1
|
Shih JY, Hsu YHH. Peptide Activator Stabilizes DJ-1 Structure and Enhances Its Activity. Int J Mol Sci 2024; 25:11075. [PMID: 39456860 PMCID: PMC11508141 DOI: 10.3390/ijms252011075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
DJ-1 is a vital enzyme involved in the maintenance of mitochondrial health, and its mutation has been associated with an increased risk of Parkinson's disease (PD). Effective regulation of DJ-1 activity is essential for the well-being of mitochondria, and DJ-1 is thus a potential target for PD drug development. In this study, two peptides (15EEMETIIPVDVMRRA29 and 47SRDVVICPDA56) were utilized with the aim of enhancing the activity of DJ-1. The mechanisms underlying the activity enhancement by these two peptides were investigated using hydrogen/deuterium exchange mass spectrometry (HDXMS). The HDXMS results revealed distinct mechanisms. Peptide 1 obstructs the access of solvent to the dimer interface and stabilizes the α/β hydrolase structure, facilitating substrate binding to a stabilized active site. Conversely, peptide 2 induces a destabilization of the α/β hydrolase core, enhancing substrate accessibility and subsequently increasing DJ-1 activity. The binding of these two peptides optimizes the activity site within the dimeric structure. These findings offer valuable insights into the mechanisms underlying the activity enhancement of DJ-1 by the two peptides, potentially aiding the development of new drugs that can enhance the activity of DJ-1 and, consequently, advance PD treatment.
Collapse
Affiliation(s)
| | - Yuan-Hao Howard Hsu
- Department of Chemistry, Tunghai University, No. 1727, Sec. 4, Taiwan Boulevard, Xitun District, Taichung 40704, Taiwan;
| |
Collapse
|
2
|
Roodveldt C, Bernardino L, Oztop-Cakmak O, Dragic M, Fladmark KE, Ertan S, Aktas B, Pita C, Ciglar L, Garraux G, Williams-Gray C, Pacheco R, Romero-Ramos M. The immune system in Parkinson's disease: what we know so far. Brain 2024; 147:3306-3324. [PMID: 38833182 PMCID: PMC11449148 DOI: 10.1093/brain/awae177] [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: 02/20/2024] [Revised: 05/02/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
Parkinson's disease is characterized neuropathologically by the degeneration of dopaminergic neurons in the ventral midbrain, the accumulation of α-synuclein (α-syn) aggregates in neurons and chronic neuroinflammation. In the past two decades, in vitro, ex vivo and in vivo studies have consistently shown the involvement of inflammatory responses mediated by microglia and astrocytes, which may be elicited by pathological α-syn or signals from affected neurons and other cell types, and are directly linked to neurodegeneration and disease development. Apart from the prominent immune alterations seen in the CNS, including the infiltration of T cells into the brain, more recent studies have demonstrated important changes in the peripheral immune profile within both the innate and adaptive compartments, particularly involving monocytes, CD4+ and CD8+ T cells. This review aims to integrate the consolidated understanding of immune-related processes underlying the pathogenesis of Parkinson's disease, focusing on both central and peripheral immune cells, neuron-glia crosstalk as well as the central-peripheral immune interaction during the development of Parkinson's disease. Our analysis seeks to provide a comprehensive view of the emerging knowledge of the mechanisms of immunity in Parkinson's disease and the implications of this for better understanding the overall pathogenesis of this disease.
Collapse
Affiliation(s)
- Cintia Roodveldt
- Centre for Molecular Biology and Regenerative Medicine-CABIMER, University of Seville-CSIC, Seville 41092, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville 41009, Spain
| | - Liliana Bernardino
- Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, 6200-506, Covilhã, Portugal
| | - Ozgur Oztop-Cakmak
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul 34010, Turkey
| | - Milorad Dragic
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
- Department of Molecular Biology and Endocrinology, ‘VINČA’ Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Kari E Fladmark
- Department of Biological Science, University of Bergen, 5006 Bergen, Norway
| | - Sibel Ertan
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul 34010, Turkey
| | - Busra Aktas
- Department of Molecular Biology and Genetics, Burdur Mehmet Akif Ersoy University, Burdur 15200, Turkey
| | - Carlos Pita
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Lucia Ciglar
- Center Health & Bioresources, Competence Unit Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, 1210 Vienna, Austria
| | - Gaetan Garraux
- Movere Group, Faculty of Medicine, GIGA Institute, University of Liège, Liège 4000, Belgium
| | | | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580702, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia 7510156, Santiago, Chile
| | - Marina Romero-Ramos
- Department of Biomedicine & The Danish Research Institute of Translational Neuroscience—DANDRITE, Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|
3
|
Alcalá-Zúniga D, Espinoza-Torres E, Das RK, Vargas M, Maldonado O, Benavides O, Manojkumar A, de la Garza R, Davila N, Perez I, Martinez AH, Roy D, López-Juárez A, Zarei MM, Baker KA, Gil M, Rodrigo H, de Erausquin GA, Roy U. Enriched Environment Contributes to the Recovery from Neurotoxin-Induced Parkinson's Disease Pathology. Mol Neurobiol 2024; 61:6734-6753. [PMID: 38349515 PMCID: PMC11339186 DOI: 10.1007/s12035-024-03951-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 01/02/2024] [Indexed: 08/22/2024]
Abstract
Parkinson's disease (PD) is a neurological disorder that affects dopaminergic neurons. The lack of understanding of the underlying molecular mechanisms of PD pathology makes treating it a challenge. Several pieces of evidence support the protective role of enriched environment (EE) and exercise on dopaminergic neurons. The specific aspect(s) of neuroprotection after exposure to EE have not been identified. Therefore, we have investigated the protective role of EE on dopamine dysregulation and subsequent downregulation of DJ1 protein using in vitro and in vivo models of PD. Our study for the first time demonstrated that DJ1 expression has a direct correlation with dopamine downregulation in PD models and exposure to EE has a significant impact on improving the behavioral changes in PD mice. This research provides evidence that exercise in EE has a positive effect on PD without interfering with the current line of therapy.
Collapse
Affiliation(s)
- Daphne Alcalá-Zúniga
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA
| | - Erika Espinoza-Torres
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA
| | - Ranjit Kumar Das
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA
| | - Magaly Vargas
- Department of Psychological Science, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Oscar Maldonado
- Institute of Neuroscience, University of Texas Rio Grande Valley School of Medicine, Harlingen, TX, USA
| | - Omar Benavides
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA
- Institute of Neuroscience, University of Texas Rio Grande Valley School of Medicine, Harlingen, TX, USA
| | - Arvind Manojkumar
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA
| | - Roberto de la Garza
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA
| | - Natalia Davila
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA
| | - Isaac Perez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA
| | - Alejandro Hernandez Martinez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA
| | - Deepa Roy
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA
| | - Alejandro López-Juárez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA
| | - Masoud M Zarei
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA
| | - Kelsey A Baker
- Institute of Neuroscience, University of Texas Rio Grande Valley School of Medicine, Harlingen, TX, USA
| | - Mario Gil
- Department of Psychological Science, University of Texas Rio Grande Valley, Brownsville, TX, USA
- Institute of Neuroscience, University of Texas Rio Grande Valley School of Medicine, Harlingen, TX, USA
| | - Hansapani Rodrigo
- School of Mathematical and Statistical Sciences, University of Rio Grande Valley, Edinburg, TX, USA
| | - Gabriel A de Erausquin
- The Glenn Biggs Institute of Alzheimer's and Neurodegenerative Disorders, Joe and Teresa Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Upal Roy
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA.
| |
Collapse
|
4
|
Wu Q, Zhu J, Zhang X, Xu X, Luo D, Lin Y, Yan M, Song Y. The antioxidant effect of tetrahedral framework nucleic acid-based delivery of small activating RNA targeting DJ-1 on retinal oxidative stress injury. Cell Prolif 2024; 57:e13635. [PMID: 38594962 PMCID: PMC11294416 DOI: 10.1111/cpr.13635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 04/11/2024] Open
Abstract
Age-related macular degeneration (AMD) and diabetic retinopathy (DR) are the world's leading causes of blindness. The retinal pigment epithelium (RPE) and vascular endothelial cell exposed to oxidative stress is the major cause of AMD and DR. DJ-1, an important endogenous antioxidant, its overexpression is considered as a promising antioxidant treatment for AMD and DR. Here, we modified the tetrahedral frame nucleic acids (tFNAs) with DJ-1 saRNAs as a delivery system, and synthesized a novel nanocomplex (tFNAs-DJ-1 saRNAs). In vitro studies show that tFNAs-DJ-1 saRNAs can efficiently transfer DJ-1 saRNAs to human umbilical vein endothelial cells (HUVECs) and ARPE-19s, and significantly increased their cellular DJ-1 level. Reactive oxygen species expression in H2O2-treated HUVECs and ARPE-19s were decreased, cell viability was enhanced and cell apoptosis were inhibited when tFNAs-DJ-1 saRNAs were delivered. Moreover, tFNAs-DJ-1 saRNAs preserved mitochondrial structure and function under oxidative stress conditions. In the aspect of molecular mechanism, tFNAs-DJ-1 saRNAs activated Erk and Nrf2 pathway, which might contribute to its protective effects against oxidative stress damage. To conclude, this study shows the successfully establishment of a simple but effective delivery system of DJ-1 saRNAs associated with antioxidant effects in AMD and DR, which may be a promising agent for future treatment in oxidative stress-related retinal disorders.
Collapse
Affiliation(s)
- Qiaowei Wu
- Department of OphthalmologyGeneral Hospital of Central Theater CommandWuhanChina
| | - Jingyi Zhu
- Department of OphthalmologyGeneral Hospital of Central Theater CommandWuhanChina
| | - Xianggui Zhang
- Department of OphthalmologyGeneral Hospital of Central Theater CommandWuhanChina
| | - Xiaoxiao Xu
- Innovative Institute of Chinese Medicine and PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Delun Luo
- Innovative Institute of Chinese Medicine and PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Yunfeng Lin
- Department of Maxillofacial Surgery, State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyChengduChina
| | - Ming Yan
- Department of OphthalmologyGeneral Hospital of Central Theater CommandWuhanChina
| | - Yanping Song
- Department of OphthalmologyGeneral Hospital of Central Theater CommandWuhanChina
| |
Collapse
|
5
|
Zohar K, Linial M. Knockdown of DJ-1 Resulted in a Coordinated Activation of the Innate Immune Antiviral Response in HEK293 Cell Line. Int J Mol Sci 2024; 25:7550. [PMID: 39062793 PMCID: PMC11277157 DOI: 10.3390/ijms25147550] [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: 06/19/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
PARK7, also known as DJ-1, plays a critical role in protecting cells by functioning as a sensitive oxidation sensor and modulator of antioxidants. DJ-1 acts to maintain mitochondrial function and regulate transcription in response to different stressors. In this study, we showed that cell lines vary based on their antioxidation potential under basal conditions. The transcriptome of HEK293 cells was tested following knockdown (KD) of DJ-1 using siRNAs, which reduced the DJ-1 transcripts to only 12% of the original level. We compared the expression levels of 14k protein-coding transcripts and 4.2k non-coding RNAs relative to cells treated with non-specific siRNAs. Among the coding genes, approximately 200 upregulated differentially expressed genes (DEGs) signified a coordinated antiviral innate immune response. Most genes were associated with the regulation of type 1 interferons (IFN) and the induction of inflammatory cytokines. About a quarter of these genes were also induced in cells treated with non-specific siRNAs that were used as a negative control. Beyond the antiviral-like response, 114 genes were specific to the KD of DJ-1 with enrichment in RNA metabolism and mitochondrial functions. A smaller set of downregulated genes (58 genes) was associated with dysregulation in membrane structure, cell viability, and mitophagy. We propose that the KD DJ-1 perturbation diminishes the protective potency against oxidative stress. Thus, it renders the cells labile and responsive to the dsRNA signal by activating a large number of genes, many of which drive apoptosis, cell death, and inflammatory signatures. The KD of DJ-1 highlights its potency in regulating genes associated with antiviral responses, RNA metabolism, and mitochondrial functions, apparently through alteration in STAT activity and downstream signaling. Given that DJ-1 also acts as an oncogene in metastatic cancers, targeting DJ-1 could be a promising therapeutic strategy where manipulation of the DJ-1 level may reduce cancer cell viability and enhance the efficacy of cancer treatments.
