1
|
Scorziello A, Sirabella R, Sisalli MJ, Tufano M, Giaccio L, D’Apolito E, Castellano L, Annunziato L. Mitochondrial Dysfunction in Parkinson's Disease: A Contribution to Cognitive Impairment? Int J Mol Sci 2024; 25:11490. [PMID: 39519043 PMCID: PMC11546611 DOI: 10.3390/ijms252111490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/05/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Among the non-motor symptoms associated with Parkinson's disease (PD), cognitive impairment is one of the most common and disabling. It can occur either early or late during the disease, and it is heterogeneous in terms of its clinical manifestations, such as Subjective Cognitive Dysfunction (SCD), Mild Cognitive Impairment (MCI), and Parkinson's Disease Dementia (PDD). The aim of the present review is to delve deeper into the molecular mechanisms underlying cognitive decline in PD. This is extremely important to delineate the guidelines for the differential diagnosis and prognosis of the dysfunction, to identify the molecular and neuronal mechanisms involved, and to plan therapeutic strategies that can halt cognitive impairment progression. Specifically, the present review will discuss the pathogenetic mechanisms involved in the progression of cognitive impairment in PD, with attention to mitochondria and their contribution to synaptic dysfunction and neuronal deterioration in the brain regions responsible for non-motor manifestations of the disease.
Collapse
Affiliation(s)
- Antonella Scorziello
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Rossana Sirabella
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Maria Josè Sisalli
- Department of Translational Medicine, Federico II University of Naples, 80138 Napoli, Italy;
| | - Michele Tufano
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Lucia Giaccio
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Elena D’Apolito
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Lorenzo Castellano
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | | |
Collapse
|
2
|
Lee JS, Park HJ, Kang SO, Lee SH, Lee CK. The effects of light emitting diodes on mitochondrial function and cellular viability of M-1 cell and mouse CD1 brain cortex neurons. PLoS One 2024; 19:e0306656. [PMID: 39213294 PMCID: PMC11364243 DOI: 10.1371/journal.pone.0306656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/20/2024] [Indexed: 09/04/2024] Open
Abstract
The invention of Light Emitting Diode (LED) revolutionized energy-efficient illumination, but concerns persist regarding the potential harm of blue light to our eyes. In this study, we scrutinized the impact of LED light characteristics on eyes using two cell types: M-1 (rich in mitochondria) and CD-1 (neuronal). Variations in color rendering index (CRI) and correlated color temperature (CCT) were investigated, alongside exposure durations ranging from 0 to 24 hours. The findings illuminated the potential benefits of high-quality LED lighting, characterized by a high CRI and low CCT, which emits a greater proportion of red light. This form of lighting was associated with enhanced cell proliferation, elevated ATP levels, and reduced oxidative stress. In contrast, LEDs with low CRI and high CCT exhibited adverse effects, diminishing cell viability and increasing oxidative stress. These results suggest that high-quality LED lighting may have neuroprotective potential as a treatment option, such as for retinal ganglion cells.
Collapse
Affiliation(s)
- Jong Soo Lee
- Department of Ophthalmology, Pusan National University College of Medicine, Busan, Korea
| | - Hyun Jin Park
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Sang Ook Kang
- Department of Advanced Materials Chemistry, Korea University, Sejong, Korea
| | - Sang Hak Lee
- Department of Chemistry, Pusan National University, Busan, Korea
| | - Chang Kyu Lee
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
- Department of Ophthalmology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| |
Collapse
|
3
|
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
|
4
|
Schmidt S, Stautner C, Vu DT, Heinz A, Regensburger M, Karayel O, Trümbach D, Artati A, Kaltenhäuser S, Nassef MZ, Hembach S, Steinert L, Winner B, Jürgen W, Jastroch M, Luecken MD, Theis FJ, Westmeyer GG, Adamski J, Mann M, Hiller K, Giesert F, Vogt Weisenhorn DM, Wurst W. A reversible state of hypometabolism in a human cellular model of sporadic Parkinson's disease. Nat Commun 2023; 14:7674. [PMID: 37996418 PMCID: PMC10667251 DOI: 10.1038/s41467-023-42862-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 10/23/2023] [Indexed: 11/25/2023] Open
Abstract
Sporadic Parkinson's Disease (sPD) is a progressive neurodegenerative disorder caused by multiple genetic and environmental factors. Mitochondrial dysfunction is one contributing factor, but its role at different stages of disease progression is not fully understood. Here, we showed that neural precursor cells and dopaminergic neurons derived from induced pluripotent stem cells (hiPSCs) from sPD patients exhibited a hypometabolism. Further analysis based on transcriptomics, proteomics, and metabolomics identified the citric acid cycle, specifically the α-ketoglutarate dehydrogenase complex (OGDHC), as bottleneck in sPD metabolism. A follow-up study of the patients approximately 10 years after initial biopsy demonstrated a correlation between OGDHC activity in our cellular model and the disease progression. In addition, the alterations in cellular metabolism observed in our cellular model were restored by interfering with the enhanced SHH signal transduction in sPD. Thus, inhibiting overactive SHH signaling may have potential as neuroprotective therapy during early stages of sPD.
Collapse
Affiliation(s)
- Sebastian Schmidt
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany.
- Munich Institute of Biomedical Engineering, Department of Chemistry, Technical University of Munich, Munich, Germany.
| | - Constantin Stautner
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Duc Tung Vu
- Department for Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Alexander Heinz
- Department of Bioinformatics and Biochemistry and Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Martin Regensburger
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ozge Karayel
- Department for Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Anna Artati
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, Neuherberg, Germany
| | - Sabine Kaltenhäuser
- Department of Bioinformatics and Biochemistry and Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Mohamed Zakaria Nassef
- Department of Bioinformatics and Biochemistry and Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Sina Hembach
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Letyfee Steinert
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Winkler Jürgen
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Martin Jastroch
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Malte D Luecken
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Mathematics, Technische Universität München, Garching bei München, Germany
| | - Gil Gregor Westmeyer
- Munich Institute of Biomedical Engineering, Department of Chemistry, Technical University of Munich, Munich, Germany
- Institute for Synthetic Biomedicine, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Matthias Mann
- Department for Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry and Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Florian Giesert
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | | | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany.
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE) site Munich, Munich, Germany.
| |
Collapse
|
5
|
Abrishamdar M, Jalali MS, Yazdanfar N. The role of exosomes in pathogenesis and the therapeutic efficacy of mesenchymal stem cell-derived exosomes against Parkinson's disease. Neurol Sci 2023:10.1007/s10072-023-06706-y. [PMID: 36949298 DOI: 10.1007/s10072-023-06706-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 02/21/2023] [Indexed: 03/24/2023]
Abstract
Parkinson's disease (PD) is a chronic, progressive, neurodegenerative disease. The predominant pathology of PD is the loss of dopaminergic cells in the substantia nigra. Cell transplantation is a strategy with significant potential for treating PD; mesenchymal stem cells (MSCs) are a tremendous therapeutic cell source because they are easily accessible. MSC-derived exosomes with potential protective action in lesioned sites serve as an essential promoter of neuroprotection, and neurodifferentiation, by modulating neural stem cells, neurons, glial cells, and axonal growth in vitro and in vivo environments. The biological properties of MSC-derived exosomes have been proposed as a beneficial tool in different pathological conditions, including PD. Therefore, in this review, we assort the current understanding of MSC-derived exosomes as a new possible therapeutic strategy for PD by providing an overview of the potential role of miRNAs as a component of exosomes in the cellular and molecular basis of PD.
Collapse
Affiliation(s)
- Maryam Abrishamdar
- Department of Physiology, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Sadat Jalali
- Department of Physiology, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Neda Yazdanfar
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Jana M, Dasarathy S, Ghosh S, Pahan K. Upregulation of DJ-1 in Dopaminergic Neurons by a Physically-Modified Saline: Implications for Parkinson's Disease. Int J Mol Sci 2023; 24:ijms24054652. [PMID: 36902085 PMCID: PMC10002578 DOI: 10.3390/ijms24054652] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/21/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder in human and loss-of-functions DJ-1 mutations are associated with a familial form of early onset PD. Functionally, DJ-1 (PARK7), a neuroprotective protein, is known to support mitochondria and protect cells from oxidative stress. Mechanisms and agents by which the level of DJ-1 could be increased in the CNS are poorly described. RNS60 is a bioactive aqueous solution created by exposing normal saline to Taylor-Couette-Poiseuille flow under high oxygen pressure. Recently we have described neuroprotective, immunomodulatory and promyelinogenic properties of RNS60. Here we delineate that RNS60 is also capable of increasing the level of DJ-1 in mouse MN9D neuronal cells and primary dopaminergic neurons, highlighting another new neuroprotective effect of RNS60. While investigating the mechanism we found the presence of cAMP response element (CRE) in DJ-1 gene promoter and stimulation of CREB activation in neuronal cells by RNS60. Accordingly, RNS60 treatment increased the recruitment of CREB to the DJ-1 gene promoter in neuronal cells. Interestingly, RNS60 treatment also induced the enrollment of CREB-binding protein (CBP), but not the other histone acetyl transferase p300, to the promoter of DJ-1 gene. Moreover, knockdown of CREB by siRNA led to the inhibition of RNS60-mediated DJ-1 upregulation, indicating an important role of CREB in DJ-1 upregulation by RNS60. Together, these results indicate that RNS60 upregulates DJ-1 in neuronal cells via CREB-CBP pathway. It may be of benefit for PD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sridevi Dasarathy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | | | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
- Correspondence:
| |
Collapse
|
7
|
Duan G, Li J, Duan Y, Zheng C, Guo Q, Li F, Zheng J, Yu J, Zhang P, Wan M, Long C. Mitochondrial Iron Metabolism: The Crucial Actors in Diseases. Molecules 2022; 28:29. [PMID: 36615225 PMCID: PMC9822237 DOI: 10.3390/molecules28010029] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Iron is a trace element necessary for cell growth, development, and cellular homeostasis, but insufficient or excessive level of iron is toxic. Intracellularly, sufficient amounts of iron are required for mitochondria (the center of iron utilization) to maintain their normal physiologic function. Iron deficiency impairs mitochondrial metabolism and respiratory activity, while mitochondrial iron overload promotes ROS production during mitochondrial electron transport, thus promoting potential disease development. This review provides an overview of iron homeostasis, mitochondrial iron metabolism, and how mitochondrial iron imbalances-induced mitochondrial dysfunction contribute to diseases.
