. The success of intracerebral transplantation is highly dependent on the integration of the grafted cells with the host brain. In this process, glial cells are clearly more than passive bystanders. They provide transplanted cells with mechanical support, trophics, mediate synapse formation, and participate in graft vascularization. At the same time, glial cells mediate scarring, graft rejection, and neuroinflammation, which can be detrimental. We can use this information to try to understand the mechanisms behind the glial reaction to intracerebral transplantation. Recognizing and utilizing glial reactivity can move translational research forward and provide an insight not only to post-transplantation events but also to mechanisms of neuronal death and degeneration. Knowledge about glial reactivity to transplanted cells could also be a key for optimization of transplantation protocols, which ultimately should contribute to greater patient benefit.
Collapse
Affiliation(s)
- Nikola Tomov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland
| |
Collapse
|