Collapse
Affiliation(s)
| | - Michal Linial
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel;
| |
Collapse
|
6
|
Ilieva NM, Hoffman EK, Ghalib MA, Greenamyre JT, De Miranda BR. LRRK2 kinase inhibition protects against Parkinson's disease-associated environmental toxicants. Neurobiol Dis 2024; 196:106522. [PMID: 38705492 PMCID: PMC11332574 DOI: 10.1016/j.nbd.2024.106522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024] Open
Abstract
Idiopathic Parkinson's disease (PD) is epidemiologically linked with exposure to toxicants such as pesticides and solvents, which comprise a wide array of chemicals that pollute our environment. While most are structurally distinct, a common cellular target for their toxicity is mitochondrial dysfunction, a key pathological trigger involved in the selective vulnerability of dopaminergic neurons. We and others have shown that environmental mitochondrial toxicants such as the pesticides rotenone and paraquat, and the organic solvent trichloroethylene (TCE) appear to be influenced by the protein LRRK2, a genetic risk factor for PD. As LRRK2 mediates vesicular trafficking and influences endolysosomal function, we postulated that LRRK2 kinase activity may inhibit the autophagic removal of toxicant damaged mitochondria, resulting in elevated oxidative stress. Conversely, we suspected that inhibition of LRRK2, which has been shown to be protective against dopaminergic neurodegeneration caused by mitochondrial toxicants, would reduce the intracellular production of reactive oxygen species (ROS) and prevent mitochondrial toxicity from inducing cell death. To do this, we tested in vitro if genetic or pharmacologic inhibition of LRRK2 (MLi2) protected against ROS caused by four toxicants associated with PD risk - rotenone, paraquat, TCE, and tetrachloroethylene (PERC). In parallel, we assessed if LRRK2 inhibition with MLi2 could protect against TCE-induced toxicity in vivo, in a follow up study from our observation that TCE elevated LRRK2 kinase activity in the nigrostriatal tract of rats prior to dopaminergic neurodegeneration. We found that LRRK2 inhibition blocked toxicant-induced ROS and promoted mitophagy in vitro, and protected against dopaminergic neurodegeneration, neuroinflammation, and mitochondrial damage caused by TCE in vivo. We also found that cells with the LRRK2 G2019S mutation displayed exacerbated levels of toxicant induced ROS, but this was ameliorated by LRRK2 inhibition with MLi2. Collectively, these data support a role for LRRK2 in toxicant-induced mitochondrial dysfunction linked to PD risk through oxidative stress and the autophagic removal of damaged mitochondria.
Collapse
Affiliation(s)
- Neda M Ilieva
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eric K Hoffman
- Pittsburgh Institute for Neurodegenerative Diseases, Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mohammed A Ghalib
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Briana R De Miranda
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
7
|
Naoi M, Maruyama W, Shamoto-Nagai M, Riederer P. Toxic interactions between dopamine, α-synuclein, monoamine oxidase, and genes in mitochondria of Parkinson's disease. J Neural Transm (Vienna) 2024; 131:639-661. [PMID: 38196001 DOI: 10.1007/s00702-023-02730-6] [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: 10/15/2023] [Accepted: 12/15/2023] [Indexed: 01/11/2024]
Abstract
Parkinson's disease is characterized by its distinct pathological features; loss of dopamine neurons in the substantia nigra pars compacta and accumulation of Lewy bodies and Lewy neurites containing modified α-synuclein. Beneficial effects of L-DOPA and dopamine replacement therapy indicate dopamine deficit as one of the main pathogenic factors. Dopamine and its oxidation products are proposed to induce selective vulnerability in dopamine neurons. However, Parkinson's disease is now considered as a generalized disease with dysfunction of several neurotransmitter systems caused by multiple genetic and environmental factors. The pathogenic factors include oxidative stress, mitochondrial dysfunction, α-synuclein accumulation, programmed cell death, impaired proteolytic systems, neuroinflammation, and decline of neurotrophic factors. This paper presents interactions among dopamine, α-synuclein, monoamine oxidase, its inhibitors, and related genes in mitochondria. α-Synuclein inhibits dopamine synthesis and function. Vice versa, dopamine oxidation by monoamine oxidase produces toxic aldehydes, reactive oxygen species, and quinones, which modify α-synuclein, and promote its fibril production and accumulation in mitochondria. Excessive dopamine in experimental models modifies proteins in the mitochondrial electron transport chain and inhibits the function. α-Synuclein and familiar Parkinson's disease-related gene products modify the expression and activity of monoamine oxidase. Type A monoamine oxidase is associated with neuroprotection by an unspecific dose of inhibitors of type B monoamine oxidase, rasagiline and selegiline. Rasagiline and selegiline prevent α-synuclein fibrillization, modulate this toxic collaboration, and exert neuroprotection in experimental studies. Complex interactions between these pathogenic factors play a decisive role in neurodegeneration in PD and should be further defined to develop new therapies for Parkinson's disease.
Collapse
Affiliation(s)
- Makoto Naoi
- Department of Health and Nutritional Sciences, Faculty of Health Sciences, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 320-0195, Japan.
| | - Wakako Maruyama
- Department of Health and Nutritional Sciences, Faculty of Health Sciences, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 320-0195, Japan
| | - Masayo Shamoto-Nagai
- Department of Health and Nutritional Sciences, Faculty of Health Sciences, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 320-0195, Japan
| | - Peter Riederer
- Clinical Neurochemistry, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
- Department of Psychiatry, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
8
|
Jia Q, Li J, Guo X, Li Y, Wu Y, Peng Y, Fang Z, Zhang X. Neuroprotective effects of chaperone-mediated autophagy in neurodegenerative diseases. Neural Regen Res 2024; 19:1291-1298. [PMID: 37905878 PMCID: PMC11467915 DOI: 10.4103/1673-5374.385848] [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: 03/20/2023] [Revised: 05/30/2023] [Accepted: 07/17/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Chaperone-mediated autophagy is one of three types of autophagy and is characterized by the selective degradation of proteins. Chaperone-mediated autophagy contributes to energy balance and helps maintain cellular homeostasis, while providing nutrients and support for cell survival. Chaperone-mediated autophagy activity can be detected in almost all cells, including neurons. Owing to the extreme sensitivity of neurons to their environmental changes, maintaining neuronal homeostasis is critical for neuronal growth and survival. Chaperone-mediated autophagy dysfunction is closely related to central nervous system diseases. It has been shown that neuronal damage and cell death are accompanied by chaperone-mediated autophagy dysfunction. Under certain conditions, regulation of chaperone-mediated autophagy activity attenuates neurotoxicity. In this paper, we review the changes in chaperone-mediated autophagy in neurodegenerative diseases, brain injury, glioma, and autoimmune diseases. We also summarize the most recent research progress on chaperone-mediated autophagy regulation and discuss the potential of chaperone-mediated autophagy as a therapeutic target for central nervous system diseases.
Collapse
Affiliation(s)
- Qi Jia
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi Province, China
| | - Jin Li
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi Province, China
- Department of Critical Care Medicine, Air Force Medical Center, Beijing, China
| | - Xiaofeng Guo
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi Province, China
| | - Yi Li
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi Province, China
| | - You Wu
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi Province, China
| | - Yuliang Peng
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi Province, China
| | - Zongping Fang
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi Province, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xijing Zhang
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi Province, China
| |
Collapse
|
9
|
Sarkar A, Singh MP. A Complex Interplay of DJ-1, LRRK2, and Nrf2 in the Regulation of Mitochondrial Function in Cypermethrin-Induced Parkinsonism. Mol Neurobiol 2024; 61:953-970. [PMID: 37674036 DOI: 10.1007/s12035-023-03591-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/16/2023] [Indexed: 09/08/2023]
Abstract
Cypermethrin impairs mitochondrial function, induces redox imbalance, and leads to Parkinsonism in experimental animals. Knockdown of deglycase-1 (DJ-1) gene, which encodes a redox-sensitive antioxidant protein, aggravates cypermethrin-mediated α-synuclein overexpression and oxidative alteration of proteins. DJ-1 is also reported to be essential for maintaining stability of nuclear factor erythroid 2-related factor 2 (Nrf2), shielding cells against oxidative insult. Leucine-rich repeat kinase 2 (LRRK2), another protein associated with Parkinson's disease, is also involved in regulating mitochondrial function. However, underlying molecular mechanisms remain elusive. The study intended to explore an interaction of DJ-1, LRRK2, and Nrf2 in the regulation of mitochondrial function in cypermethrin-induced Parkinsonism. Small interfering RNA-mediated knockdown of DJ-1 and LRRK2 gene and pharmacological activation of Nrf2 were performed in rats and/or human neuroblastoma cells with or without cypermethrin. Indexes of oxidative stress, mitochondrial impairment, and Parkinsonism along with α-synuclein expression, post-translational modification, and aggregation were measured. DJ-1 gene knockdown exacerbated cypermethrin-induced increase in oxidative stress and intrinsic apoptosis and reduction in expression of mitochondrial antioxidant proteins via inhibiting nuclear translocation of Nrf2. Additionally, cypermethrin-induced oxidative stress, mitochondrial impairment, and α-synuclein expression and aggregation were found to be suppressed by LRRK2 gene knockdown, by promoting Nrf2 nuclear translocation and expression of mitochondrial antioxidant proteins. Furthermore, Nrf2 activator, sulforaphane, ameliorated cypermethrin-induced mitochondrial impairment and oxidative stress and provided protection against dopaminergic neuronal death. The findings indicate that DJ-1 and LRRK2 independently alter Nrf2-mediated changes and a complex interplay among DJ-1, LRRK2, and Nrf2 exists in the regulation of mitochondrial function in cypermethrin-induced Parkinsonism.
Collapse
Affiliation(s)
- Alika Sarkar
- Toxicogenomics and Predictive Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Mahendra Pratap Singh
- Toxicogenomics and Predictive Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India.
| |
Collapse
|
10
|
Xue J, Tao K, Wang W, Wang X. What Can Inflammation Tell Us about Therapeutic Strategies for Parkinson's Disease? Int J Mol Sci 2024; 25:1641. [PMID: 38338925 PMCID: PMC10855787 DOI: 10.3390/ijms25031641] [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: 01/01/2024] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder with a complicated etiology and pathogenesis. α-Synuclein aggregation, dopaminergic (DA) neuron loss, mitochondrial injury, oxidative stress, and inflammation are involved in the process of PD. Neuroinflammation has been recognized as a key element in the initiation and progression of PD. In this review, we summarize the inflammatory response and pathogenic mechanisms of PD. Additionally, we describe the potential anti-inflammatory therapies, including nod-like receptor pyrin domain containing protein 3 (NLRP3) inflammasome inhibition, nuclear factor κB (NF-κB) inhibition, microglia inhibition, astrocyte inhibition, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibition, the peroxisome proliferator-activated receptor γ (PPARγ) agonist, targeting the mitogen-activated protein kinase (MAPK) pathway, targeting the adenosine monophosphate-activated protein kinase (AMPK)-dependent pathway, targeting α-synuclein, targeting miRNA, acupuncture, and exercise. The review focuses on inflammation and will help in designing new prevention strategies for PD.
Collapse
Affiliation(s)
- Jinsong Xue
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China; (K.T.); (W.W.)
| | | | | | - Xiaofei Wang
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China; (K.T.); (W.W.)
| |
Collapse
|
11
|
Morrone Parfitt G, Coccia E, Goldman C, Whitney K, Reyes R, Sarrafha L, Nam KH, Sohail S, Jones DR, Crary JF, Ordureau A, Blanchard J, Ahfeldt T. Disruption of lysosomal proteolysis in astrocytes facilitates midbrain organoid proteostasis failure in an early-onset Parkinson's disease model. Nat Commun 2024; 15:447. [PMID: 38200091 PMCID: PMC10781970 DOI: 10.1038/s41467-024-44732-2] [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: 10/05/2022] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Accumulation of advanced glycation end products (AGEs) on biopolymers accompanies cellular aging and drives poorly understood disease processes. Here, we studied how AGEs contribute to development of early onset Parkinson's Disease (PD) caused by loss-of-function of DJ1, a protein deglycase. In induced pluripotent stem cell (iPSC)-derived midbrain organoid models deficient for DJ1 activity, we find that lysosomal proteolysis is impaired, causing AGEs to accumulate, α-synuclein (α-syn) phosphorylation to increase, and proteins to aggregate. We demonstrated these processes are at least partly driven by astrocytes, as DJ1 loss reduces their capacity to provide metabolic support and triggers acquisition of a pro-inflammatory phenotype. Consistently, in co-cultures, we find that DJ1-expressing astrocytes are able to reverse the proteolysis deficits of DJ1 knockout midbrain neurons. In conclusion, astrocytes' capacity to clear toxic damaged proteins is critical to preserve neuronal function and their dysfunction contributes to the neurodegeneration observed in a DJ1 loss-of-function PD model.
Collapse
Affiliation(s)
- Gustavo Morrone Parfitt
- Nash Family Department of Neuroscience at Mount Sinai, New York, NY, USA.
- Ronald M. Loeb Center for Alzheimer's Disease at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute at Mount Sinai, New York, NY, USA.