Collapse
Affiliation(s)
- Geyan Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianjun Li
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yehui Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Changbing Zheng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Qiuping Guo
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fengna Li
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Zheng
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiayi Yu
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peiwen Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Mengliao Wan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Cimin Long
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
8
|
Rai P, Kumar Roy J. Endosomal recycling protein Rab11 in Parkin and Pink1 signaling in Drosophila model of Parkinson's disease. Exp Cell Res 2022; 420:113357. [PMID: 36116557 DOI: 10.1016/j.yexcr.2022.113357] [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: 06/30/2022] [Revised: 08/23/2022] [Accepted: 09/11/2022] [Indexed: 11/16/2022]
Abstract
Neurodegenerative diseases are progressive disorders of the nervous system primarily affecting the loss of neuronal cells present in the brain. Although most neurodegenerative cases are sporadic, some familial genes are found to be involved in the neurodegenerative diseases. The extensively studied parkin and pink1 gene products are known to be involved in the removal of damaged mitochondria via autophagy (mitophagy), a quality control process. If the function of any of these genes is somehow disrupted, accumulation of damaged mitochondria occurs in the forms of protein aggregates in the cytoplasm, leading to formation of the Lewy-bodies. Autophagy is an important catabolic process where the endosomal Rab proteins are seen to be involved. Rab11, an endosomal recycling protein, serves as an ATG9A carrier that helps in autophagosome formation and maturation. Earlier studies have reported that loss of Rab11 prevents the fusion of autophagosomes with the late endosomes hampering the autophagy pathway resulting in apoptosis of cells. In this study, we have emphasized on the importance and functional role of Rab11 in the molecular pathway of Parkin/Pink1 in Parkinson's disease.
Collapse
Affiliation(s)
- Pooja Rai
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| | - Jagat Kumar Roy
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
9
|
Tiwari S, Gupta P, Singh A, Chaturvedi S, Wahajuddin M, Mishra A, Singh S. 4-Phenylbutyrate Mitigates the Motor Impairment and Dopaminergic Neuronal Death During Parkinson's Disease Pathology via Targeting VDAC1 Mediated Mitochondrial Function and Astrocytes Activation. Neurochem Res 2022; 47:3385-3401. [PMID: 35922743 DOI: 10.1007/s11064-022-03691-0] [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: 03/02/2022] [Revised: 06/15/2022] [Accepted: 07/12/2022] [Indexed: 10/16/2022]
Abstract
Parkinson's disease (PD) is a progressive motor neurodegenerative disorder significantly associated with protein aggregation related neurodegenerative mechanisms. In view of no disease modifying drugs, the present study was targeted to investigate the therapeutic effects of pharmacological agent 4-phenylbutyric acid (4PBA) in PD pathology. 4PBA is an FDA approved monocarboxylic acid with inhibitory activity towards histone deacetylase and clinically treats urea cycle disorder. First, we observed the significant protective effects of 4PBA on PD specific neuromuscular coordination, level of tyrosine hydroxylase, α-synuclein level and neurotransmitter dopamine in both substantia nigra and striatal regions of the experimental rat model of PD. Further results revealed that treatment with 4PBA drug exhibited significant protection against disease related oxidative stress and augmented nitrite levels. The disease pathology-related depletion in mitochondrial membrane potential and augmented level of calcium as well as mitochondrion membrane located VDAC1 protein level and cytochrome-c translocation were also significantly attenuated with 4PBA administration. Inhibited neuronal apoptosis and restored neuronal morphology were also observed with 4PBA treatment as measured by level of pro-apoptotic proteins t-Bid, Bax and cleaved caspase-3 along with cresyl violet staining in both substantia nigra and striatal regions. Lastly, PD-linked astrocyte activation was significantly inhibited with 4PBA treatment. Altogether, our findings suggest that 4PBA exerts broad-spectrum neuroprotective effects in PD animal model.
Collapse
Affiliation(s)
- Shubhangini Tiwari
- Division of Neuroscience and Ageing Biology, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Parul Gupta
- Division of Neuroscience and Ageing Biology, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Abhishek Singh
- Division of Neuroscience and Ageing Biology, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Swati Chaturvedi
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - M Wahajuddin
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology, Jodhpur, Rajasthan, 342011, India
| | - Sarika Singh
- Division of Neuroscience and Ageing Biology, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India. .,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
10
|
Robustness of the Krebs Cycle under Physiological Conditions and in Cancer: New Clues for Evaluating Metabolism-Modifying Drug Therapies. Biomedicines 2022; 10:biomedicines10051199. [PMID: 35625935 PMCID: PMC9138339 DOI: 10.3390/biomedicines10051199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/10/2022] Open
Abstract
The Krebs cycle in cells that contain mitochondria is necessary for both energy production and anabolic processes. In given cell/condition, the Krebs cycle is dynamic but remains at a steady state. In this article, we first aimed at comparing the properties of a closed cycle versus the same metabolism in a linear array. The main finding is that, unlike a linear metabolism, the closed cycle can reach a steady state (SS) regardless of the nature and magnitude of the disturbance. When the cycle is modeled with input and output reactions, the “open” cycle is robust and reaches a steady state but with exceptions that lead to sustained accumulation of intermediate metabolites, i.e., conditions at which no SS can be achieved. The modeling of the cycle in cancer, trying to obtain marked reductions in flux, shows that these reductions are limited and therefore the Warburg effect is moderate at most. In general, our results of modeling the cycle in different conditions and looking for the achievement, or not, of SS, suggest that the cycle may have a regulation, not yet discovered, to go from an open cycle to a closed one. Said regulation could allow for reaching the steady state, thus avoiding the unwanted effects derived from the aberrant accumulation of metabolites in the mitochondria. The information in this paper might be useful to evaluate metabolism-modifying medicines.
Collapse
|
11
|
Xie L, Xiao Y, Meng F, Li Y, Shi Z, Qian K. Functions and Mechanisms of Lysine Glutarylation in Eukaryotes. Front Cell Dev Biol 2021; 9:667684. [PMID: 34249920 PMCID: PMC8264553 DOI: 10.3389/fcell.2021.667684] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/01/2021] [Indexed: 01/22/2023] Open
Abstract
Lysine glutarylation (Kglu) is a newly discovered post-translational modification (PTM), which is considered to be reversible, dynamic, and conserved in prokaryotes and eukaryotes. Recent developments in the identification of Kglu by mass spectrometry have shown that Kglu is mainly involved in the regulation of metabolism, oxidative damage, chromatin dynamics and is associated with various diseases. In this review, we firstly summarize the development history of glutarylation, the biochemical processes of glutarylation and deglutarylation. Then we focus on the pathophysiological functions such as glutaric acidemia 1, asthenospermia, etc. Finally, the current computational tools for predicting glutarylation sites are discussed. These emerging findings point to new functions for lysine glutarylation and related enzymes, and also highlight the mechanisms by which glutarylation regulates diverse cellular processes.
Collapse
Affiliation(s)
- Longxiang Xie
- Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Huaihe Hospital, Henan University, Kaifeng, China
| | - Yafei Xiao
- Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Huaihe Hospital, Henan University, Kaifeng, China
| | - Fucheng Meng
- Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Huaihe Hospital, Henan University, Kaifeng, China
| | - Yongqiang Li
- Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Huaihe Hospital, Henan University, Kaifeng, China
| | - Zhenyu Shi
- Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Huaihe Hospital, Henan University, Kaifeng, China
| | - Keli Qian
- Infection Control Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
12
|
Artero-Castro A, Rodriguez-Jimenez FJ, Jendelova P, VanderWall KB, Meyer JS, Erceg S. Glaucoma as a Neurodegenerative Disease Caused by Intrinsic Vulnerability Factors. Prog Neurobiol 2020; 193:101817. [PMID: 32360241 DOI: 10.1016/j.pneurobio.2020.101817] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/30/2020] [Accepted: 04/23/2020] [Indexed: 01/08/2023]
Abstract
Glaucoma, one of the most common causes of blindness in developing countries today, involves a progressive loss of neural cells in the optic nerve that leads to progressive, irreversible vision loss. Increased intraocular pressure (IOP) presents as a major risk factor for glaucoma, although there exist cases of glaucoma patients with normal IOP that exhibit damage to retinal ganglion cells (RGCs) and the optic nerve. However, treatment approaches have maintained their focus on modifying IOP due to a lack of other modifiable risks factors. Traditional concepts in glaucoma involve the neuronal environment and external effects as a source of causative factors; however, studies have yet to investigate whether the molecular profile of RGCs in glaucoma patients makes them more vulnerable and/or susceptible to external damage. Our hypothesis states that molecular changes at the whole cell, gene expression, and electrophysiological level of the neurons can contribute to their degeneration. Herein, we briefly describe different types of glaucoma and any similarities to different molecular and cellular features of neurodegeneration. To test our hypothesis, we describe human induced pluripotent stem cells (hiPSCs) as a reliable cellular tool to model neurodegenerative aspects of glaucoma to reveal the multiple pathological molecular mechanisms underlying disease development.