- Black Family Stem Cell Institute at Mount Sinai, New York, NY, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA, 94080, USA.
| | - Elena Coccia
- Nash Family Department of Neuroscience at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease at Mount Sinai, New York, NY, USA
- Friedman Brain Institute at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute at Mount Sinai, New York, NY, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Camille Goldman
- Nash Family Department of Neuroscience at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease at Mount Sinai, New York, NY, USA
- Friedman Brain Institute at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute at Mount Sinai, New York, NY, USA
| | - Kristen Whitney
- Nash Family Department of Neuroscience at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease at Mount Sinai, New York, NY, USA
- Friedman Brain Institute at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular, and Cell-Based Medicine at Mount Sinai, New York, NY, USA
| | - Ricardo Reyes
- Nash Family Department of Neuroscience at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease at Mount Sinai, New York, NY, USA
- Friedman Brain Institute at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute at Mount Sinai, New York, NY, USA
| | - Lily Sarrafha
- Nash Family Department of Neuroscience at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease at Mount Sinai, New York, NY, USA
- Friedman Brain Institute at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute at Mount Sinai, New York, NY, USA
| | - Ki Hong Nam
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Soha Sohail
- Nash Family Department of Neuroscience at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease at Mount Sinai, New York, NY, USA
- Friedman Brain Institute at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute at Mount Sinai, New York, NY, USA
| | - Drew R Jones
- Metabolomics Core Resource Laboratory, NYU Langone Health, New York, NY, USA
| | - John F Crary
- Nash Family Department of Neuroscience at Mount Sinai, New York, NY, USA
- Friedman Brain Institute at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular, and Cell-Based Medicine at Mount Sinai, New York, NY, USA
| | - Alban Ordureau
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joel Blanchard
- Nash Family Department of Neuroscience at Mount Sinai, New York, NY, USA.
- Ronald M. Loeb Center for Alzheimer's Disease at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute at Mount Sinai, New York, NY, USA.
- Black Family Stem Cell Institute at Mount Sinai, New York, NY, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Tim Ahfeldt
- Nash Family Department of Neuroscience at Mount Sinai, New York, NY, USA.
- Ronald M. Loeb Center for Alzheimer's Disease at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute at Mount Sinai, New York, NY, USA.
- Black Family Stem Cell Institute at Mount Sinai, New York, NY, USA.
- Recursion Pharmaceuticals, Salt Lake City, UT, USA.
| |
Collapse
|
12
|
Buneeva OA, Kapitsa IG, Zgoda VG, Medvedev AE. Neuroprotective effects of isatin and afobazole in rats with rotenone-induced Parkinsonism are accompanied by increased brain levels of Triton X-100 soluble alpha-synuclein. BIOMEDITSINSKAIA KHIMIIA 2023; 69:290-299. [PMID: 37937431 DOI: 10.18097/pbmc20236905290] [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: 11/09/2023]
Abstract
Effects of the endogenous neuroprotector isatin and the pharmacological drug afobazole (exhibiting neuroprotective properties) on behavioral reactions and quantitative changes in the brain proteomic profile have been investigated in rats with experimental rotenone Parkinsonism. A single dose of isatin (100 mg/kg subcutaneously on the last day of a 7-day course of rotenone administration) improved the motor activity of rats with rotenone-induced Parkinsonism in the open field test (horizontal movements) and the rotating rod test. Afobazole (10 mg/kg intraperitoneally, daily during the 7-day course of rotenone administration) reduced the manifestations of rigidity and postural instability. Proteomic analysis, performed using brain samples obtained the day after the last administration of rotenone and neuroprotectors, revealed similar quantitative changes in the brain of rats with rotenone Parkinsonism. An increase in the relative content of 65 proteins and a decrease in the relative content of 21 proteins were detected. The most pronounced changes - an almost ninety-fold increase in the alpha-synuclein content - were found in the brains of rats treated with isatin. In animals of the experimental groups treated with "Rotenone + Isatin", as well as "Rotenone + Afobazole", the increase in the relative content of this protein in the brain was almost 60 and 50 times higher than the control values. Taking into consideration the known data on the physiological role of alpha-synuclein, an increase in the content of this protein in the brain upon administration of neuroprotectors to animals with rotenone Parkinsonism may represent a compensatory reaction, at least in the early stages of this disease and the beginning of its treatment.
Collapse
Affiliation(s)
- O A Buneeva
- Institute of Biomedical Chemistry, Moscow, Russia
| | - I G Kapitsa
- Institute of Biomedical Chemistry, Moscow, Russia; Zakusov Institute of Pharmacology, Moscow, Russia
| | - V G Zgoda
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A E Medvedev
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
13
|
Liu Y, Tan L, Tan MS. Chaperone-mediated autophagy in neurodegenerative diseases: mechanisms and therapy. Mol Cell Biochem 2023; 478:2173-2190. [PMID: 36695937 DOI: 10.1007/s11010-022-04640-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 12/09/2022] [Indexed: 01/26/2023]
Abstract
Chaperone-mediated autophagy (CMA) is the selective degradation process of intracellular components by lysosomes, which is required for the degradation of aggregate-prone proteins and contributes to proteostasis maintenance. Proteostasis is essential for normal cell function and survival, and it is determined by the balance of protein synthesis and degradation. Because postmitotic neurons are highly susceptible to proteostasis disruption, CMA is vital for the nervous system. Since Parkinson's disease (PD) was first linked to CMA dysfunction, an increasing number of studies have shown that CMA loss, as seen during aging, occurs in the pathogenetic process of neurodegenerative diseases. Here, we review the molecular mechanisms of CMA, as well as the physiological function and regulation of this autophagy pathway. Following, we highlight its potential role in neurodegenerative diseases, and the latest advances and challenges in targeting CMA in therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi Liu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.
| | - Meng-Shan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.
| |
Collapse
|
14
|
Yao R, Shen J. Chaperone-mediated autophagy: Molecular mechanisms, biological functions, and diseases. MedComm (Beijing) 2023; 4:e347. [PMID: 37655052 PMCID: PMC10466100 DOI: 10.1002/mco2.347] [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: 12/15/2022] [Revised: 07/23/2023] [Accepted: 07/27/2023] [Indexed: 09/02/2023] Open
Abstract
Chaperone-mediated autophagy (CMA) is a lysosomal degradation pathway that eliminates substrate proteins through heat-shock cognate protein 70 recognition and lysosome-associated membrane protein type 2A-assisted translocation. It is distinct from macroautophagy and microautophagy. In recent years, the regulatory mechanisms of CMA have been gradually enriched, including the newly discovered NRF2 and p38-TFEB signaling, as positive and negative regulatory pathways of CMA, respectively. Normal CMA activity is involved in the regulation of metabolism, aging, immunity, cell cycle, and other physiological processes, while CMA dysfunction may be involved in the occurrence of neurodegenerative disorders, tumors, intestinal disorders, atherosclerosis, and so on, which provides potential targets for the treatment and prediction of related diseases. This article describes the general process of CMA and its role in physiological activities and summarizes the connection between CMA and macroautophagy. In addition, human diseases that concern the dysfunction or protective role of CMA are discussed. Our review deepens the understanding of the mechanisms and physiological functions of CMA and provides a summary of past CMA research and a vision of future directions.
Collapse
Affiliation(s)
- Ruchen Yao
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of Health, Inflammatory Bowel Disease Research CenterShanghaiChina
- Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Institute of Digestive DiseaseShanghaiChina
| | - Jun Shen
- Division of Gastroenterology and HepatologyKey Laboratory of Gastroenterology and HepatologyMinistry of Health, Inflammatory Bowel Disease Research CenterShanghaiChina
- Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Institute of Digestive DiseaseShanghaiChina
| |
Collapse
|
15
|
Fagen SJ, Burgess JD, Lim MJ, Amerna D, Kaya ZB, Faroqi AH, Perisetla P, DeMeo NN, Stojkovska I, Quiriconi DJ, Mazzulli JR, Delenclos M, Boschen SL, McLean PJ. Honokiol decreases alpha-synuclein mRNA levels and reveals novel targets for modulating alpha-synuclein expression. Front Aging Neurosci 2023; 15:1179086. [PMID: 37637959 PMCID: PMC10449643 DOI: 10.3389/fnagi.2023.1179086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
Background Intracytoplasmic inclusions comprised of aggregated alpha-synuclein (αsyn) represent a key histopathological feature of neurological disorders collectively termed "synucleinopathies," which includes Parkinson's disease (PD). Mutations and multiplications in the SNCA gene encoding αsyn cause familial forms of PD and a large body of evidence indicate a correlation between αsyn accumulation and disease. Decreasing αsyn expression is recognized as a valid target for PD therapeutics, with down-regulation of SNCA expression potentially attenuating downstream cascades of pathologic events. Here, we evaluated if Honokiol (HKL), a polyphenolic compound derived from magnolia tree bark with demonstrated neuroprotective properties, can modulate αsyn levels in multiple experimental models. Methods Human neuroglioma cells stably overexpressing αsyn, mouse primary neurons, and human iPSC-derived neurons were exposed to HKL and αsyn protein and SNCA messenger RNA levels were assessed. The effect of HKL on rotenone-induced overexpression of αsyn levels was further assessed and transcriptional profiling of mouse cortical neurons treated with HKL was performed to identify potential targets of HKL. Results We demonstrate that HKL can successfully reduce αsyn protein levels and SNCA expression in multiple in vitro models of PD with our data supporting a mechanism whereby HKL acts by post-transcriptional modulation of SNCA rather than modulating αsyn protein degradation. Transcriptional profiling of mouse cortical neurons treated with HKL identifies several differentially expressed genes (DEG) as potential targets to modulate SNCA expression. Conclusion This study supports a HKL-mediated downregulation of SNCA as a viable strategy to modify disease progression in PD and other synucleinopathies. HKL has potential as a powerful tool for investigating SNCA gene modulation and its downstream effects.
Collapse
Affiliation(s)
- Sara J. Fagen
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Jeremy D. Burgess
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Melina J. Lim
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Danilyn Amerna
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Zeynep B. Kaya
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Ayman H. Faroqi
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Priyanka Perisetla
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Natasha N. DeMeo
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Iva Stojkovska
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Drew J. Quiriconi
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Joseph R. Mazzulli
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Marion Delenclos
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Suelen L. Boschen
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Rochester, MN, United States
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, United States
| | - Pamela J. McLean
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Rochester, MN, United States
| |
Collapse
|
16
|
Buneeva OA, Kapitsa IG, Kazieva LS, Vavilov NE, Zgoda VG. Quantitative changes of brain isatin-binding proteins of rats with the rotenone-induced experimental parkinsonism. BIOMEDITSINSKAIA KHIMIIA 2023; 69:188-192. [PMID: 37384911 DOI: 10.18097/pbmc20236903188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Isatin (indoldione-2,3) is an endogenous regulator found in humans and animals. It exhibits a broad range of biological activity mediated by numerous isatin-binding proteins. Isatin produces neuroprotective effects in several experimental models of diseases, including Parkinsonism induced by the neurotoxin MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine).Rotenone (a neurotoxin used to modeling Parkinson's disease in rodents) causes significant changes in the profile of isatin-binding proteins of rat brain. Comparative proteomic identification of brain proteins of control rats and the rats with the rotenone-induced Parkinsonian syndrome (PS) revealed significant quantitative changes of 86 proteins under the influence of rotenone. This neurotoxin mainly caused the increase of the quantity of proteins involved in signal transduction and regulation of enzyme activity (24), proteins involved in cytoskeleton formation and exocytosis (23), and enzymes involved in energy generation and carbohydrate metabolism (19). However, only 11 of these proteins referred to isatin-binding proteins; the content of eight of them increased while the content of three proteins decreased. This suggests that the dramatic change of the profile of isatin-binding proteins, found in the development of the rotenone-induced PS, comes from changes in the state of the pre-existing molecules of proteins, rather than altered expression of corresponding genes.
Collapse
Affiliation(s)
- O A Buneeva
- Institute of Biomedical Chemistry, Moscow, Russia
| | - I G Kapitsa
- Institute of Biomedical Chemistry, Moscow, Russia; Zakusov Institute of Pharmacology, Moscow, Russia
| | - L Sh Kazieva
- Institute of Biomedical Chemistry, Moscow, Russia
| | - N E Vavilov
- Institute of Biomedical Chemistry, Moscow, Russia
| | - V G Zgoda
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
17
|
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: 4] [Impact Index Per Article: 4.0] [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
|
18
|
Chavali LNM, Yddal I, Bifulco E, Mannsåker S, Røise D, Law JO, Frøyset AK, Grellscheid SN, Fladmark KE. Progressive Motor and Non-Motor Symptoms in Park7 Knockout Zebrafish. Int J Mol Sci 2023; 24:ijms24076456. [PMID: 37047429 PMCID: PMC10094626 DOI: 10.3390/ijms24076456] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
DJ-1 is a redox sensitive protein with a wide range of functions related to oxidative stress protection. Mutations in the park7 gene, which codes for DJ-1 are associated with early onset familial Parkinson’s disease and increased astrocytic DJ-1 levels are found in pathologic tissues from idiopathic Parkinson’s disease. We have previously established a DJ-1 knockout zebrafish line that developed normally, but with aging the DJ-1 null fish had a lowered level of tyrosine hydroxylase, respiratory mitochondrial failure and a lower body mass. Here we have examined the DJ-1 knockout from the early adult stage and show that loss of DJ-1 results in a progressive, age-dependent increase in both motoric and non-motoric symptoms associated to Parkinson’s disease. These changes coincide with changes in mitochondrial and mitochondrial associated proteins. Recent studies have suggested that a decline in NAD+ can contribute to Parkinson’s disease and that supplementation of NAD+ precursors may delay disease progression. We found that the brain NAD+/NADH ratio decreased in aging zebrafish but did not correlate with DJ-1 induced altered behavior. Differences were first observed at the late adult stage in which NAD+ and NADPH levels were decreased in DJ-1 knockouts. Considering the experimental power of zebrafish and the development of Parkinson’s disease-related symptoms in the DJ-1 null fish, this model can serve as a useful tool both to understand the progression of the disease and the effect of suggested treatments.
Collapse
|
19
|
Kim S, Pajarillo E, Nyarko-Danquah I, Aschner M, Lee E. Role of Astrocytes in Parkinson's Disease Associated with Genetic Mutations and Neurotoxicants. Cells 2023; 12:622. [PMID: 36831289 PMCID: PMC9953822 DOI: 10.3390/cells12040622] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/02/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons and the aggregation of Lewy bodies in the basal ganglia, resulting in movement impairment referred to as parkinsonism. However, the etiology of PD is not well known, with genetic factors accounting only for 10-15% of all PD cases. The pathogenetic mechanism of PD is not completely understood, although several mechanisms, such as oxidative stress and inflammation, have been suggested. Understanding the mechanisms of PD pathogenesis is critical for developing highly efficacious therapeutics. In the PD brain, dopaminergic neurons degenerate mainly in the basal ganglia, but recently emerging evidence has shown that astrocytes also significantly contribute to dopaminergic neuronal death. In this review, we discuss the role of astrocytes in PD pathogenesis due to mutations in α-synuclein (PARK1), DJ-1 (PARK7), parkin (PARK2), leucine-rich repeat kinase 2 (LRRK2, PARK8), and PTEN-induced kinase 1 (PINK1, PARK6). We also discuss PD experimental models using neurotoxins, such as paraquat, rotenone, 6-hydroxydopamine, and MPTP/MPP+. A more precise and comprehensive understanding of astrocytes' modulatory roles in dopaminergic neurodegeneration in PD will help develop novel strategies for effective PD therapeutics.
Collapse
Affiliation(s)
- Sanghoon Kim
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Edward Pajarillo
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Ivan Nyarko-Danquah
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Eunsook Lee
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|
20
|
Jokanović M, Oleksak P, Kuca K. Multiple neurological effects associated with exposure to organophosphorus pesticides in man. Toxicology 2023; 484:153407. [PMID: 36543276 DOI: 10.1016/j.tox.2022.153407] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/02/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
This article reviews available data regarding the possible association of organophosphorus (OP) pesticides with neurological disorders such as dementia, attention deficit hyperactivity disorder, neurodevelopment, autism, cognitive development, Parkinson's disease and chronic organophosphate-induced neuropsychiatric disorder. These effects mainly develop after repeated (chronic) human exposure to low doses of OP. In addition, three well defined neurotoxic effects in humans caused by single doses of OP compounds are discussed. Those effects are the cholinergic syndrome, the intermediate syndrome and organophosphate-induced delayed polyneuropathy. Usually, the poisoning can be avoided by an improved administrative control, limited access to OP pesticides, efficient measures of personal protection and education of OP pesticide applicators and medical staff.
Collapse
Affiliation(s)
- Milan Jokanović
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech republic
| | - Patrik Oleksak
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech republic; Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech republic.
| |
Collapse
|
21
|
Heng Y, Li YY, Wen L, Yan JQ, Chen NH, Yuan YH. Gastric Enteric Glial Cells: A New Contributor to the Synucleinopathies in the MPTP-Induced Parkinsonism Mouse. Molecules 2022; 27:7414. [PMID: 36364248 PMCID: PMC9656042 DOI: 10.3390/molecules27217414] [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: 10/06/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 05/19/2024] Open
Abstract
Accumulating evidence has shown that Parkinson's disease (PD) is a systemic disease other than a mere central nervous system (CNS) disorder. One of the most important peripheral symptoms is gastrointestinal dysfunction. The enteric nervous system (ENS) is regarded as an essential gateway to the environment. The discovery of the prion-like behavior of α-synuclein makes it possible for the neurodegenerative process to start in the ENS and spread via the gut-brain axis to the CNS. We first confirmed that synucleinopathies existed in the stomachs of chronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/probenecid (MPTP/p)-induced PD mice, as indicated by the significant increase in abnormal aggregated and nitrated α-synuclein in the TH-positive neurons and enteric glial cells (EGCs) of the gastric myenteric plexus. Next, we attempted to clarify the mechanisms in single MPTP-injected mice. The stomach naturally possesses high monoamine oxidase-B (MAO-B) activity and low superoxide dismutase (SOD) activity, making the stomach susceptible to MPTP-induced oxidative stress, as indicated by the significant increase in reactive oxygen species (ROS) in the stomach and elevated 4-hydroxynonenal (4-HNE) in the EGCs after MPTP exposure for 3 h. Additionally, stomach synucleinopathies appear before those of the nigrostriatal system, as determined by Western blotting 12 h after MPTP injection. Notably, nitrated α-synuclein was considerably increased in the EGCs after 3 h and 12 h of MPTP exposure. Taken together, our work demonstrated that the EGCs could be new contributors to synucleinopathies in the stomach. The early-initiated synucleinopathies might further influence neighboring neurons in the myenteric plexus and the CNS. Our results offer a new experimental clue for interpreting the etiology of PD.
Collapse
Affiliation(s)
- Yang Heng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yan-Yan Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lu Wen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jia-Qing Yan
- Department of Pharmacy, National Cancer Center/National, Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union, Medical College, Beijing 100021, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yu-He Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
22
|
Chen W, Liu H, Liu S, Kang Y, Nie Z, Lei H. Altered prefrontal neurochemistry in the DJ-1 knockout mouse model of Parkinson's disease: complementary semi-quantitative analyses with in vivo magnetic resonance spectroscopy and MALDI-MSI. Anal Bioanal Chem 2022; 414:7977-7987. [PMID: 36208327 DOI: 10.1007/s00216-022-04341-8] [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: 07/19/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 11/24/2022]
Abstract
In vivo proton magnetic resonance spectroscopy (1H-MRS) and matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) are two semi-quantitative analytical methods commonly used in neurochemical research. In this study, the two methods were used complementarily, in parallel, to investigate neurochemical perturbations in the medial prefrontal cortex (mPFC) of 9-month-old DJ-1 knockout mice, a well-established transgenic model for Parkinson's diseases. Convergingly, the results obtained with the two methods demonstrated that, compared with the wild-type (WT) mice, the DJ-1 knockout mice had significantly increased glutathione (GSH) level and GSH/glutamate (Glu) ratio in the mPFC, which likely presented an astrocytic compensatory mechanism in response to elevated regional oxidative stress induced by the loss of DJ-1 function. The results from this study also highlighted (1) the need to be cautious when interpreting the in vivo 1H-MRS results obtained from aged transgenic animals, in which the concentration of internal reference, being whether water or total creatine, could no longer be assumed to be the same as that in the age-matched WT animals, and (2) the necessity and importance of complementary analyses with more than one method under such circumstances.
Collapse
Affiliation(s)
- Wei Chen
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 30# Xiaohongshan West, Wuhan, 430071, Hubei, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Huihui Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Zhongguancun North First, Street 2, Beijing, 100190, China
| | - Sijie Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 30# Xiaohongshan West, Wuhan, 430071, Hubei, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Yan Kang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 30# Xiaohongshan West, Wuhan, 430071, Hubei, People's Republic of China
| | - Zongxiu Nie
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China. .,Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Zhongguancun North First, Street 2, Beijing, 100190, China.
| | - Hao Lei
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 30# Xiaohongshan West, Wuhan, 430071, Hubei, People's Republic of China. .,University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China.
| |
Collapse
|
23
|
Yin DX, Toyoda H, Nozaki K, Satoh K, Katagiri A, Adachi K, Kato T, Sato H. Taste Impairments in a Parkinson’s Disease Model Featuring Intranasal Rotenone Administration in Mice. JOURNAL OF PARKINSON'S DISEASE 2022; 12:1863-1880. [PMID: 35848036 PMCID: PMC9535587 DOI: 10.3233/jpd-223273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Taste impairments are often accompanied by olfactory impairments in the early stage of Parkinson’s disease (PD). The development of animal models is required to elucidate the mechanisms underlying taste impairments in PD. Objective: This study was conducted to clarify whether the intranasal administration of rotenone causes taste impairments prior to motor deficits in mice. Methods: Rotenone was administrated to the right nose of mice once a day for 1 or 4 week(s). In the 1-week group, taste, olfactory, and motor function was assessed before and after a 1-week recovery period following the rotenone administration. Motor function was also continuously examined in the 4-weeks group from 0 to 5 weeks. After a behavioral test, the number of catecholamine neurons (CA-Nos) was counted in the regions responsible for taste, olfactory, and motor function. Results: taste and olfactory impairments were simultaneously observed without locomotor impairments in the 1-week group. The CA-Nos was significantly reduced in the olfactory bulb and nucleus of the solitary tract. In the 4-week group, locomotor impairments were observed from the third week, and a significant reduction in the CA-Nos was observed in the substantia nigra (SN) and ventral tegmental area (VTA) at the fifth week along with the weight loss. Conclusion: The intranasal administration of rotenone caused chemosensory and motor impairments in an administration time-period dependent manner. Since chemosensory impairments were expressed prior to the locomotor impairments followed by SN/VTA CA neurons loss, this rotenone administration model may contribute to the clarification of the prodromal symptoms of PD.
Collapse
Affiliation(s)
- Dong Xu Yin
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Hiroki Toyoda
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Kazunori Nozaki
- Division of Medical Information, Osaka University Dental Hospital, Suita, Osaka, Japan
| | - Keitaro Satoh
- Division of Pharmacology, Meikai University School of Dentistry, Sakado, Saitama, Japan
| | - Ayano Katagiri
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Kazunori Adachi
- Division of Pharmacology, Meikai University School of Dentistry, Sakado, Saitama, Japan
| | - Takafumi Kato
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Hajime Sato
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
- Division of Pharmacology, Meikai University School of Dentistry, Sakado, Saitama, Japan
| |
Collapse
|
24
|
Cai L, Gao L, Zhang G, Zeng H, Wu X, Tan X, Qian C, Chen G. DJ-1 Alleviates Neuroinflammation and the Related Blood-Spinal Cord Barrier Destruction by Suppressing NLRP3 Inflammasome Activation via SOCS1/Rac1/ROS Pathway in a Rat Model of Traumatic Spinal Cord Injury. J Clin Med 2022; 11:jcm11133716. [PMID: 35807002 PMCID: PMC9267719 DOI: 10.3390/jcm11133716] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/19/2022] [Accepted: 06/22/2022] [Indexed: 02/06/2023] Open
Abstract
DJ-1 has been shown to play essential roles in neuronal protection and anti-inflammation in nervous system diseases. This study aimed to explore how DJ-1 regulates neuroinflammation after traumatic spinal cord injury (t-SCI). The rat model of spinal cord injury was established by the clamping method. The Basso, Beattie, Bresnahan (BBB) score and the inclined plane test (IPT) were used to evaluate neurological function. Western blot was then applied to test the levels of DJ-1, NLRP3, SOCS1, and related proinflammatory factors (cleaved caspase 1, IL-1β and IL-18); ROS level was also examined. The distribution of DJ-1 was assessed by immunofluorescence staining (IF). BSCB integrity was assessed by the level of MMP-9 and tight junction proteins (Claudin-5, Occludin and ZO-1). We found that DJ-1 became significantly elevated after t-SCI and was mainly located in neurons. Knockdown of DJ-1 with specific siRNA aggravated NLRP3 inflammasome-related neuroinflammation and strengthened the disruption of BSCB integrity. However, the upregulation of DJ-1 by Sodium benzoate (SB) reversed these effects and improved neurological function. Furthermore, SOCS1-siRNA attenuated the neuroprotective effects of DJ-1 and increased the ROS, Rac1 and NLRP3. In conclusion, DJ-1 may alleviate neuroinflammation and the related BSCB destruction after t-SCI by suppressing NLRP3 inflammasome activation by SOCS1/Rac1/ROS pathways. DJ-1 shows potential as a feasible target for mediating neuroinflammation after t-SCI.