Collapse
Affiliation(s)
- Ana Artero-Castro
- Stem Cells Therapies in Neurodegenerative Diseases Lab, Research Center "Principe Felipe", Valencia, Spain.
| | | | - Pavla Jendelova
- Institute of Experimental Medicine, Czech Academy of Sciences, Department of Neuroregeneration, Prague, Czech Republic.
| | - Kirstin B VanderWall
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA.
| | - Jason S Meyer
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Slaven Erceg
- Stem Cells Therapies in Neurodegenerative Diseases Lab, Research Center "Principe Felipe", Valencia, Spain; National Stem Cell Bank-Valencia Node, Platform for Proteomics, Genotyping and Cell Lines, PRB3,ISCIII, Research Center "Principe Felipe", Valencia, Spain; Institute of Experimental Medicine, Czech Academy of Sciences, Department of Neuroregeneration, Prague, Czech Republic.
| |
Collapse
|
13
|
Franco R, Rivas-Santisteban R, Reyes-Resina I, Navarro G, Martínez-Pinilla E. Microbiota and Other Preventive Strategies and Non-genetic Risk Factors in Parkinson's Disease. Front Aging Neurosci 2020; 12:12. [PMID: 32226375 PMCID: PMC7080700 DOI: 10.3389/fnagi.2020.00012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/15/2020] [Indexed: 12/15/2022] Open
Abstract
The exact cause of Parkinson’s disease (PD), the second most prevalent neurodegenerative disease in modern societies, is still unknown. Many scientists point out that PD is caused by a complex interaction between different factors. Although the main risk factor is age, there are other influences, genetic and environmental, that individually or in combination may trigger neurodegenerative changes leading to PD. Nowadays, research remains focused on better understanding which environmental factors are related to the risk of developing PD and why. In line with the knowledge on evidence on exposures that prevent/delay PD onset or that impact on disease progression, the aims of this review were: (i) to comment on the non-genetic risk factors that mainly affect idiopathic PD; and (ii) to comment on seemingly reliable preventive interventions. We discuss both environmental factors that may affect the central nervous system (CNS) or the intestinal tract, and the likely mechanisms underlying noxious or protective actions. Knowledge on risk, protective factors, and mechanisms may help to envisage why nigral dopaminergic neurons are so vulnerable in PD and, eventually, to design new strategies for PD prevention and/or anti-PD therapy. This article reviews the variety of the known and suspected environmental factors, such as lifestyle, gut microbiota or pesticide exposition, and distinguishes between those that are harmful or beneficial for the PD acquisition or progression. In fact, the review covers one of the most novel players in the whole picture, and we address the role of microbiota on keeping a healthy CNS and/or on preventing the “side-effects” related to aging.
Collapse
Affiliation(s)
- Rafael Franco
- Chemistry School, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael Rivas-Santisteban
- Chemistry School, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Gemma Navarro
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain.,Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| | - Eva Martínez-Pinilla
- Departamento de Morfología y Biología Celular, Facultad de Medicina, Universidad de Oviedo, Oviedo, Spain.,Instituto de Neurociencias del Principado de Asturias (INEUROPA), Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
14
|
Imai Y. PINK1-Parkin signaling in Parkinson's disease: Lessons from Drosophila. Neurosci Res 2020; 159:40-46. [PMID: 32035987 DOI: 10.1016/j.neures.2020.01.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 01/30/2020] [Indexed: 12/30/2022]
Abstract
The mitochondrial protein kinase PINK1 activates Parkin ubiquitin ligase by phosphorylating Parkin and ubiquitin, which are required for mitochondrial maintenance in dopaminergic (DA) neurons whose degeneration leads to the development of Parkinson's disease (PD). Loss of PINK1 and Parkin leads to mitochondrial degeneration and abnormal wing posture in Drosophila. Modifier screening using the Drosophila wing phenotype showed that the inactivation of Miro, a mitochondrial adaptor protein, suppresses the phenotype caused by mitochondrial degeneration. When activated by PINK1, Parkin suppresses mitochondrial transport by reducing Miro levels in Drosophila DA neurons. In human DA neurons, PINK1-Parkin signaling also regulates axonal mitochondrial re-distribution in response to reduced mitochondrial membrane potential, which is impaired in the DA neurons of patients with PINK1 and Parkin mutations. Phospho-ubiquitin signals amplified by PINK1 and Parkin are stronger in DA neurons than other neurons, suggesting that PINK1-Parkin signaling is particularly important for DA neuron activity. Moreover, the recently identified PD-associated protein CHCHD2 may ensure proper electron transfer during mitochondrial respiration. The genetic interaction between PINK1/Parkin and CHCHD2 in Drosophila indicates that they are not directly associated and CHCHD2-linked PD exhibits a very different pathology to PINK1/Parkin PD. I suggest a complex pathogenesis for mitochondrial dysregulation in PD.
Collapse
Affiliation(s)
- Yuzuru Imai
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.
| |
Collapse
|
15
|
Ikeda A, Nishioka K, Meng H, Takanashi M, Hasegawa I, Inoshita T, Shiba-Fukushima K, Li Y, Yoshino H, Mori A, Okuzumi A, Yamaguchi A, Nonaka R, Izawa N, Ishikawa KI, Saiki H, Morita M, Hasegawa M, Hasegawa K, Elahi M, Funayama M, Okano H, Akamatsu W, Imai Y, Hattori N. Mutations in CHCHD2 cause α-synuclein aggregation. Hum Mol Genet 2019; 28:3895-3911. [DOI: 10.1093/hmg/ddz241] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 02/02/2023] Open
Abstract
Abstract
Mutations in CHCHD2 are linked to a familial, autosomal dominant form of Parkinson’s disease (PD). The gene product may regulate mitochondrial respiratory function. However, whether mitochondrial dysfunction induced by CHCHD2 mutations further yields α-synuclein pathology is unclear. Here, we provide compelling genetic evidence that mitochondrial dysfunction induced by PD-linked CHCHD2 T61I mutation promotes α-synuclein aggregation using brain autopsy, induced pluripotent stem cells (iPSCs) and Drosophila genetics. An autopsy of an individual with CHCHD2 T61I revealed widespread Lewy pathology with both amyloid plaques and neurofibrillary tangles that appeared in the brain stem, limbic regions and neocortex. A prominent accumulation of sarkosyl-insoluble α-synuclein aggregates, the extent of which was comparable to that of a case with α-synuclein (SNCA) duplication, was observed in CHCHD2 T61I brain tissue. The prion-like activity and morphology of α-synuclein fibrils from the CHCHD2 T61I brain tissue were similar to those of fibrils from SNCA duplication and sporadic PD brain tissues. α-Synuclein insolubilization was reproduced in dopaminergic neuron cultures from CHCHD2 T61I iPSCs and Drosophila lacking the CHCHD2 ortholog or expressing the human CHCHD2 T61I. Moreover, the combination of ectopic α-synuclein expression and CHCHD2 null or T61I enhanced the toxicity in Drosophila dopaminergic neurons, altering the proteolysis pathways. Furthermore, CHCHD2 T61I lost its mitochondrial localization by α-synuclein in Drosophila. The mislocalization of CHCHD2 T61I was also observed in the patient brain. Our study suggests that CHCHD2 is a significant mitochondrial factor that determines α-synuclein stability in the etiology of PD.
Collapse
Affiliation(s)
- Aya Ikeda
- Department of Neurology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Kenya Nishioka
- Department of Neurology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Hongrui Meng
- Department of Neurodegenerative and Demented Disorders, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Masashi Takanashi
- Department of Neurology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Iwao Hasegawa
- University Center of Legal Medicine, Kanagawa Dental University, Kanagawa 238-8580, Japan
| | - Tsuyoshi Inoshita
- Department of Treatment and Research in Multiple Sclerosis and Neuro-intractable Disease, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Kahori Shiba-Fukushima
- Department of Treatment and Research in Multiple Sclerosis and Neuro-intractable Disease, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Yuanzhe Li
- Department of Neurology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Hiroyo Yoshino
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Akio Mori
- Department of Neurology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Ayami Okuzumi
- Department of Neurology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Akihiro Yamaguchi
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Risa Nonaka
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
- Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Nana Izawa
- Department of Neurology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Kei-ichi Ishikawa
- Department of Neurology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Hidemoto Saiki
- Department of Neurology, Tazuke Kofukai Medical Research Institute and Kitano Hospital, Osaka 530-8480, Japan
| | - Masayo Morita
- Department of Neurology, Jikei University Katsushika Medical Center, Tokyo 125-8506, Japan
| | - Masato Hasegawa
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Kazuko Hasegawa
- Department of Neurology, NHO Sagamihara National Hospital, Kanagawa 252-0392, Japan
| | - Montasir Elahi
- Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Manabu Funayama
- Department of Neurology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Wado Akamatsu
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Yuzuru Imai
- Department of Neurology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Department of Research for Parkinson’s Disease, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Department of Neurodegenerative and Demented Disorders, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
- Department of Treatment and Research in Multiple Sclerosis and Neuro-intractable Disease, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| |
Collapse
|
16
|
Pariary R, Bhattacharyya D, Bhunia A. Mitochondrial-membrane association of α-synuclein: Pros and cons in consequence of Parkinson's disease pathophysiology. GENE REPORTS 2019. [DOI: 10.1016/j.genrep.2019.100423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
17
|
Yamashita S, Tanaka M, Nodono H, Hamada A, Hamada T, Hasegawa M, Nishi Y, Moss J, Miwa M. Human alcohol dehydrogenase 1 is an acceptor protein for polyADP-ribosylation. Biochem Pharmacol 2019; 167:27-32. [PMID: 30936015 PMCID: PMC9872671 DOI: 10.1016/j.bcp.2019.03.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 03/19/2019] [Indexed: 01/26/2023]
Abstract
Alcohol dehydrogenase (ADH) is important for preventing alcohol toxicity and developmental disorders, and may be involved in other diseases including neurodegenerative diseases. We found that the major acceptor protein of polyADP-ribosylation in a model organism of neurodegeneration using a Drosophila melanogaster mutant lacking poly(ADP-ribose) glycohydrolase, was ADH. Thus we postulated that human ADH activity might be regulated by polyADP-ribosylation, a post-translational modification. The radioactivity of [32P]NAD+ was incorporated into human ADH1 by human poly(ADP-ribose) polymerase 1 in vitro, but was not incorporated when heat-inactivated PARP1 or a PARP inhibitor, 3-aminobenzamide, was used. The incorporated radioactivity was not released from ADH1 protein in the presence of excess amount of ADP-ribose or poly(ADP-ribose) as competitors. However, it was released by incubation with 1 M neutral NH2OH or 0.1 N NaOH, but was not with 0.1 N HCl, suggesting the bond between ADH1 and poly(ADP-ribose) is an ester linkage. When HepG2 cells, a human hepatoma cell line, were cultured in the presence of another PARP inhibitor, olaparib, ADH activity of the cell was significantly increased. These results suggest that polyADP-ribosylation could regulate ADH activity in vivo and might be involved in neurodegeneration.