Collapse
Affiliation(s)
- Lingxin Cai
- Department of Neurological Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (L.C.); (L.G.); (G.Z.); (H.Z.); (X.W.); (X.T.)
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou 310009, China
| | - Liansheng Gao
- Department of Neurological Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (L.C.); (L.G.); (G.Z.); (H.Z.); (X.W.); (X.T.)
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou 310009, China
| | - Guoqiang Zhang
- Department of Neurological Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (L.C.); (L.G.); (G.Z.); (H.Z.); (X.W.); (X.T.)
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou 310009, China
| | - Hanhai Zeng
- Department of Neurological Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (L.C.); (L.G.); (G.Z.); (H.Z.); (X.W.); (X.T.)
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou 310009, China
| | - Xinyan Wu
- Department of Neurological Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (L.C.); (L.G.); (G.Z.); (H.Z.); (X.W.); (X.T.)
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou 310009, China
| | - Xiaoxiao Tan
- Department of Neurological Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (L.C.); (L.G.); (G.Z.); (H.Z.); (X.W.); (X.T.)
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou 310009, China
| | - Cong Qian
- Department of Neurological Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (L.C.); (L.G.); (G.Z.); (H.Z.); (X.W.); (X.T.)
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou 310009, China
- Correspondence: (C.Q.); (G.C.)
| | - Gao Chen
- Department of Neurological Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (L.C.); (L.G.); (G.Z.); (H.Z.); (X.W.); (X.T.)
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou 310009, China
- Correspondence: (C.Q.); (G.C.)
| |
Collapse
|
25
|
Huang J, Li C, Shang H. Astrocytes in Neurodegeneration: Inspiration From Genetics. Front Neurosci 2022; 16:882316. [PMID: 35812232 PMCID: PMC9268899 DOI: 10.3389/fnins.2022.882316] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/06/2022] [Indexed: 12/19/2022] Open
Abstract
Despite the discovery of numerous molecules and pathologies, the pathophysiology of various neurodegenerative diseases remains unknown. Genetics participates in the pathogenesis of neurodegeneration. Neural dysfunction, which is thought to be a cell-autonomous mechanism, is insufficient to explain the development of neurodegenerative disease, implying that other cells surrounding or related to neurons, such as glial cells, are involved in the pathogenesis. As the primary component of glial cells, astrocytes play a variety of roles in the maintenance of physiological functions in neurons and other glial cells. The pathophysiology of neurodegeneration is also influenced by reactive astrogliosis in response to central nervous system (CNS) injuries. Furthermore, those risk-gene variants identified in neurodegenerations are involved in astrocyte activation and senescence. In this review, we summarized the relationships between gene variants and astrocytes in four neurodegenerative diseases, including Alzheimer’s disease (AD), amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and Parkinson’s disease (PD), and provided insights into the implications of astrocytes in the neurodegenerations.
Collapse
|
26
|
Rocha SM, Bantle CM, Aboellail T, Chatterjee D, Smeyne RJ, Tjalkens RB. Rotenone induces regionally distinct α-synuclein protein aggregation and activation of glia prior to loss of dopaminergic neurons in C57Bl/6 mice. Neurobiol Dis 2022; 167:105685. [DOI: 10.1016/j.nbd.2022.105685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/14/2022] [Accepted: 03/02/2022] [Indexed: 12/21/2022] Open
|
27
|
Le S, Fu X, Pang M, Zhou Y, Yin G, Zhang J, Fan D. The Antioxidative Role of Chaperone-Mediated Autophagy as a Downstream Regulator of Oxidative Stress in Human Diseases. Technol Cancer Res Treat 2022; 21:15330338221114178. [PMID: 36131551 PMCID: PMC9500268 DOI: 10.1177/15330338221114178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Chaperone-mediated autophagy (CMA) plays an important role in regulating a variety of cellular functions by selectively degrading damaged or functional proteins in the cytoplasm. One of the cellular processes in which CMA participates is the oxidative stress response. Oxidative stress regulates CMA activity, while CMA protects cells from oxidative damage by degrading oxidized proteins and preventing the accumulation of excessive reactive oxygen species (ROS). Changes in CMA activity have been found in many human diseases, and oxidative stress is also involved. Therefore, understanding the interaction mechanism of ROS and CMA will provide new targets for disease treatment. In this review, we discuss the role of CMA in combatting oxidative stress during the development of different conditions, such as aging, neurodegeneration, liver diseases, infections, pulmonary disorders, and cancers.
Collapse
Affiliation(s)
- Shuangshuang Le
- Guangxi Key Laboratory of Bio-Targeting Theranostics, National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, 74626Guangxi Medical University, Nanning, China.,State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, 12644Air Force Military Medical University, Xi'an, China
| | - Xin Fu
- Guangxi Key Laboratory of Bio-Targeting Theranostics, National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, 74626Guangxi Medical University, Nanning, China.,State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, 12644Air Force Military Medical University, Xi'an, China
| | - Maogui Pang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, 12644Air Force Military Medical University, Xi'an, China
| | - Yao Zhou
- Guangxi Key Laboratory of Bio-Targeting Theranostics, National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, 74626Guangxi Medical University, Nanning, China.,State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, 12644Air Force Military Medical University, Xi'an, China
| | - Guoqing Yin
- Department of Oncology, 572481Xianyang Hospital of Yan'an University, Xianyang, China
| | - Jie Zhang
- Department of Oncology, 572481Xianyang Hospital of Yan'an University, Xianyang, China
| | - Daiming Fan
- Guangxi Key Laboratory of Bio-Targeting Theranostics, National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, 74626Guangxi Medical University, Nanning, China.,State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, 12644Air Force Military Medical University, Xi'an, China
| |
Collapse
|
28
|
Yuan X, Tian Y, Liu C, Zhang Z. Environmental factors in Parkinson's disease: New insights into the molecular mechanisms. Toxicol Lett 2021; 356:1-10. [PMID: 34864130 DOI: 10.1016/j.toxlet.2021.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/25/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023]
Abstract
Parkinson's disease is a chronic, progressive neurodegenerative disorder affecting 2-3% of the population ≥65 years. It has long been characterized by motor impairment, autonomic dysfunction, and psychological and cognitive changes. The pathological hallmarks are intracellular inclusions containing α-synuclein aggregates and the loss of dopaminergic neurons in the substantia nigra. Parkinson's disease is thought to be caused by a combination of various pathogenic factors, including genetic factors, environmental factors, and lifestyles. Although much research has focused on the genetic causes of PD, environmental risk factors also play a crucial role in the development of the disease. Here, we summarize the environmental risk factors that may increase the occurrence of PD, as well as the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Xin Yuan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ye Tian
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chaoyang Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, 430073, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
29
|
Gharbi N, Røise D, Førre JE, Edson AJ, Hushagen HA, Tronci V, Frøyset AK, Fladmark KE. Reintroduction of DJ-1 in Müller Cells Inhibits Retinal Degeneration in the DJ-1 Deficient Retina. Antioxidants (Basel) 2021; 10:1862. [PMID: 34942966 PMCID: PMC8698414 DOI: 10.3390/antiox10121862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 12/19/2022] Open
Abstract
The eye is continuously under oxidative stress due to high metabolic activity and reactive oxygen species generated by daily light exposure. The redox-sensitive protein DJ-1 has proven to be essential in order to protect retina and retinal pigment epithelium (RPE) from oxidative-stress-induced degeneration. Here, we analyzed the specific role of Müller cell DJ-1 in the adult zebrafish retina by re-establishing Müller-cell-specific DJ-1 expression in a DJ-1 knockout retina. Loss of DJ-1 resulted in an age-dependent retinal degeneration, including loss of cells in the ganglion cell layer, retinal thinning, photoreceptor disorganization and RPE cell dysfunction. The degenerative phenotype induced by the absence of DJ-1 was inhibited by solely expressing DJ-1 in Müller cells. The protective effect was dependent upon the cysteine-106 residue of DJ-1, which has been shown to be an oxidative sensor of DJ-1. In a label-free proteomics analysis of isolated retinas, we identified proteins differentially expressed after DJ-1 knockout, but with restored levels after Müller cell DJ-1 re-insertion. Our data show that Müller cell DJ-1 has a major role in protecting the retina from age-dependent oxidative stress.
Collapse
Affiliation(s)
- Naouel Gharbi
- Integrative Fish Biology Group (IFB), NORCE Norwegian Research Center AS, N-5020 Bergen, Norway; (N.G.); (V.T.)
| | - Dagne Røise
- Department of Biological Science, University of Bergen, N-5020 Bergen, Norway; (D.R.); (J.-E.F.); (A.J.E.); (H.A.H.); (A.-K.F.)
| | - Jorunn-Elise Førre
- Department of Biological Science, University of Bergen, N-5020 Bergen, Norway; (D.R.); (J.-E.F.); (A.J.E.); (H.A.H.); (A.-K.F.)
| | - Amanda J. Edson
- Department of Biological Science, University of Bergen, N-5020 Bergen, Norway; (D.R.); (J.-E.F.); (A.J.E.); (H.A.H.); (A.-K.F.)
| | - Helena A. Hushagen
- Department of Biological Science, University of Bergen, N-5020 Bergen, Norway; (D.R.); (J.-E.F.); (A.J.E.); (H.A.H.); (A.-K.F.)
| | - Valentina Tronci
- Integrative Fish Biology Group (IFB), NORCE Norwegian Research Center AS, N-5020 Bergen, Norway; (N.G.); (V.T.)
| | - Ann-Kristin Frøyset
- Department of Biological Science, University of Bergen, N-5020 Bergen, Norway; (D.R.); (J.-E.F.); (A.J.E.); (H.A.H.); (A.-K.F.)
| | - Kari E. Fladmark
- Department of Biological Science, University of Bergen, N-5020 Bergen, Norway; (D.R.); (J.-E.F.); (A.J.E.); (H.A.H.); (A.-K.F.)
| |
Collapse
|
30
|
Autophagy in α-Synucleinopathies-An Overstrained System. Cells 2021; 10:cells10113143. [PMID: 34831366 PMCID: PMC8618716 DOI: 10.3390/cells10113143] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 01/01/2023] Open
Abstract
Alpha-synucleinopathies comprise progressive neurodegenerative diseases, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). They all exhibit the same pathological hallmark, which is the formation of α-synuclein positive deposits in neuronal or glial cells. The aggregation of α-synuclein in the cell body of neurons, giving rise to the so-called Lewy bodies (LBs), is the major characteristic for PD and DLB, whereas the accumulation of α-synuclein in oligodendroglial cells, so-called glial cytoplasmic inclusions (GCIs), is the hallmark for MSA. The mechanisms involved in the intracytoplasmic inclusion formation in neuronal and oligodendroglial cells are not fully understood to date. A possible mechanism could be an impaired autophagic machinery that cannot cope with the high intracellular amount of α-synuclein. In fact, different studies showed that reduced autophagy is involved in α-synuclein aggregation. Furthermore, altered levels of different autophagy markers were reported in PD, DLB, and MSA brains. To date, the trigger point in disease initiation is not entirely clear; that is, whether autophagy dysfunction alone suffices to increase α-synuclein or whether α-synuclein is the pathogenic driver. In the current review, we discuss the involvement of defective autophagy machinery in the formation of α-synuclein aggregates, propagation of α-synuclein, and the resulting neurodegenerative processes in α-synucleinopathies.
Collapse
|
31
|
Wang C, Yang T, Liang M, Xie J, Song N. Astrocyte dysfunction in Parkinson's disease: from the perspectives of transmitted α-synuclein and genetic modulation. Transl Neurodegener 2021; 10:39. [PMID: 34657636 PMCID: PMC8522040 DOI: 10.1186/s40035-021-00265-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/05/2021] [Indexed: 01/20/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder that primarily affects the elderly. While the etiology of PD is likely multifactorial with the involvement of genetic, environmental, aging and other factors, α-synuclein (α-syn) pathology is a pivotal mechanism underlying the development of PD. In recent years, astrocytes have attracted considerable attention in the field. Although astrocytes perform a variety of physiological functions in the brain, they are pivotal mediators of α-syn toxicity since they internalize α-syn released from damaged neurons, and this triggers an inflammatory response, protein degradation dysfunction, mitochondrial dysfunction and endoplasmic reticulum stress. Astrocytes are indispensable coordinators in the background of several genetic mutations, including PARK7, GBA1, LRRK2, ATP13A2, PINK1, PRKN and PLA2G6. As the most abundant glial cells in the brain, functional astrocytes can be replenished and even converted to functional neurons. In this review, we discuss astrocyte dysfunction in PD with an emphasis on α-syn toxicity and genetic modulation and conclude that astrocyte replenishment is a valuable therapeutic approach in PD.
Collapse
Affiliation(s)
- Changjing Wang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Tongtong Yang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Meiyu Liang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Junxia Xie
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China.
| | - Ning Song
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
32
|
Sanchez A, Morales I, Rodriguez-Sabate C, Sole-Sabater M, Rodriguez M. Astrocytes, a Promising Opportunity to Control the Progress of Parkinson's Disease. Biomedicines 2021; 9:biomedicines9101341. [PMID: 34680458 PMCID: PMC8533570 DOI: 10.3390/biomedicines9101341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 12/21/2022] Open
Abstract
At present, there is no efficient treatment to prevent the evolution of Parkinson’s disease (PD). PD is generated by the concurrent activity of multiple factors, which is a serious obstacle for the development of etio-pathogenic treatments. Astrocytes may act on most factors involved in PD and the promotion of their neuroprotection activity may be particularly suitable to prevent the onset and progression of this basal ganglia (BG) disorder. The main causes proposed for PD, the ability of astrocytes to control these causes, and the procedures that can be used to promote the neuroprotective action of astrocytes will be commented upon, here.
Collapse
Affiliation(s)
- Alberto Sanchez
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna, 38200 Tenerife, Spain; (A.S.); (I.M.); (C.R.-S.)
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Ingrid Morales
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna, 38200 Tenerife, Spain; (A.S.); (I.M.); (C.R.-S.)
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Clara Rodriguez-Sabate
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna, 38200 Tenerife, Spain; (A.S.); (I.M.); (C.R.-S.)
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Department of Psychiatry, Getafe University Hospital, 28905 Madrid, Spain
| | - Miguel Sole-Sabater
- Department of Neurology, La Candelaria University Hospital, 38010 Tenerife, Spain;
| | - Manuel Rodriguez
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna, 38200 Tenerife, Spain; (A.S.); (I.M.); (C.R.-S.)
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Correspondence: ; Tel.: +34-922-319361; Fax: +34-922-319397
| |
Collapse
|
33
|
Hanslik KL, Marino KM, Ulland TK. Modulation of Glial Function in Health, Aging, and Neurodegenerative Disease. Front Cell Neurosci 2021; 15:718324. [PMID: 34531726 PMCID: PMC8439422 DOI: 10.3389/fncel.2021.718324] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/27/2021] [Indexed: 01/15/2023] Open
Abstract
In the central nervous system (CNS), glial cells, such as microglia and astrocytes, are normally associated with support roles including contributions to energy metabolism, synaptic plasticity, and ion homeostasis. In addition to providing support for neurons, microglia and astrocytes function as the resident immune cells in the brain. The glial function is impacted by multiple aspects including aging and local CNS changes caused by neurodegeneration. During aging, microglia and astrocytes display alterations in their homeostatic functions. For example, aged microglia and astrocytes exhibit impairments in the lysosome and mitochondrial function as well as in their regulation of synaptic plasticity. Recent evidence suggests that glia can also alter the pathology associated with many neurodegenerative disorders including Alzheimer's disease (AD) and Parkinson's disease (PD). Shifts in the microbiome can impact glial function as well. Disruptions in the microbiome can lead to aberrant microglial and astrocytic reactivity, which can contribute to an exacerbation of disease and neuronal dysfunction. In this review, we will discuss the normal physiological functions of microglia and astrocytes, summarize novel findings highlighting the role of glia in aging and neurodegenerative diseases, and examine the contribution of microglia and astrocytes to disease progression.
Collapse
Affiliation(s)
- Kendra L. Hanslik
- Neuroscience Training Program, University of Wisconsin, Madison, WI, United States
| | - Kaitlyn M. Marino
- Neuroscience Training Program, University of Wisconsin, Madison, WI, United States
| | - Tyler K. Ulland
- Neuroscience Training Program, University of Wisconsin, Madison, WI, United States
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
34
|
Latif S, Jahangeer M, Maknoon Razia D, Ashiq M, Ghaffar A, Akram M, El Allam A, Bouyahya A, Garipova L, Ali Shariati M, Thiruvengadam M, Azam Ansari M. Dopamine in Parkinson's disease. Clin Chim Acta 2021; 522:114-126. [PMID: 34389279 DOI: 10.1016/j.cca.2021.08.009] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/04/2021] [Accepted: 08/07/2021] [Indexed: 12/12/2022]
Abstract
Parkinson's disease is a neurodegenerative disease caused by the death of neurons, ie, cells critical to the production of dopamine, an important neurotransmitter in the brain. Here, we present a brief review of the dopamine synthetic pathway, binding to the dopamine receptors, and subsequent action. The production of dopamine (a monoamine neurotransmitter) occurs in the ventral tegmental area (VTA) of the substantia nigra, specifically in the hypothalamic nucleus and midbrain. Compared to other monoamines, dopamine is widely distributed in the olfactory bulb, midbrain substantia nigra, hypothalamus, VTA, retina, and the periaqueductal gray area. Dopamine receptors are large G-protein coupled receptor family members, of which there are five subtypes including D1, D2, D3, D4, and D5. These subtypes are further divided into two subclasses: D1-like family receptors (types 1 and 5) and D2-like family receptors (types 2, 3, and 4). Four different pathways and functions of the dopaminergic system are presented in this review. In the oxidation of dopamine, 5,6-indolequinone, dopamine-o-quinone, and aminochrome are formed. It is difficult to separate the roles of 5,6-indolequinone and dopamine-o-quinone in the degenerative process of Parkinson's diseases due to their instability. The role of aminochrome in Parkinson's disease is to form and stabilize the neurotoxic protofibrils of alpha-synuclein, mitochondrial dysfunction, oxidative stress, and the degradation of protein by lysosomal systems and proteasomes. The neurotoxic effects of aminochrome can be inhibited by preventing the polymerization of 5,6-indolequinone, dopamine-o-quinone, and aminochrome into neuromelanin, by reducing aminochrome catalysis by DT-diaphorase, and by preventing dopamine oxidative deamination catalyzed by monoamine oxidase. In addition to these, the conversion of dopamine in the neuromelanin (NM) shows both protective and toxic roles. Therefore, the aims of this review were to discuss and explain the role of dopamine and explore its physiology and specificity in Parkinson's disease, as well as its role in other physiological functions.
Collapse
Affiliation(s)
- Saad Latif
- Department of Biochemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Muhammad Jahangeer
- Department of Biochemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Dure Maknoon Razia
- Department of Biochemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Mehvish Ashiq
- Department of Chemistry, The Women University Multan, Multan, Pakistan
| | - Abdul Ghaffar
- Department of Biochemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan.
| | - Muhammad Akram
- Department of Eastern Medicine, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Aicha El Allam
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, And Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, And Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Larisa Garipova
- K.G. Razumovsky Moscow State University of technologies and management (the First Cossack University), 73 Zemlyanoy Val, Moscow 109004, Russian Federation
| | - Mohammad Ali Shariati
- K.G. Razumovsky Moscow State University of technologies and management (the First Cossack University), 73 Zemlyanoy Val, Moscow 109004, Russian Federation
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul 05029, Republic of Korea.
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research & Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
35
|
Buneeva OA, Medvedev AE. DJ-1 Protein and Its Role in the Development of Parkinson's Disease: Studies on Experimental Models. BIOCHEMISTRY (MOSCOW) 2021; 86:627-640. [PMID: 34225587 DOI: 10.1134/s000629792106002x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
DJ-1, also known as Parkinson's disease protein 7, is a multifunctional protein ubiquitously expressed in cells and tissues. Interacting with proteins of various intracellular compartments, DJ-1 plays an important role in maintaining different cellular functions. Mutant DJ-1 forms containing amino acid substitutions (especially L166P), typical of Parkinson's disease, are characterized by impaired dimerization, stability, and folding. DJ-1 exhibits several types of catalytic activity; however, in the enzyme classification it exists as protein deglycase (EC 3.5.1.124). Apparently, in different cell compartments DJ-1 exhibits catalytic and non-catalytic functions, and their ratio still remains unknown. Oxidative stress promotes dissociation of cytoplasmic DJ-1 dimers into monomers, which are translocated to the nucleus, where this protein acts as a coactivator of various signaling pathways, preventing cell death. In mitochondria, DJ-1 is found in the synthasome, where it interacts with the β ATP synthase subunit. Downregulation of the DJ-1 gene under conditions of experimental PD increases sensitivity of the cells to neurotoxins, and introduction of the recombinant DJ-1 protein attenuates manifestation of this pathology. The thirteen-membered fragment of the DJ-1 amino acid sequence attached to the heptapeptide of the TAT protein penetrating into the cells exhibited neuroprotective properties in various PD models both in cell cultures and after administration to animals. Low molecular weight DJ-1 ligands also demonstrate therapeutic potential, providing neuroprotective effects seen during their incubation with cells and administration to animals.
Collapse
Affiliation(s)
- Olga A Buneeva
- Institute of Biomedical Chemistry, Moscow, 119121, Russia
| | | |
Collapse
|
36
|
Huang M, Chen S. DJ-1 in neurodegenerative diseases: Pathogenesis and clinical application. Prog Neurobiol 2021; 204:102114. [PMID: 34174373 DOI: 10.1016/j.pneurobio.2021.102114] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/22/2021] [Accepted: 06/21/2021] [Indexed: 12/23/2022]
Abstract
Neurodegenerative diseases (NDs) are one of the major health threats to human characterized by selective and progressive neuronal loss. The mechanisms of NDs are still not fully understood. The study of genetic defects and disease-related proteins offers us a window into the mystery of it, and the extension of knowledge indicates that different NDs share similar features, mechanisms, and even genetic or protein abnormalities. Among these findings, PARK7 and its production DJ-1 protein, which was initially found implicated in PD, have also been found altered in other NDs. PARK7 mutations, altered expression and posttranslational modification (PTM) cause DJ-1 abnormalities, which in turn lead to downstream mechanisms shared by most NDs, such as mitochondrial dysfunction, oxidative stress, protein aggregation, autophagy defects, and so on. The knowledge of DJ-1 derived from PD researches might apply to other NDs in both basic research and clinical application, and might yield novel insights into and alternative approaches for dealing with NDs.
Collapse
Affiliation(s)
- Maoxin Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China; Lab for Translational Research of Neurodegenerative Diseases, Institute of Immunochemistry, Shanghai Tech University, 201210, Shanghai, China.
| |
Collapse
|
37
|
Ren H, Zhai W, Lu X, Wang G. The Cross-Links of Endoplasmic Reticulum Stress, Autophagy, and Neurodegeneration in Parkinson's Disease. Front Aging Neurosci 2021; 13:691881. [PMID: 34168552 PMCID: PMC8218021 DOI: 10.3389/fnagi.2021.691881] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/10/2021] [Indexed: 12/17/2022] Open
Abstract
Parkinson’s disease (PD) is the most common neurodegenerative movement disorder, and it is characterized by the selective loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc), as well as the presence of intracellular inclusions with α-synuclein as the main component in surviving DA neurons. Emerging evidence suggests that the imbalance of proteostasis is a key pathogenic factor for PD. Endoplasmic reticulum (ER) stress-induced unfolded protein response (UPR) and autophagy, two major pathways for maintaining proteostasis, play important roles in PD pathology and are considered as attractive therapeutic targets for PD treatment. However, although ER stress/UPR and autophagy appear to be independent cellular processes, they are closely related to each other. In this review, we focused on the roles and molecular cross-links between ER stress/UPR and autophagy in PD pathology. We systematically reviewed and summarized the most recent advances in regulation of ER stress/UPR and autophagy, and their cross-linking mechanisms. We also reviewed and discussed the mechanisms of the coexisting ER stress/UPR activation and dysregulated autophagy in the lesion regions of PD patients, and the underlying roles and molecular crosslinks between ER stress/UPR activation and the dysregulated autophagy in DA neurodegeneration induced by PD-associated genetic factors and PD-related neurotoxins. Finally, we indicate that the combined regulation of ER stress/UPR and autophagy would be a more effective treatment for PD rather than regulating one of these conditions alone.