Collapse
Affiliation(s)
- Sachiko Yamashita
- Faculty of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan,Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1590, USA
| | - Masakazu Tanaka
- Department of Microbiology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan,Division of Molecular Pathology, Center for Chronic Viral Diseases, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Hiroto Nodono
- Faculty of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan
| | - Akiko Hamada
- Faculty of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan
| | - Takashi Hamada
- Faculty of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan
| | - Makoto Hasegawa
- Faculty of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan
| | - Yoshisuke Nishi
- Faculty of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan
| | - Joel Moss
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1590, USA
| | - Masanao Miwa
- Faculty of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan,Corresponding author. (M. Miwa)
| |
Collapse
|
18
|
Mondal S, Rangasamy SB, Roy A, Dasarathy S, Kordower JH, Pahan K. Low-Dose Maraviroc, an Antiretroviral Drug, Attenuates the Infiltration of T Cells into the Central Nervous System and Protects the Nigrostriatum in Hemiparkinsonian Monkeys. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 202:3412-3422. [PMID: 31043478 PMCID: PMC6824976 DOI: 10.4049/jimmunol.1800587] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 04/05/2019] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder in humans. Despite intense investigation, no effective therapy is available to stop the progression of this disease. It is becoming clear that both innate and adaptive immune responses are active in PD. Accordingly, we have reported a marked increase in RANTES and eotaxin, chemokines that are involved in T cell trafficking, in vivo in the substantia nigra (SN) and the serum of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-intoxicated hemiparkinsonian monkeys. Because RANTES and eotaxin share a common receptor, CCR5, we examined the efficacy of maraviroc, an inhibitor of CCR5 and a Food and Drug Administration-approved drug against HIV infection, in hemiparkinsonian rhesus monkeys. First, we found glial limitans injury, loss of GFAP immunostaining, and infiltration of T cells across the endothelial monolayer in SN of hemiparkinsonian monkeys. However, oral administration of a low dose of maraviroc protected glia limitans partially, maintained the integrity of endothelial monolayer, reduced the infiltration of T cells, attenuated neuroinflammation, and decreased α-synucleinopathy in the SN. Accordingly, maraviroc treatment also protected both the nigrostriatal axis and neurotransmitters and improved motor functions in hemiparkinsonian monkeys. These results suggest that low-dose maraviroc and other CCR5 antagonists may be helpful for PD patients.
Collapse
Affiliation(s)
- Susanta Mondal
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| | - Suresh B Rangasamy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| | - Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| | - Sridevi Dasarathy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| | - Jeffrey H Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| |
Collapse
|
19
|
Mitochondria in Neuroprotection by Phytochemicals: Bioactive Polyphenols Modulate Mitochondrial Apoptosis System, Function and Structure. Int J Mol Sci 2019; 20:ijms20102451. [PMID: 31108962 PMCID: PMC6566187 DOI: 10.3390/ijms20102451] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 05/11/2019] [Accepted: 05/15/2019] [Indexed: 12/15/2022] Open
Abstract
In aging and neurodegenerative diseases, loss of distinct type of neurons characterizes disease-specific pathological and clinical features, and mitochondria play a pivotal role in neuronal survival and death. Mitochondria are now considered as the organelle to modulate cellular signal pathways and functions, not only to produce energy and reactive oxygen species. Oxidative stress, deficit of neurotrophic factors, and multiple other factors impair mitochondrial function and induce cell death. Multi-functional plant polyphenols, major groups of phytochemicals, are proposed as one of most promising mitochondria-targeting medicine to preserve the activity and structure of mitochondria and neurons. Polyphenols can scavenge reactive oxygen and nitrogen species and activate redox-responsible transcription factors to regulate expression of genes, coding antioxidants, anti-apoptotic Bcl-2 protein family, and pro-survival neurotrophic factors. In mitochondria, polyphenols can directly regulate the mitochondrial apoptosis system either in preventing or promoting way. Polyphenols also modulate mitochondrial biogenesis, dynamics (fission and fusion), and autophagic degradation to keep the quality and number. This review presents the role of polyphenols in regulation of mitochondrial redox state, death signal system, and homeostasis. The dualistic redox properties of polyphenols are associated with controversial regulation of mitochondrial apoptosis system involved in the neuroprotective and anti-carcinogenic functions. Mitochondria-targeted phytochemical derivatives were synthesized based on the phenolic structure to develop a novel series of neuroprotective and anticancer compounds, which promote the bioavailability and effectiveness. Phytochemicals have shown the multiple beneficial effects in mitochondria, but further investigation is required for the clinical application.
Collapse
|
20
|
Abstract
Mitochondria are an iconic distinguishing feature of eukaryotic cells. Mitochondria encompass an active organellar network that fuses, divides, and directs a myriad of vital biological functions, including energy metabolism, cell death regulation, and innate immune signaling in different tissues. Another crucial and often underappreciated function of these dynamic organelles is their central role in the metabolism of the most abundant and biologically versatile transition metals in mammalian cells, iron. In recent years, cellular and animal models of mitochondrial iron dysfunction have provided vital information in identifying new proteins that have elucidated the pathways involved in mitochondrial homeostasis and iron metabolism. Specific signatures of mitochondrial iron dysregulation that are associated with disease pathogenesis and/or progression are becoming increasingly important. Understanding the molecular mechanisms regulating mitochondrial iron pathways will help better define the role of this important metal in mitochondrial function and in human health and disease.
Collapse
Affiliation(s)
- Diane M Ward
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA
| | - Suzanne M Cloonan
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY 10065, USA;
| |
Collapse
|
21
|
Supandi F, van Beek JHGM. Computational prediction of changes in brain metabolic fluxes during Parkinson's disease from mRNA expression. PLoS One 2018; 13:e0203687. [PMID: 30208076 PMCID: PMC6135490 DOI: 10.1371/journal.pone.0203687] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 08/24/2018] [Indexed: 12/12/2022] Open
Abstract
Background Parkinson’s disease is a widespread neurodegenerative disorder which affects brain metabolism. Although changes in gene expression during disease are often measured, it is difficult to predict metabolic fluxes from gene expression data. Here we explore the hypothesis that changes in gene expression for enzymes tend to parallel flux changes in biochemical reaction pathways in the brain metabolic network. This hypothesis is the basis of a computational method to predict metabolic flux changes from post-mortem gene expression measurements in Parkinson’s disease (PD) brain. Results We use a network model of central metabolism and optimize the correspondence between relative changes in fluxes and in gene expression. To this end we apply the Least-squares with Equalities and Inequalities algorithm integrated with Flux Balance Analysis (Lsei-FBA). We predict for PD (1) decreases in glycolytic rate and oxygen consumption and an increase in lactate production in brain cortex that correspond with measurements (2) relative flux decreases in ATP synthesis, in the malate-aspartate shuttle and midway in the TCA cycle that are substantially larger than relative changes in glucose uptake in the substantia nigra, dopaminergic neurons and most other brain regions (3) shifts in redox shuttles between cytosol and mitochondria (4) in contrast to Alzheimer’s disease: little activation of the gamma-aminobutyric acid shunt pathway in compensation for decreased alpha-ketoglutarate dehydrogenase activity (5) in the globus pallidus internus, metabolic fluxes are increased, reflecting increased functional activity. Conclusion Our method predicts metabolic changes from gene expression data that correspond in direction and order of magnitude with presently available experimental observations during Parkinson’s disease, indicating that the hypothesis may be useful for some biochemical pathways. Lsei-FBA generates predictions of flux distributions in neurons and small brain regions for which accurate metabolic flux measurements are not yet possible.
Collapse
Affiliation(s)
- Farahaniza Supandi
- Department of Clinical Genetics, VU University Medical Centre, Amsterdam, the Netherlands
- Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
- * E-mail:
| | - Johannes H. G. M. van Beek
- Department of Clinical Genetics, VU University Medical Centre, Amsterdam, the Netherlands
- Department of Experimental Vascular Medicine, Academic Medical Center, AZ Amsterdam, the Netherlands
| |
Collapse
|
22
|
Increase in Mitochondrial Biogenesis in Neuronal Cells by RNS60, a Physically-Modified Saline, via Phosphatidylinositol 3-Kinase-Mediated Upregulation of PGC1α. J Neuroimmune Pharmacol 2017; 13:143-162. [PMID: 29188424 PMCID: PMC5928179 DOI: 10.1007/s11481-017-9771-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 11/10/2017] [Indexed: 11/25/2022]
Abstract
This study highlights a novel approach to upregulate mitochondrial biogenesis in neuronal cells. RNS60 is a 0.9% saline solution containing oxygenated nanobubbles that is generated by subjecting normal saline to Taylor-Couette-Poiseuille (TCP) flow under elevated oxygen pressure. RNS60, but not NS (normal saline), PNS60 (saline containing a comparable level of oxygen without the TCP modification), or RNS10.3 (TCP-modified normal saline without excess oxygen), increased the expression of Nrf1, Tfam, Mcu, and Tom20 (genes associated with mitochondrial biogenesis) and upregulated mitochondrial biogenesis in MN9D dopaminergic neuronal cells. Similarly RNS60 also increased mitochondrial biogenesis in primary dopaminergic neurons and in the nigra of MPTP-intoxicated mice. However, RNS60 had no effect on lysosomal biogenesis. Interestingly, we found that RNS60 upregulated PGC1α and siRNA knockdown of PGC1α abrogated the ability of RNS60 to increase mitochondrial biogenesis. Furthermore, we delineated that RNS60 increased the transcription of Pgc1a via type IA phosphatidylinositol (PI) 3-kinase-mediated activation of cAMP-response element-binding protein (CREB). Accordingly, knockdown of the PI3K – CREB pathway suppressed RNS60-mediated mitochondrial biogenesis. These results describe a novel property of RNS60 of enhancing mitochondrial biogenesis via PI 3-kinase-CREB-mediated up-regulation of PGC1α, which may be of therapeutic benefit in different neurodegenerative disorders.