Collapse
Affiliation(s)
- Haigang Ren
- Department of Neurology, Center of Translational Medicine, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Suzhou, China.,Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Wanqing Zhai
- Department of Neurology, Center of Translational Medicine, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Suzhou, China
| | - Xiaojun Lu
- Department of Neurology, Center of Translational Medicine, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Suzhou, China
| | - Guanghui Wang
- Department of Neurology, Center of Translational Medicine, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Suzhou, China.,Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
38
|
Buck SA, De Miranda BR, Logan RW, Fish KN, Greenamyre JT, Freyberg Z. VGLUT2 Is a Determinant of Dopamine Neuron Resilience in a Rotenone Model of Dopamine Neurodegeneration. J Neurosci 2021; 41:4937-4947. [PMID: 33893220 PMCID: PMC8260163 DOI: 10.1523/jneurosci.2770-20.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is characterized by progressive dopamine (DA) neuron loss in the SNc. In contrast, DA neurons in the VTA are relatively protected from neurodegeneration, but the underlying mechanisms for this resilience remain poorly understood. Recent work suggests that expression of the vesicular glutamate transporter 2 (VGLUT2) selectively impacts midbrain DA neuron vulnerability. We investigated whether altered DA neuron VGLUT2 expression determines neuronal resilience in rats exposed to rotenone, a mitochondrial complex I inhibitor and toxicant model of PD. We discovered that VTA/SNc DA neurons that expressed VGLUT2 are more resilient to rotenone-induced DA neurodegeneration. Surprisingly, the density of neurons with detectable VGLUT2 expression in the VTA and SNc increases in response to rotenone. Furthermore, dopaminergic terminals within the NAc, where the majority of VGLUT2-expressing DA neurons project, exhibit greater resilience compared with DA terminals in the caudate/putamen. More broadly, VGLUT2-expressing terminals are protected throughout the striatum from rotenone-induced degeneration. Together, our data demonstrate that a distinct subpopulation of VGLUT2-expressing DA neurons are relatively protected from rotenone neurotoxicity. Rotenone-induced upregulation of the glutamatergic machinery in VTA and SNc neurons and their projections may be part of a broader neuroprotective mechanism. These findings offer a putative new target for neuronal resilience that can be manipulated to prevent toxicant-induced DA neurodegeneration in PD.SIGNIFICANCE STATEMENT Environmental exposures to pesticides contribute significantly to pathologic processes that culminate in Parkinson's disease (PD). The pesticide rotenone has been used to generate a PD model that replicates key features of the illness, including dopamine neurodegeneration. To date, longstanding questions remain: are there dopamine neuron subpopulations resilient to rotenone; and if so, what are the molecular determinants of this resilience? Here we show that the subpopulation of midbrain dopaminergic neurons that express the vesicular glutamate transporter 2 (VGLUT2) are more resilient to rotenone-induced neurodegeneration. Rotenone also upregulates VGLUT2 more broadly in the midbrain, suggesting that VGLUT2 expression generally confers increased resilience to rotenone. VGLUT2 may therefore be a new target for boosting neuronal resilience to prevent toxicant-induced DA neurodegeneration in PD.
Collapse
Affiliation(s)
- Silas A Buck
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| | - Briana R De Miranda
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Ryan W Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, 02118
- Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, Maine, 04609
| | - Kenneth N Fish
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| | - J Timothy Greenamyre
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| |
Collapse
|
39
|
Jing L, Hou L, Zhang D, Li S, Ruan Z, Zhang X, Hong JS, Wang Q. Microglial Activation Mediates Noradrenergic Locus Coeruleus Neurodegeneration via Complement Receptor 3 in a Rotenone-Induced Parkinson's Disease Mouse Model. J Inflamm Res 2021; 14:1341-1356. [PMID: 33859489 PMCID: PMC8044341 DOI: 10.2147/jir.s299927] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/24/2021] [Indexed: 12/17/2022] Open
Abstract
Background Chronic exposure to the insecticide rotenone can damage dopaminergic neurons and lead to an increased risk of Parkinson’s disease (PD). Whereas it is not clear whether rotenone induces neurodegeneration of noradrenergic locus coeruleus (LC/NE) neurons. Chronic neuroinflammation mediated by microglia has been involved in the pathogenesis of PD. Evidence shows that complement receptor 3 (CR3) is a crucial regulator of microglial activation and related neurodegeneration. However, it is not clear whether CR3 mediates rotenone-elicited degeneration of LC/NE neurons through microglia-mediated neuroinflammation. Materials and Methods Wild type (WT) and CR3 knockout (KO) mice were treated with rotenone. PLX3397 and minocycline were used to deplete or inactivate the microglia. Leukadherin-1 (LA-1) was used to modulate CR3. LC/NE neurodegeneration, microglial phenotype, and expression of CR3 were determined by using immunohistochemistry, Western blot and real-time polymerase chain reaction (PCR) techniques. The glutathione (GSH) and malondialdehyde (MDA) contents were measured by using commercial kits. Results Rotenone exposure led to dose- and time-dependent LC/NE neuronal loss and microglial activation in mice. Depletion of microglia by PLX3397 or inhibition of microglial activation by minocycline significantly reduced rotenone-induced LC/NE neurodegeneration. Mechanistic studies revealed that CR3 played an essential role in the rotenone-induced activation of microglia and neurodegeneration of LC/NE neurons. Rotenone elevated the expression of CR3, and genetic ablation of CR3 markedly reduced rotenone-induced microglial activation and M1 polarization. LA-1 also suppressed rotenone-induced toxic microglial M1 activation. Furthermore, lack of CR3 or treatment with LA-1 reduced oxidative stress in the brainstem of rotenone-intoxicated mice. Finally, we found that mice deficient in CR3 or treated with LA-1 were more resistant to rotenone-induced LC/NE neurodegeneration than WT or vehicle-treated mice, respectively. Conclusion Our results indicate that CR3-mediated microglial activation participates in rotenone-induced LC/NE neurodegeneration, providing novel insight into environmental toxin-induced neurotoxicity and related Parkinsonism.
Collapse
Affiliation(s)
- Lu Jing
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China.,Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Liyan Hou
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Dongdong Zhang
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Sheng Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Zhengzheng Ruan
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Xiaomeng Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Qingshan Wang
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China.,National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, People's Republic of China
| |
Collapse
|
40
|
De Miranda BR, Castro SL, Rocha EM, Bodle CR, Johnson KE, Greenamyre JT. The industrial solvent trichloroethylene induces LRRK2 kinase activity and dopaminergic neurodegeneration in a rat model of Parkinson's disease. Neurobiol Dis 2021; 153:105312. [PMID: 33636387 DOI: 10.1016/j.nbd.2021.105312] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/05/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
Gene-environment interaction is implicated in the majority of idiopathic Parkinson's disease (PD) risk, and some of the most widespread environmental contaminants are selectively toxic to dopaminergic neurons. Pesticides have long been connected to PD incidence, however, it has become increasingly apparent that other industrial byproducts likely influence neurodegeneration. For example, organic solvents, which are used in chemical, machining, and dry-cleaning industries, are of growing concern, as decades of solvent use and their effluence into the environment has contaminated much of the world's groundwater and soil. Like some pesticides, certain organic solvents, such as the chlorinated halocarbon trichloroethylene (TCE), are mitochondrial toxicants, which are collectively implicated in the pathogenesis of dopaminergic neurodegeneration. Recently, we hypothesized a possible gene-environment interaction may occur between environmental mitochondrial toxicants and the protein kinase LRRK2, mutations of which are the most common genetic cause of familial and sporadic PD. In addition, emerging data suggests that elevated wildtype LRRK2 kinase activity also contributes to the pathogenesis of idiopathic PD. To this end, we investigated whether chronic, systemic TCE exposure (200 mg/kg) in aged rats produced wildtype LRRK2 activation and caused nigrostriatal dopaminergic dysfunction. Interestingly, we found that TCE not only induced LRRK2 kinase activity in the brain, but produced a significant dopaminergic lesion in the nigrostriatal tract, elevated oxidative stress, and caused endolysosomal dysfunction and α-synuclein accumulation. Together, these data suggest that TCE-induced LRRK2 kinase activity contributed to the selective toxicity of dopaminergic neurons. We conclude that gene-environment interactions between certain industrial contaminants and LRRK2 likely influence PD risk.
Collapse
Affiliation(s)
- Briana R De Miranda
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America.
| | - Sandra L Castro
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Emily M Rocha
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Christopher R Bodle
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Katrina E Johnson
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
41
|
Auzmendi-Iriarte J, Matheu A. Impact of Chaperone-Mediated Autophagy in Brain Aging: Neurodegenerative Diseases and Glioblastoma. Front Aging Neurosci 2021; 12:630743. [PMID: 33633561 PMCID: PMC7901968 DOI: 10.3389/fnagi.2020.630743] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022] Open
Abstract
Brain aging is characterized by a time-dependent decline of tissue integrity and function, and it is a major risk for neurodegenerative diseases and brain cancer. Chaperone-mediated autophagy (CMA) is a selective form of autophagy specialized in protein degradation, which is based on the individual translocation of a cargo protein through the lysosomal membrane. Regulation of processes such as proteostasis, cellular energetics, or immune system activity has been associated with CMA, indicating its pivotal role in tissue homeostasis. Since first studies associating Parkinson’s disease (PD) to CMA dysfunction, increasing evidence points out that CMA is altered in both physiological and pathological brain aging. In this review article, we summarize the current knowledge regarding the impact of CMA during aging in brain physiopathology, highlighting the role of CMA in neurodegenerative diseases and glioblastoma, the most common and aggressive brain tumor in adults.
Collapse
Affiliation(s)
| | - Ander Matheu
- Cellular Oncology Group, Biodonostia Health Research Institute, San Sebastian, Spain.,CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Madrid, Spain.,IKERBASQUE, Basque Foundation, Bilbao, Spain
| |
Collapse
|
42
|
Acioglu C, Li L, Elkabes S. Contribution of astrocytes to neuropathology of neurodegenerative diseases. Brain Res 2021; 1758:147291. [PMID: 33516810 DOI: 10.1016/j.brainres.2021.147291] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/10/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023]
Abstract
Classically, the loss of vulnerable neuronal populations in neurodegenerative diseases was considered to be the consequence of cell autonomous degeneration of neurons. However, progress in the understanding of glial function, the availability of improved animal models recapitulating the features of the human diseases, and the development of new approaches to derive glia and neurons from induced pluripotent stem cells obtained from patients, provided novel information that altered this view. Current evidence strongly supports the notion that non-cell autonomous mechanisms contribute to the demise of neurons in neurodegenerative disorders, and glia causally participate in the pathogenesis and progression of these diseases. In addition to microglia, astrocytes have emerged as key players in neurodegenerative diseases and will be the focus of the present review. Under the influence of pathological stimuli present in the microenvironment of the diseased CNS, astrocytes undergo morphological, transcriptional, and functional changes and become reactive. Reactive astrocytes are heterogeneous and exhibit neurotoxic (A1) or neuroprotective (A2) phenotypes. In recent years, single-cell or single-nucleus transcriptome analyses unraveled new, disease-specific phenotypes beyond A1/A2. These investigations highlighted the complexity of the astrocytic responses to CNS pathology. The present review will discuss the contribution of astrocytes to neurodegenerative diseases with particular emphasis on Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and frontotemporal dementia. Some of the commonalties and differences in astrocyte-mediated mechanisms that possibly drive the pathogenesis or progression of the diseases will be summarized. The emerging view is that astrocytes are potential new targets for therapeutic interventions. A comprehensive understanding of astrocyte heterogeneity and disease-specific phenotypic complexity could facilitate the design of novel strategies to treat neurodegenerative disorders.
Collapse
Affiliation(s)
- Cigdem Acioglu
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| | - Lun Li
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| | - Stella Elkabes
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| |
Collapse
|
43
|
Bantle CM, Hirst WD, Weihofen A, Shlevkov E. Mitochondrial Dysfunction in Astrocytes: A Role in Parkinson's Disease? Front Cell Dev Biol 2021; 8:608026. [PMID: 33537300 PMCID: PMC7849831 DOI: 10.3389/fcell.2020.608026] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/25/2020] [Indexed: 12/20/2022] Open
Abstract
Mitochondrial dysfunction is a hallmark of Parkinson’s disease (PD). Astrocytes are the most abundant glial cell type in the brain and are thought to play a pivotal role in the progression of PD. Emerging evidence suggests that many astrocytic functions, including glutamate metabolism, Ca2+ signaling, fatty acid metabolism, antioxidant production, and inflammation are dependent on healthy mitochondria. Here, we review how mitochondrial dysfunction impacts astrocytes, highlighting translational gaps and opening new questions for therapeutic development.
Collapse
Affiliation(s)
- Collin M Bantle
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, United States
| | - Warren D Hirst
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, United States
| | - Andreas Weihofen
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, United States
| | - Evgeny Shlevkov
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, United States
| |
Collapse
|
44
|
Ding ZB, Song LJ, Wang Q, Kumar G, Yan YQ, Ma CG. Astrocytes: a double-edged sword in neurodegenerative diseases. Neural Regen Res 2021; 16:1702-1710. [PMID: 33510058 PMCID: PMC8328766 DOI: 10.4103/1673-5374.306064] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Astrocytes play multifaceted and vital roles in maintaining neurophysiological function of the central nervous system by regulating homeostasis, increasing synaptic plasticity, and sustaining neuroprotective effects. Astrocytes become activated as a result of inflammatory responses during the progression of pathological changes associated with neurodegenerative disorders. Reactive astrocytes (neurotoxic A1 and neuroprotective A2) are triggered during disease progression and pathogenesis due to neuroinflammation and ischemia. However, only a limited body of literature describes morphological and functional changes of astrocytes during the progression of neurodegenerative diseases. The present review investigated the detrimental and beneficial roles of astrocytes in neurodegenerative diseases reported in recent studies, as these cells have promising therapeutic potential and offer new approaches for treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Zhi-Bin Ding
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine; Department of Neurology, Affiliated Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Li-Juan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine; Department of Neurology, Affiliated Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Qing Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, Shanxi Province, China
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Hong Kong Special Administrative Region, China
| | - Yu-Qing Yan
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan; Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, Shanxi Province, China
| | - Cun-Gen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan; Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, Shanxi Province, China
| |
Collapse
|
45
|
Du RW, Bu WG. Simvastatin Prevents Neurodegeneration in the MPTP Mouse Model of Parkinson's Disease via Inhibition of A1 Reactive Astrocytes. Neuroimmunomodulation 2021; 28:82-89. [PMID: 33735898 DOI: 10.1159/000513678] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/09/2020] [Indexed: 11/19/2022] Open
Abstract
Emerging evidence indicates that A1 reactive astrocytes play crucial roles in the pathogenesis of Parkinson's disease (PD). Thus, development of agents that could inhibit the formation of A1 reactive astrocytes could be used to treat PD. Simvastatin has been touted as a potential neuroprotective agent for neurologic disorders such as PD, but the specific underlying mechanism remains unclear. The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD and primary astrocytes/neurons were prepared to investigate the effects of simvastatin on PD and its underlying mechanisms in vitro and in vivo. We show that simvastatin protects against the loss of dopamine neurons and behavioral deficits in the MPTP mouse model of PD. We also found that simvastatin suppressed the expression of A1 astrocytic specific markers in vivo and in vitro. In addition, simvastatin alleviated neuron death induced by A1 astrocytes. Our findings reveal that simvastatin is neuroprotective via the prevention of conversion of astrocytes to an A1 neurotoxic phenotype. In light of simvastatin favorable properties, it should be evaluated in the treatment of PD and related neurologic disorders characterized by A1 reactive astrocytes.
Collapse
Affiliation(s)
- Ren-Wei Du
- Department of Neurology, Chaoyang Hospital, Huainan, China,
| | - Wen-Guang Bu
- Department of Neurology, Chaoyang Hospital, Huainan, China
| |
Collapse
|
46
|
De Miranda BR, Rocha EM, Castro SL, Greenamyre JT. Protection from α-Synuclein induced dopaminergic neurodegeneration by overexpression of the mitochondrial import receptor TOM20. NPJ Parkinsons Dis 2020; 6:38. [PMID: 33293540 PMCID: PMC7722884 DOI: 10.1038/s41531-020-00139-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/21/2020] [Indexed: 12/15/2022] Open
Abstract
Dopaminergic neurons of the substantia nigra are selectively vulnerable to mitochondrial dysfunction, which is hypothesized to be an early and fundamental pathogenic mechanism in Parkinson's disease (PD). Mitochondrial function depends on the successful import of nuclear-encoded proteins, many of which are transported through the TOM20-TOM22 outer mitochondrial membrane import receptor machinery. Recent data suggests that post-translational modifications of α-synuclein promote its interaction with TOM20 at the outer mitochondrial membrane and thereby inhibit normal protein import, leading to dysfunction, and death of dopaminergic neurons. As such, preservation of mitochondrial import in the face of α-synuclein accumulation might be a strategy to prevent dopaminergic neurodegeneration, however, this is difficult to assess using current in vivo models of PD. To this end, we established an exogenous co-expression system, utilizing AAV2 vectors to overexpress human α-synuclein and TOM20, individually or together, in the adult Lewis rat substantia nigra to assess whether TOM20 overexpression attenuates α-synuclein-induced dopaminergic neurodegeneration. Twelve weeks after viral injection, we observed that AAV2-TOM20 expression was sufficient to prevent loss of nigral dopaminergic neurons caused by AAV2-αSyn overexpression. The observed TOM20-mediated dopaminergic neuron preservation appeared to be due, in part, to the rescued expression (and presumed import) of nuclear-encoded mitochondrial electron transport chain proteins that were inhibited by α-synuclein overexpression. In addition, TOM20 overexpression rescued the expression of the chaperone protein GRP75/mtHSP70/mortalin, a stress-response protein involved in α-synuclein-induced injury. Collectively, these data indicate that TOM20 expression prevents α-synuclein-induced mitochondrial dysfunction, which is sufficient to rescue dopaminergic neurons in the adult rat brain.
Collapse
Affiliation(s)
- Briana R De Miranda
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Emily M Rocha
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sandra L Castro
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
47
|
Miyazaki I, Asanuma M. Neuron-Astrocyte Interactions in Parkinson's Disease. Cells 2020; 9:cells9122623. [PMID: 33297340 PMCID: PMC7762285 DOI: 10.3390/cells9122623] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/20/2020] [Accepted: 12/05/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease. PD patients exhibit motor symptoms such as akinesia/bradykinesia, tremor, rigidity, and postural instability due to a loss of nigrostriatal dopaminergic neurons. Although the pathogenesis in sporadic PD remains unknown, there is a consensus on the involvement of non-neuronal cells in the progression of PD pathology. Astrocytes are the most numerous glial cells in the central nervous system. Normally, astrocytes protect neurons by releasing neurotrophic factors, producing antioxidants, and disposing of neuronal waste products. However, in pathological situations, astrocytes are known to produce inflammatory cytokines. In addition, various studies have reported that astrocyte dysfunction also leads to neurodegeneration in PD. In this article, we summarize the interaction of astrocytes and dopaminergic neurons, review the pathogenic role of astrocytes in PD, and discuss therapeutic strategies for the prevention of dopaminergic neurodegeneration. This review highlights neuron-astrocyte interaction as a target for the development of disease-modifying drugs for PD in the future.
Collapse
|
48
|
Domingues AV, Pereira IM, Vilaça-Faria H, Salgado AJ, Rodrigues AJ, Teixeira FG. Glial cells in Parkinson´s disease: protective or deleterious? Cell Mol Life Sci 2020; 77:5171-5188. [PMID: 32617639 PMCID: PMC11104819 DOI: 10.1007/s00018-020-03584-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 05/25/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023]
Abstract
Glial cells have been identified more than 100 years ago, and are known to play a key role in the central nervous system (CNS) function. A recent piece of evidence is emerging showing that in addition to the capacity of CNS modulation and homeostasis, glial cells are also being looked like as a promising cell source not only to study CNS pathologies initiation and progression but also to the establishment and development of new therapeutic strategies. Thus, in the present review, we will discuss the current evidence regarding glial cells' contribution to neurodegenerative diseases as Parkinson's disease, providing cellular, molecular, functional, and behavioral data supporting its active role in disease initiation, progression, and treatment. As so, considering their functional relevance, glial cells may be important to the understanding of the underlying mechanisms regarding neuronal-glial networks in neurodegeneration/regeneration processes, which may open new research opportunities for their future use as a target or treatment in human clinical trials.
Collapse
Affiliation(s)
- Ana V Domingues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Laboratory, Braga/Guimarães, Portugal
| | - Inês M Pereira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Laboratory, Braga/Guimarães, Portugal
| | - Helena Vilaça-Faria
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Laboratory, Braga/Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Laboratory, Braga/Guimarães, Portugal
| | - Ana J Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.
- ICVS/3B's Associate Lab, PT Government Associated Laboratory, Braga/Guimarães, Portugal.
| | - Fábio G Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.
- ICVS/3B's Associate Lab, PT Government Associated Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
49
|
Marchetti B. Nrf2/Wnt resilience orchestrates rejuvenation of glia-neuron dialogue in Parkinson's disease. Redox Biol 2020; 36:101664. [PMID: 32863224 PMCID: PMC7395594 DOI: 10.1016/j.redox.2020.101664] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/15/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress and inflammation have long been recognized to contribute to Parkinson's disease (PD), a common movement disorder characterized by the selective loss of midbrain dopaminergic neurons (mDAn) of the substantia nigra pars compacta (SNpc). The causes and mechanisms still remain elusive, but a complex interplay between several genes and a number of interconnected environmental factors, are chiefly involved in mDAn demise, as they intersect the key cellular functions affected in PD, such as the inflammatory response, mitochondrial, lysosomal, proteosomal and autophagic functions. Nuclear factor erythroid 2 -like 2 (NFE2L2/Nrf2), the master regulator of cellular defense against oxidative stress and inflammation, and Wingless (Wnt)/β-catenin signaling cascade, a vital pathway for mDAn neurogenesis and neuroprotection, emerge as critical intertwinned actors in mDAn physiopathology, as a decline of an Nrf2/Wnt/β-catenin prosurvival axis with age underlying PD mutations and a variety of noxious environmental exposures drive PD neurodegeneration. Unexpectedly, astrocytes, the so-called "star-shaped" cells, harbouring an arsenal of "beneficial" and "harmful" molecules represent the turning point in the physiopathological and therapeutical scenario of PD. Fascinatingly, "astrocyte's fil rouge" brings back to Nrf2/Wnt resilience, as boosting the Nrf2/Wnt resilience program rejuvenates astrocytes, in turn (i) mitigating nigrostriatal degeneration of aged mice, (ii) reactivating neural stem progenitor cell proliferation and neuron differentiation in the brain and (iii) promoting a beneficial immunomodulation via bidirectional communication with mDAns. Then, through resilience of Nrf2/Wnt/β-catenin anti-ageing, prosurvival and proregenerative molecular programs, it seems possible to boost the inherent endogenous self-repair mechanisms. Here, the cellular and molecular aspects as well as the therapeutical options for rejuvenating glia-neuron dialogue will be discussed together with major glial-derived mechanisms and therapies that will be fundamental to the identification of novel diagnostic tools and treatments for neurodegenerative diseases (NDs), to fight ageing and nigrostriatal DAergic degeneration and promote functional recovery.
Collapse
Affiliation(s)
- Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Pharmacology Section, Medical School, University of Catania, Via S. Sofia 65, 95125, Catania, Italy; Oasi Research Institute-IRCCS, Neuropharmacology Section, Via Conte Ruggero 73, 94018, Troina, EN, Italy.
| |
Collapse
|
50
|
De Miranda BR, Fazzari M, Rocha EM, Castro S, Greenamyre JT. Sex Differences in Rotenone Sensitivity Reflect the Male-to-Female Ratio in Human Parkinson's Disease Incidence. Toxicol Sci 2020; 170:133-143. [PMID: 30907971 DOI: 10.1093/toxsci/kfz082] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
There is a critical need to include female subjects in disease research; however, in Parkinson's disease, where the male-to-female incidence is about 1.5-to-1, the majority of preclinical research is conducted in male animals. The mitochondrial complex I inhibitor, rotenone, is selectively toxic to dopaminergic neurons, and reproduces several neuropathological features of Parkinson's disease, including α-synuclein pathology. Rotenone has been primarily utilized in male Lewis rats; however, pilot studies in age-matched female Lewis rats revealed that our usual dose (2.8 mg/kg/day intraperitoneal [i.p.]) did not cause dopaminergic neurodegeneration. Therefore, we compared rotenone-treated males (2.8 mg/kg/day, i.p.) to females at increasing doses (2.8 mg/kg/day, 3.2 mg/kg/day, 3.6 mg/kg/day, and 1.6 mg/kg bis in die, i.p.). Female rats receiving 3.2 mg/kg, and 3.6 mg/kg rotenone displayed significant loss of dopaminergic neurons in the substantia nigra as assessed by stereology, which was accompanied by a loss of striatal dopaminergic terminals. Even at these higher doses, however, females showed less inflammation, and less accumulation of α-synuclein and transferrin, possibly as a result of preserved autophagy. Thus, the bias toward increased male incidence of human Parkinson's disease is reflected in the rotenone model. Whether such sex differences will translate into differences in responses to mechanism-driven therapeutic interventions remains to be determined.
Collapse
Affiliation(s)
- Briana R De Miranda
- Pittsburgh Institute for Neurodegenerative Diseases.,Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| | - Marco Fazzari
- Geriatric Research, Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, 15261.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261.,Fondazione Ri.MED, Via Bandiera 11, Palermo 90133, Italy
| | - Emily M Rocha
- Pittsburgh Institute for Neurodegenerative Diseases.,Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| | - Sandra Castro
- Pittsburgh Institute for Neurodegenerative Diseases.,Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases.,Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| |
Collapse
|