Collapse
|
23
|
Ducray AD, Felser A, Zielinski J, Bittner A, Bürgi JV, Nuoffer JM, Frenz M, Mevissen M. Effects of silica nanoparticle exposure on mitochondrial function during neuronal differentiation. J Nanobiotechnology 2017; 15:49. [PMID: 28676089 PMCID: PMC5496409 DOI: 10.1186/s12951-017-0284-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 06/17/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Nanomedicine offers a promising tool for therapies of brain diseases, but potential effects on neuronal health and neuronal differentiation need to be investigated to assess potential risks. The aim of this study was to investigate effects of silica-indocyanine green/poly (ε-caprolactone) nanoparticles (PCL-NPs) engineered for laser tissue soldering in the brain before and during differentiation of SH-SY5Y cells. Considering adaptations in mitochondrial homeostasis during neuronal differentiation, metabolic effects of PCL-NP exposure before and during neuronal differentiation were studied. In addition, kinases of the PI3 kinase (PI3-K/Akt) and the MAP kinase (MAP-K/ERK) pathways related to neuronal differentiation and mitochondrial function were investigated. RESULTS Differentiation resulted in a decrease in the cellular respiration rate and the extracellular acidification rate (ECAR). PCL-NP exposure impaired mitochondrial function depending on the time of exposure. The cellular respiration rate was significantly reduced compared to differentiated controls when PCL-NPs were given before differentiation. The shift in ECAR was less pronounced in PCL-NP exposure during differentiation. Differentiation and PCL-NP exposure had no effect on expression levels and the enzymatic activity of respiratory chain complexes. The activity of the glycolytic enzyme phosphofructokinase was significantly reduced after differentiation with the effect being more pronounced after PCL-NP exposure before differentiation. The increase in mitochondrial membrane potential observed after differentiation was not found in SH-SY5Y cells exposed to PCL-NPs before differentiation. The cellular adenosine triphosphate (ATP) production significantly dropped during differentiation, and this effect was independent of the PCL-NP exposure. Differentiation and nanoparticle exposure had no effect on superoxide levels at the endpoint of the experiments. A slight decrease in the expression of the neuronal differentiation markers was found after PCL-NP exposure, but no morphological variation was observed. CONCLUSIONS PCL-NP exposure affects mitochondrial function depending on the time of exposure before and during neuronal differentiation. PCL-NP exposure during differentiation was associated with impaired mitochondrial function, which may affect differentiation. Considering the importance of adaptations in cellular respiration for neuronal differentiation and function, further studies are needed to unravel the underlying mechanisms and consequences to assess the possible risks including neurodegeneration.
Collapse
Affiliation(s)
- Angélique D Ducray
- Division of Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 124, 3012, Bern, Switzerland
| | - Andrea Felser
- Institute of Clinical Chemistry, University Hospital Bern, 3010, Bern, Switzerland
| | - Jana Zielinski
- Division of Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 124, 3012, Bern, Switzerland
| | - Aniela Bittner
- Division of Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 124, 3012, Bern, Switzerland
| | - Julia V Bürgi
- Division of Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 124, 3012, Bern, Switzerland
| | - Jean-Marc Nuoffer
- Institute of Clinical Chemistry, University Hospital Bern, 3010, Bern, Switzerland
| | - Martin Frenz
- Institute of Applied Physics, University of Bern, Sidlerstrasse 5, 3012, Bern, Switzerland
| | - Meike Mevissen
- Division of Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 124, 3012, Bern, Switzerland.
| |
Collapse
|
24
|
Andres RH, Ducray AD, Pérez-Bouza A, Schlattner U, Huber AW, Krebs SH, Seiler RW, Wallimann T, Widmer HR. Creatine Supplementation Improves Dopaminergic Cell Survival and Protects against MPP+ Toxicity in an Organotypic Tissue Culture System. Cell Transplant 2017; 14:537-50. [PMID: 16355565 DOI: 10.3727/000000005783982756] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cell replacement therapy using mesencephalic precursor cells is an experimental approach for the treatment of Parkinson's disease (PD). A significant problem associated with this procedure is the poor survival of grafted neurons. Impaired energy metabolism is considered to contribute to neuronal cell death after transplantation. Creatine is a substrate for mitochondrial and cytosolic creatine kinases (CK) and buffers cellular ATP resources. Furthermore, elevated cellular creatine levels facilitate metabolic channeling and show anti-apoptotic properties. Exogenous creatine supplementation therefore might offer a tool for improvement of dopaminergic neuron survival. The present study aimed at investigating the effects of creatine on cell survival of rat embryonic day 14 (E14) ventral mesencephalic neurons grown as organotypic free-floating roller tube (FFRT) cultures. We found that the brain-specific isoform of CK (BB-CK) and the ubiquitous mitochondrial isoform (uMt-CK) are expressed at high levels in FFRT cultures and colocalize with tyrosine hydroxylase immunoreactive (TH-ir) cells. Exposure of these cultures to creatine induced an increase in the content of the BB-CK isotype. Creatine (5 mM) administration starting at day in vitro (DIV) 7 resulted in a significant increase (+35%) in TH-ir cell density at DIV21. In addition, we observed that creatine treatment provided neuroprotection against 1-methyl-4-phenyl pyridinium ion (MPP+)-induced TH-ir cell loss in the FFRT culture system, resulting in a significantly higher density (+19%) of TH-ir neurons in creatine-treated cultures compared to corresponding controls. The decrease of TH-ir neurons in the MPP+-treated group corresponded with an increase in immunoreactivity for active caspase-3, an effect that was not seen in the group receiving creatine supplementation. In conclusion, our data imply that creatine administration is beneficial for the survival of TH-ir neurons encountering harmful conditions.
Collapse
Affiliation(s)
- Robert H Andres
- Department of Neurosurgery, University Hospital, Berne, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Barodia SK, Creed RB, Goldberg MS. Parkin and PINK1 functions in oxidative stress and neurodegeneration. Brain Res Bull 2016; 133:51-59. [PMID: 28017782 DOI: 10.1016/j.brainresbull.2016.12.004] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 12/07/2016] [Accepted: 12/15/2016] [Indexed: 12/12/2022]
Abstract
Loss-of-function mutations in the genes encoding Parkin and PINK1 are causally linked to autosomal recessive Parkinson's disease (PD). Parkin, an E3 ubiquitin ligase, and PINK1, a mitochondrial-targeted kinase, function together in a common pathway to remove dysfunctional mitochondria by autophagy. Presumably, deficiency for Parkin or PINK1 impairs mitochondrial autophagy and thereby increases oxidative stress due to the accumulation of dysfunctional mitochondria that release reactive oxygen species. Parkin and PINK1 likely have additional functions that may be relevant to the mechanisms by which mutations in these genes cause neurodegeneration, such as regulating inflammation, apoptosis, or dendritic morphogenesis. Here we briefly review what is known about functions of Parkin and PINK1 related to oxidative stress and neurodegeneration.
Collapse
Affiliation(s)
- Sandeep K Barodia
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Rose B Creed
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Matthew S Goldberg
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States; Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
26
|
Parkinson's Disease: The Mitochondria-Iron Link. PARKINSONS DISEASE 2016; 2016:7049108. [PMID: 27293957 PMCID: PMC4886095 DOI: 10.1155/2016/7049108] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 12/14/2022]
Abstract
Mitochondrial dysfunction, iron accumulation, and oxidative damage are conditions often found in damaged brain areas of Parkinson's disease. We propose that a causal link exists between these three events. Mitochondrial dysfunction results not only in increased reactive oxygen species production but also in decreased iron-sulfur cluster synthesis and unorthodox activation of Iron Regulatory Protein 1 (IRP1), a key regulator of cell iron homeostasis. In turn, IRP1 activation results in iron accumulation and hydroxyl radical-mediated damage. These three occurrences-mitochondrial dysfunction, iron accumulation, and oxidative damage-generate a positive feedback loop of increased iron accumulation and oxidative stress. Here, we review the evidence that points to a link between mitochondrial dysfunction and iron accumulation as early events in the development of sporadic and genetic cases of Parkinson's disease. Finally, an attempt is done to contextualize the possible relationship between mitochondria dysfunction and iron dyshomeostasis. Based on published evidence, we propose that iron chelation-by decreasing iron-associated oxidative damage and by inducing cell survival and cell-rescue pathways-is a viable therapy for retarding this cycle.
Collapse
|
27
|
Abstract
BACKGROUND The primary pathophysiological feature of glaucoma is a progressive optic neuropathy with characteristic morphological changes of the optic disc and risk factors of age and intraocular pressure. Recently, involvement of other areas of the central nervous system (CNS) beyond the optic nerve has been demonstrated. This article addresses the proposition that glaucoma shares mechanistic and pathophysiologic features with neurodegenerations in the CNS. METHODS The literature on CNS alterations in patients with glaucoma is reviewed with particular focus on neuroimaging and pathological studies. A theoretical framework for assessing whether glaucoma is truly a neurodegenerative disease is developed based on the comparison with neurodegenerative and nonneurodegenerative diseases. RESULTS Although there is convincing evidence of abnormalities in CNS regions distal to the optic nerve in glaucoma, these are similar to those seen in other disorders of the proximal visual pathways, such as other optic neuropathies or retinal diseases. Similarly, features of glaucoma that are similar to neurodegenerations are also seen in nonneurodegenerative diseases. CONCLUSIONS Glaucoma is less likely a primary neurodegeneration affecting the CNS and more likely a primary optic neuropathy with secondary effects in the CNS.
Collapse
|
28
|
|
29
|
Wolff JN, Ladoukakis ED, Enríquez JA, Dowling DK. Mitonuclear interactions: evolutionary consequences over multiple biological scales. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130443. [PMID: 24864313 PMCID: PMC4032519 DOI: 10.1098/rstb.2013.0443] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Fundamental biological processes hinge on coordinated interactions between genes spanning two obligate genomes--mitochondrial and nuclear. These interactions are key to complex life, and allelic variation that accumulates and persists at the loci embroiled in such intergenomic interactions should therefore be subjected to intense selection to maintain integrity of the mitochondrial electron transport system. Here, we compile evidence that suggests that mitochondrial-nuclear (mitonuclear) allelic interactions are evolutionarily significant modulators of the expression of key health-related and life-history phenotypes, across several biological scales--within species (intra- and interpopulational) and between species. We then introduce a new frontier for the study of mitonuclear interactions--those that occur within individuals, and are fuelled by the mtDNA heteroplasmy and the existence of nuclear-encoded mitochondrial gene duplicates and isoforms. Empirical evidence supports the idea of high-resolution tissue- and environment-specific modulation of intraindividual mitonuclear interactions. Predicting the penetrance, severity and expression patterns of mtDNA-induced mitochondrial diseases remains a conundrum. We contend that a deeper understanding of the dynamics and ramifications of mitonuclear interactions, across all biological levels, will provide key insights that tangibly advance our understanding, not only of core evolutionary processes, but also of the complex genetics underlying human mitochondrial disease.
Collapse
Affiliation(s)
- Jonci N Wolff
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney 2052, New South Wales, Australia Evolution and Ecology Research Centre, University of New South Wales, Sydney 2052, New South Wales, Australia School of Biological Sciences, Monash University, Clayton 3800, Victoria, Australia
| | | | - José A Enríquez
- Regenerative Cardiology Department, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain Departamento de Bioquímica, Universidad de Zaragoza, Zaragoza, Spain
| | - Damian K Dowling
- School of Biological Sciences, Monash University, Clayton 3800, Victoria, Australia
| |
Collapse
|
30
|
Im AR, Kim YH, Uddin MR, Chae S, Lee HW, Kim YS, Lee MY. Neuroprotective effects of Lycium chinense Miller against rotenone-induced neurotoxicity in PC12 cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2014; 41:1343-59. [PMID: 24228605 DOI: 10.1142/s0192415x13500900] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Rotenone, an inhibitor of mitochondrial complex I, has been widely regarded as a neurotoxin because it induces a Parkinson's disease-like syndrome. The fruit and root bark of Lycium chinense Miller have been used as traditional medicines in Asia to treat neurodegenerative diseases. In this study, we examined the neuroprotective effects of Lycium chinense Miller extracts in rotenone-treated PC12 cells. Treatment with rotenone reduced PC12 cell viability and cellular ATP levels. Conversely, caspase 3/7 activity, the ratio of Bax:Bcl-2 expression levels, mitochondrial superoxide level, and intracellular calcium (Ca(2+)) concentration were elevated. Pretreatment with Lycium chinense Miller extracts significantly increased cell viability and ATP levels. Additionally, they attenuated caspase activation, mitochondrial membrane depolarization and mitochondrial superoxide production. Moreover, confocal microscopy showed that the mitochondrial staining pattern was restored from that of extracts treated cells and that the increase in intracellular Ca (2+) level was blunted by treatment with the extracts. Our results suggest that Lycium chinense Miller extracts may have the possible beneficial effects in Parkinson's disease by attenuating rotenone induced toxicity.
Collapse
Affiliation(s)
- A-Rang Im
- KM-Based Herbal Drug Research Group, Korea Institute of Oriental Medicine, Daejeon 305-811, South Korea
| | | | | | | | | | | | | |
Collapse
|
31
|
Dowling DK. Evolutionary perspectives on the links between mitochondrial genotype and disease phenotype. Biochim Biophys Acta Gen Subj 2013; 1840:1393-403. [PMID: 24246955 DOI: 10.1016/j.bbagen.2013.11.013] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 10/24/2013] [Accepted: 11/11/2013] [Indexed: 01/09/2023]
Abstract
BACKGROUND Disorders of the mitochondrial respiratory chain are heterogeneous in their symptoms and underlying genetics. Simple links between candidate mutations and expression of disease phenotype typically do not exist. It thus remains unclear how the genetic variation in the mitochondrial genome contributes to the phenotypic expression of complex traits and disease phenotypes. SCOPE OF REVIEW I summarize the basic genetic processes known to underpin mitochondrial disease. I highlight other plausible processes, drawn from the evolutionary biological literature, whose contribution to mitochondrial disease expression remains largely empirically unexplored. I highlight recent advances to the field, and discuss common-ground and -goals shared by researchers across medical and evolutionary domains. MAJOR CONCLUSIONS Mitochondrial genetic variance is linked to phenotypic variance across a variety of traits (e.g. reproductive function, life expectancy) fundamental to the upkeep of good health. Evolutionary theory predicts that mitochondrial genomes are destined to accumulate male-harming (but female-friendly) mutations, and this prediction has received proof-of-principle support. Furthermore, mitochondrial effects on the phenotype are typically manifested via interactions between mitochondrial and nuclear genes. Thus, whether a mitochondrial mutation is pathogenic in effect can depend on the nuclear genotype in which is it expressed. GENERAL SIGNIFICANCE Many disease phenotypes associated with OXPHOS malfunction might be determined by the outcomes of mitochondrial-nuclear interactions, and by the evolutionary forces that historically shaped mitochondrial DNA (mtDNA) sequences. Concepts and results drawn from the evolutionary sciences can have broad, but currently under-utilized, applicability to the medical sciences and provide new insights into understanding the complex genetics of mitochondrial disease. This article is part of a Special Issue entitled Frontiers of Mitochondrial Research.
Collapse
Affiliation(s)
- Damian K Dowling
- School of Biological Sciences, Monash University, Clayton 3800, VIC Australia
| |
Collapse
|
32
|
Antiparkinson's and free radical scavenging study of ethyl acetate fraction of ethanolic extract of Leucas lanata. ACTA ACUST UNITED AC 2013. [DOI: 10.1016/j.dit.2013.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
33
|
Lee MY, Choi EJ, Lee MK, Lee JJ. Epigallocatechin gallate attenuates L-DOPA-induced apoptosis in rat PC12 cells. Nutr Res Pract 2013; 7:249-55. [PMID: 23964310 PMCID: PMC3746157 DOI: 10.4162/nrp.2013.7.4.249] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 05/11/2013] [Accepted: 05/16/2013] [Indexed: 11/09/2022] Open
Abstract
In this study, the protective effects of EGCG on L-3,4-dihydroxyphenylalanine (L-DOPA)-induced oxidative cell death in catecholaminergic PC12 cells, the in vitro model of Parkinson's disease, were investigated. Treatment with L-DOPA at concentrations higher than 150 µM caused cytotoxicity in PC12 cells, as determined using the 3-(4,5-dimetylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometry detection. The apoptotic ratio was similar in cells treated with 100 µM EGCG plus 150 µM L-DOPA (5.02%) and the control (0.96%) (P > 0.05), and was lower than that of cells treated with L-DOPA only (32.24%, P < 0.05). The generation level of ROS (% of control) in cells treated with EGCG plus L-DOPA was lower than that in cells treated with L-DOPA only (123.90% vs 272.32%, P < 0.05). The optical density in production of TBARS in cells treated with L-DOPA only was higher than that in the control (0.27 ± 0.05 vs 0.08 ± 0.04, P < 0.05), and in cells treated with EGCG only (0.14 ± 0.02, P < 0.05), and EGCG plus L-DOPA (0.13 ± 0.02, P < 0.05). The intracellular level of GSH in cells treated with EGCG plus L-DOPA was higher than that in cells treated with L-DOPA only (233.25 ± 16.44 vs 119.23 ± 10.25, P < 0.05). These results suggest that EGCG protects against L-DOPA-induced oxidative apoptosis in PC12 cells, and might be a potent neuroprotective agent.
Collapse
Affiliation(s)
- Myung-Yul Lee
- Department of Food and Nutrition, College of Natural Science, Chosun University, 301, Pilmun-daero, Dong-gu, Gwangju 501-759, Korea
| | | | | | | |
Collapse
|
34
|
Zhang L, Karsten P, Hamm S, Pogson JH, Müller-Rischart AK, Exner N, Haass C, Whitworth AJ, Winklhofer KF, Schulz JB, Voigt A. TRAP1 rescues PINK1 loss-of-function phenotypes. Hum Mol Genet 2013; 22:2829-41. [PMID: 23525905 DOI: 10.1093/hmg/ddt132] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PTEN-induced kinase 1 (PINK1) is a serine/threonine kinase that is localized to mitochondria. It protects cells from oxidative stress by suppressing mitochondrial cytochrome c release, thereby preventing cell death. Mutations in Pink1 cause early-onset Parkinson's disease (PD). Consistently, mitochondrial function is impaired in Pink1-linked PD patients and model systems. Previously, in vitro analysis implied that the protective effects of PINK1 depend on phosphorylation of the downstream factor, TNF receptor-associated protein 1 (TRAP1). Furthermore, TRAP1 has been shown to mitigate α-Synuclein-induced toxicity, linking α-Synuclein directly to mitochondrial dysfunction. These data suggest that TRAP1 seems to mediate protective effects on mitochondrial function in pathways that are affected in PD. Here we investigated the potential of TRAP1 to rescue dysfunction induced by either PINK1 or Parkin deficiency in vivo and in vitro. We show that overexpression of human TRAP1 is able to mitigate Pink1 but not parkin loss-of-function phenotypes in Drosophila. In addition, detrimental effects observed after RNAi-mediated silencing of complex I subunits were rescued by TRAP1 in Drosophila. Moreover, TRAP1 was able to rescue mitochondrial fragmentation and dysfunction upon siRNA-induced silencing of Pink1 but not parkin in human neuronal SH-SY5Y cells. Thus, our data suggest a functional role of TRAP1 in maintaining mitochondrial integrity downstream of PINK1 and complex I deficits but parallel to or upstream of Parkin.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen D-52074, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Mitochondria-specific accumulation of amyloid β induces mitochondrial dysfunction leading to apoptotic cell death. PLoS One 2012; 7:e34929. [PMID: 22514691 PMCID: PMC3325919 DOI: 10.1371/journal.pone.0034929] [Citation(s) in RCA: 184] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 03/07/2012] [Indexed: 12/30/2022] Open
Abstract
Mitochondria are best known as the essential intracellular organelles that host the homeostasis required for cellular survival, but they also have relevance in diverse disease-related conditions, including Alzheimer's disease (AD). Amyloid β (Aβ) peptide is the key molecule in AD pathogenesis, and has been highlighted in the implication of mitochondrial abnormality during the disease progress. Neuronal exposure to Aβ impairs mitochondrial dynamics and function. Furthermore, mitochondrial Aβ accumulation has been detected in the AD brain. However, the underlying mechanism of how Aβ affects mitochondrial function remains uncertain, and it is questionable whether mitochondrial Aβ accumulation followed by mitochondrial dysfunction leads directly to neuronal toxicity. This study demonstrated that an exogenous Aβ1–42 treatment, when applied to the hippocampal cell line of mice (specifically HT22 cells), caused a deleterious alteration in mitochondria in both morphology and function. A clathrin-mediated endocytosis blocker rescued the exogenous Aβ1–42-mediated mitochondrial dysfunction. Furthermore, the mitochondria-targeted accumulation of Aβ1–42 in HT22 cells using Aβ1–42 with a mitochondria-targeting sequence induced the identical morphological alteration of mitochondria as that observed in the APP/PS AD mouse model and exogenous Aβ1–42-treated HT22 cells. In addition, subsequent mitochondrial dysfunctions were demonstrated in the mitochondria-specific Aβ1–42 accumulation model, which proved indistinguishable from the mitochondrial impairment induced by exogenous Aβ1–42-treated HT22 cells. Finally, cellular toxicity was directly induced by mitochondria-targeted Aβ1–42 accumulation, which mimics the apoptosis process in exogenous Aβ1–42-treated HT22 cells. Taken together, these results indicate that mitochondria-targeted Aβ1–42 accumulation is the necessary and sufficient condition for Aβ-mediated mitochondria impairments, and leads directly to cellular death rather than along with other Aβ-mediated signaling alterations.
Collapse
|
36
|
Lu'o'ng KVQ, Nguyên LTH. Thiamine and Parkinson's disease. J Neurol Sci 2012; 316:1-8. [PMID: 22385680 DOI: 10.1016/j.jns.2012.02.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 02/03/2012] [Accepted: 02/08/2012] [Indexed: 11/20/2022]
Abstract
Parkinson's disease (PD) is the second most common form of neurodegeneration in the elderly population. PD is clinically characterized by tremors, rigidity, slowness of movement and postural imbalance. A significant association has been demonstrated between PD and low levels of thiamine in the serum, which suggests that elevated thiamine levels might provide protection against PD. Genetic studies have helped identify a number of factors that link thiamine to PD pathology, including the DJ-1 gene, excitatory amino acid transporters (EAATs), the α-ketoglutarate dehydrogenase complex (KGDHC), coenzyme Q10 (CoQ10 or ubiquinone), lipoamide dehydrogenase (LAD), chromosome 7, transcription factor p53, the renin-angiotensin system (RAS), heme oxygenase-1 (HO-1), and poly(ADP-ribose) polymerase-1 gene (PARP-1). Thiamine has also been implicated in PD through its effects on L-type voltage-sensitive calcium channels (L-VSCC), matrix metalloproteinases (MMPs), prostaglandins (PGs), cyclooxygenase-2 (COX-2), reactive oxygen species (ROS), and nitric oxide synthase (NOS). Recent studies highlight a possible relationship between thiamine and PD. Genetic studies provide opportunities to determine which proteins may link thiamine to PD pathology. Thiamine can also act through a number of non-genomic mechanisms that include protein expression, oxidative stress, inflammation, and cellular metabolism. Further studies are needed to determine the benefits of using thiamine as a treatment for PD.
Collapse
|
37
|
Butler EK, Voigt A, Lutz AK, Toegel JP, Gerhardt E, Karsten P, Falkenburger B, Reinartz A, Winklhofer KF, Schulz JB. The mitochondrial chaperone protein TRAP1 mitigates α-Synuclein toxicity. PLoS Genet 2012; 8:e1002488. [PMID: 22319455 PMCID: PMC3271059 DOI: 10.1371/journal.pgen.1002488] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 12/02/2011] [Indexed: 01/13/2023] Open
Abstract
Overexpression or mutation of α-Synuclein is associated with protein aggregation and interferes with a number of cellular processes, including mitochondrial integrity and function. We used a whole-genome screen in the fruit fly Drosophila melanogaster to search for novel genetic modifiers of human [A53T]α-Synuclein-induced neurotoxicity. Decreased expression of the mitochondrial chaperone protein tumor necrosis factor receptor associated protein-1 (TRAP1) was found to enhance age-dependent loss of fly head dopamine (DA) and DA neuron number resulting from [A53T]α-Synuclein expression. In addition, decreased TRAP1 expression in [A53T]α-Synuclein-expressing flies resulted in enhanced loss of climbing ability and sensitivity to oxidative stress. Overexpression of human TRAP1 was able to rescue these phenotypes. Similarly, human TRAP1 overexpression in rat primary cortical neurons rescued [A53T]α-Synuclein-induced sensitivity to rotenone treatment. In human (non)neuronal cell lines, small interfering RNA directed against TRAP1 enhanced [A53T]α-Synuclein-induced sensitivity to oxidative stress treatment. [A53T]α-Synuclein directly interfered with mitochondrial function, as its expression reduced Complex I activity in HEK293 cells. These effects were blocked by TRAP1 overexpression. Moreover, TRAP1 was able to prevent alteration in mitochondrial morphology caused by [A53T]α-Synuclein overexpression in human SH-SY5Y cells. These results indicate that [A53T]α-Synuclein toxicity is intimately connected to mitochondrial dysfunction and that toxicity reduction in fly and rat primary neurons and human cell lines can be achieved using overexpression of the mitochondrial chaperone TRAP1. Interestingly, TRAP1 has previously been shown to be phosphorylated by the serine/threonine kinase PINK1, thus providing a potential link of PINK1 via TRAP1 to α-Synuclein.
Collapse
Affiliation(s)
- Erin K. Butler
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
- Department of Neurodegeneration and Restorative Research, Center Molecular Physiology of the Brain (CMPB), Georg-August University Göttingen, Göttingen, Germany
- Göttingen Graduate School for Neurosciences and Molecular Biology (GGNB), Göttingen, Germany
| | - Aaron Voigt
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - A. Kathrin Lutz
- Neurobiochemistry, Adolf-Butenandt-Institute, Ludwig Maximilians University, Munich, Germany
| | - Jane P. Toegel
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - Ellen Gerhardt
- Department of Neurodegeneration and Restorative Research, Center Molecular Physiology of the Brain (CMPB), Georg-August University Göttingen, Göttingen, Germany
| | - Peter Karsten
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - Björn Falkenburger
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - Andrea Reinartz
- Department of Pathology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - Konstanze F. Winklhofer
- Neurobiochemistry, Adolf-Butenandt-Institute, Ludwig Maximilians University, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Jörg B. Schulz
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
- Jülich-Aachen Research Alliance (JARA) Brain, Jülich/Aachen, Germany
- * E-mail:
| |
Collapse
|
38
|
Infantile Progressive Hepatoencephalomyopathy with Combined OXPHOS Deficiency due to Mutations in the Mitochondrial Translation Elongation Factor Gene GFM1. JIMD Rep 2011; 5:113-22. [PMID: 23430926 DOI: 10.1007/8904_2011_107] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2011] [Revised: 10/17/2011] [Accepted: 10/20/2011] [Indexed: 12/30/2022] Open
Abstract
Mitochondrial disorders are a heterogeneous group of often multisystemic and early fatal diseases caused by defects in the oxidative phosphorylation (OXPHOS) system. Given the complexity and intricacy of the OXPHOS system, it is not surprising that the underlying molecular defect remains unidentified in many patients with a mitochondrial disorder. Here, we report the clinical features and diagnostic workup leading to the elucidation of the genetic basis for a combined complex I and IV OXPHOS deficiency secondary to a mitochondrial translational defect in an infant who presented with rapidly progressive liver failure, encephalomyopathy, and severe refractory lactic acidemia. Sequencing of the GFM1 gene revealed two inherited novel, heterozygous mutations: a.539delG (p.Gly180AlafsX11) in exon 4 which resulted in a frameshift mutation, and a second c.688G > A (p.Gly230Ser) mutation in exon 5. This missense mutation is likely to be pathogenic since it affects an amino acid residue that is highly conserved across species and is absent from the dbSNP and 1,000 genomes databases. Review of literature and comparison were made with previously reported cases of this recently identified mitochondrial disorder encoded by a nuclear gene. Although limited in number, nuclear gene defects causing mitochondrial translation abnormalities represent a new, rapidly expanding field of mitochondrial medicine and should potentially be considered in the diagnostic investigation of infants with progressive hepatoencephalomyopathy and combined OXPHOS disorders.
Collapse
|
39
|
Buneeva OA, Medvedev AE. Mitochondrial dysfunction in Parkinson’s disease. BIOCHEMISTRY MOSCOW-SUPPLEMENT SERIES B-BIOMEDICAL CHEMISTRY 2011. [DOI: 10.1134/s1990750811040032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
40
|
Baloyannis SJ. Mitochondria are related to synaptic pathology in Alzheimer's disease. Int J Alzheimers Dis 2011; 2011:305395. [PMID: 21922047 PMCID: PMC3171689 DOI: 10.4061/2011/305395] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Accepted: 07/12/2011] [Indexed: 01/08/2023] Open
Abstract
Morphological alterations of mitochondria may play an important role in the pathogenesis of Alzheimer's disease, been associated with oxidative stress and Aβ-peptide-induced toxicity. We proceeded to estimation of mitochondria on electron micrographs of autopsy specimens of Alzheimer's disease. We found substantial morphological and morphometric changes of the mitochondria in the neurons of the hippocampus, the neocortex, the cerebellar cortex, the thalamus, the globus pallidus, the red nucleus, the locus coeruleus, and the climbing fibers. The alterations consisted of considerable changes of the cristae, accumulation of osmiophilic material, and modification of the shape and size. Mitochondrial alterations were prominent in neurons, which showed a depletion of dendritic spines and loss of dendritic branches. Mitochondrial alterations are not related with the accumulation of amyloid deposits, but are prominent whenever fragmentation of the Golgi apparatus exists. Morphometric analysis showed also that mitochondria are significantly reduced in neurons, which demonstrated synaptic pathology.
Collapse
Affiliation(s)
- Stavros J Baloyannis
- Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54006 Thessaloniki, Greece
| |
Collapse
|
41
|
Gusdon AM, Chu CT. To eat or not to eat: neuronal metabolism, mitophagy, and Parkinson's disease. Antioxid Redox Signal 2011; 14:1979-87. [PMID: 21126205 PMCID: PMC3078495 DOI: 10.1089/ars.2010.3763] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Neurons are exquisitely dependent upon mitochondrial respiration to support energy-demanding functions. Mechanisms that regulate mitochondrial quality control have recently taken center stage in Parkinson's disease research, particularly the selective degradation of mitochondria by autophagy (mitophagy). Unlike other cells, neurons show limited glycolytic potential, and both insufficient and excessive mitophagy have been linked to neurodegeneration. Kinases implicated in regulating mammalian mitophagy include extracellular signal-regulated protein kinases (ERK1/2) and PTEN-induced kinase 1 (PINK1). Increased expression of full-length PINK1 enhances recruitment of parkin to chemically depolarized mitochondria, resulting in rapid mitochondrial clearance in transformed cell lines. As parkin and PINK1 mutations cause autosomal recessive parkinsonism, potential defects in clearing dysfunctional mitochondria may contribute to mitochondrial abnormalities in disease. Given the unique features of metabolic regulation in neurons, however, mechanisms regulating mitochondrial network stability and the threshold for mitophagy are likely to vary from cells that preferentially utilize aerobic glycolysis. Moreover, removal of the entire mitochondrial complement may represent part of a neuronal cell death pathway. Future work utilizing physiological injuries that affect only a subset of mitochondria would help to elucidate whether defective recognition of damaged mitochondria, or alternatively, inability to maintain or generate healthy mitochondria, play the major roles in parkinsonian neurodegeneration.
Collapse
Affiliation(s)
- Aaron M Gusdon
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | |
Collapse
|
42
|
Abstract
Mitochondrial structural and functional abnormalities in Parkinson's disease and experimental animal models of this pathology are described. Special attention is paid to the inactivation of mitochondrial enzymes, mutations in mitochondrial and nuclear DNA, and genomic and proteomic research of mitochondrial proteins in Parkinson's disease and experimental parkinsonism of animals.
Collapse
|
43
|
Neuroprotective profile of the multitarget drug rasagiline in Parkinson's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2011; 100:127-49. [DOI: 10.1016/b978-0-12-386467-3.00007-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
44
|
Sriram K, Lin GX, Jefferson AM, Roberts JR, Wirth O, Hayashi Y, Krajnak KM, Soukup JM, Ghio AJ, Reynolds SH, Castranova V, Munson AE, Antonini JM. Mitochondrial dysfunction and loss of Parkinson's disease‐linked proteins contribute to neurotoxicity of manganese‐containing welding fumes. FASEB J 2010. [DOI: 10.1096/fj.10.163964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Krishnan Sriram
- Health Effects Laboratory, National Institute for Occupational Safety and Health Morgantown West Virginia USA
| | - Gary X. Lin
- Health Effects Laboratory, National Institute for Occupational Safety and Health Morgantown West Virginia USA
| | - Amy M. Jefferson
- Health Effects Laboratory, National Institute for Occupational Safety and Health Morgantown West Virginia USA
| | - Jenny R. Roberts
- Health Effects Laboratory, National Institute for Occupational Safety and Health Morgantown West Virginia USA
| | - Oliver Wirth
- Health Effects Laboratory, National Institute for Occupational Safety and Health Morgantown West Virginia USA
| | - Yusuke Hayashi
- Health Effects Laboratory, National Institute for Occupational Safety and Health Morgantown West Virginia USA
| | - Kristine M. Krajnak
- Health Effects Laboratory, National Institute for Occupational Safety and Health Morgantown West Virginia USA
| | - Joleen M. Soukup
- National Health and Environmental Effects Research LaboratoryU.S. Environmental Protection Agency, Research Triangle Park North Carolina USA
| | - Andrew J. Ghio
- National Health and Environmental Effects Research LaboratoryU.S. Environmental Protection Agency, Research Triangle Park North Carolina USA
| | - Steven H. Reynolds
- Health Effects Laboratory, National Institute for Occupational Safety and Health Morgantown West Virginia USA
| | - Vincent Castranova
- Health Effects Laboratory, National Institute for Occupational Safety and Health Morgantown West Virginia USA
| | - Albert E. Munson
- Health Effects Laboratory, National Institute for Occupational Safety and Health Morgantown West Virginia USA
| | - James M. Antonini
- Health Effects Laboratory, National Institute for Occupational Safety and Health Morgantown West Virginia USA
| |
Collapse
|
45
|
Rasagiline: A novel anti-Parkinsonian monoamine oxidase-B inhibitor with neuroprotective activity. Prog Neurobiol 2010; 92:330-44. [DOI: 10.1016/j.pneurobio.2010.06.008] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Revised: 06/10/2010] [Accepted: 06/14/2010] [Indexed: 11/17/2022]
|
46
|
Sriram K, Lin GX, Jefferson AM, Roberts JR, Wirth O, Hayashi Y, Krajnak KM, Soukup JM, Ghio AJ, Reynolds SH, Castranova V, Munson AE, Antonini JM. Mitochondrial dysfunction and loss of Parkinson's disease-linked proteins contribute to neurotoxicity of manganese-containing welding fumes. FASEB J 2010; 24:4989-5002. [PMID: 20798247 DOI: 10.1096/fj.10-163964] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Welding generates complex metal aerosols, inhalation of which is linked to adverse health effects among welders. An important health concern of welding fume (WF) exposure is neurological dysfunction akin to Parkinson's disease (PD), thought to be mediated by manganese (Mn) in the fumes. Also, there is a proposition that welding might accelerate the onset of PD. Our recent findings link the presence of Mn in the WF with dopaminergic neurotoxicity seen in rats exposed to manual metal arc-hard surfacing (MMA-HS) or gas metal arc-mild steel (GMA-MS) fumes. To elucidate the molecular mechanisms further, we investigated the association of PD-linked (Park) genes and mitochondrial function in causing dopaminergic abnormality. Repeated instillations of the two fumes at doses that mimic ∼1 to 5 yr of worker exposure resulted in selective brain accumulation of Mn. This accumulation caused impairment of mitochondrial function and loss of tyrosine hydroxylase (TH) protein, indicative of dopaminergic injury. A fascinating finding was the altered expression of Parkin (Park2), Uchl1 (Park5), and Dj1 (Park7) proteins in dopaminergic brain areas. A similar regimen of manganese chloride (MnCl(2)) also caused extensive loss of striatal TH, mitochondrial electron transport components, and Park proteins. As mutations in PARK genes have been linked to early-onset PD in humans, and because welding is implicated as a risk factor for parkinsonism, PARK genes might play a critical role in WF-mediated dopaminergic dysfunction. Whether these molecular alterations culminate in neurobehavioral and neuropathological deficits reminiscent of PD remains to be ascertained.
Collapse
Affiliation(s)
- Krishnan Sriram
- Toxicology and Molecular Biology Branch, Mailstop L-3014, CDC-NIOSH, 1095 Willowdale Rd., Morgantown, WV 26505, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ho JWM, Ho PWL, Zhang WY, Liu HF, Kwok KHH, Yiu DCW, Chan KH, Kung MHW, Ramsden DB, Ho SL. Transcriptional regulation of UCP4 by NF-kappaB and its role in mediating protection against MPP+ toxicity. Free Radic Biol Med 2010; 49:192-204. [PMID: 20385226 DOI: 10.1016/j.freeradbiomed.2010.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Revised: 03/17/2010] [Accepted: 04/01/2010] [Indexed: 12/20/2022]
Abstract
Mitochondrial uncoupling protein-4 (UCP4) enhances neuronal cell survival in MPP(+)-induced toxicity by suppressing oxidative stress and preserving intracellular ATP and mitochondrial membrane potential. UCP4 expression is increased by MPP(+), but its regulation is unknown. Using serial human UCP4 promoter-luciferase reporter gene constructs, we identified and characterized several cis-acting elements that can regulate UCP4 expression. Core promoter activity exists within 100 bp upstream of the transcription initiation site (TIS=+1). Both CAAT box (-33/-27) and Sp1 (-62/-49) elements are crucial and act synergistically in its transcription. We identified a NF-kappaB putative binding site at -507/-495. Mutation of this site significantly decreased UCP4 promoter activity. Activation of NF-kappaB by TNFalpha or cycloheximide increased, whereas its inhibition by 4-hydroxy-2-nonenal or transfection of pIkappaBalphaM suppressed, UCP4 promoter activity. NF-kappaB inhibition significantly suppressed the MPP(+)-induced increase in UCP4 expression. MPP(+) increased specific binding of NF-kappaB protein complexes to this site in electrophoretic mobility shift assay. Both UCP4 knockdown and NF-kappaB inhibition exacerbated MPP(+)-induced cell death. We present the first direct evidence that UCP4 is regulated by NF-kappaB, mediated via a functional NF-kappaB site in its promoter region, and that UCP4 has a significant role in NF-kappaB prosurvival signaling, mediating its protection against MPP(+) toxicity.
Collapse
Affiliation(s)
- Jessica Wing-Man Ho
- Division of Neurology, University Department of Medicine, University of Hong Kong, Hong Kong, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Chu CT. A pivotal role for PINK1 and autophagy in mitochondrial quality control: implications for Parkinson disease. Hum Mol Genet 2010; 19:R28-37. [PMID: 20385539 DOI: 10.1093/hmg/ddq143] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The PTEN-induced putative kinase 1 (PINK1) is a mitochondrially targeted serine-threonine kinase, which is linked to autosomal recessive familial parkinsonism. Current literature implicates PINK1 as a pivotal regulator of mitochondrial quality control, promoting maintenance of respiring mitochondrial networks through cristae stabilization, phosphorylation of chaperones and possibly regulation of mitochondrial transport or autophagy. Pulse-chase studies indicate that PINK1 is rapidly processed into at least two shorter forms, which are distributed in both mitochondrial and cytosolic compartments. Through indirect regulation of mitochondrial proteases and Drp1, PINK1 may act to facilitate localized repair and fusion in response to minor mitochondrial stress. With severe mitochondrial damage, PINK1 facilitates aggregation and clearance of depolarized mitochondria through interactions with Parkin and possibly Beclin1. This switch in function most probably involves altered processing, post-translational modification and/or localization of PINK1, as overexpression of full-length PINK1 is required for mitochondrial Parkin recruitment. Under conditions of PINK1 deficiency, dysregulation of reactive oxygen species, electron transport chain function and calcium homeostasis trigger altered mitochondrial dynamics, indicating compromise of mitochondrial quality control mechanisms. Nevertheless, Parkin- and Beclin1-regulated mitochondrial autophagy remains effective at recycling PINK1-deficient mitochondria; failure of this final tier of mitochondrial quality control contributes to cell death. Thus, PINK1 plays a pivotal, multifactorial role in mitochondrial homeostasis. As autophagic recycling represents the final tier of mitochondrial quality control, whether PINK1 levels are enhanced or reduced, strategies to promote selective mitophagy and mitochondrial biogenesis may prove effective for multiple forms of Parkinson's disease.
Collapse
Affiliation(s)
- Charleen T Chu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
49
|
Goswami B, Tayal D, Gupta N, Mallika V. Paraoxonase: a multifaceted biomolecule. Clin Chim Acta 2009; 410:1-12. [PMID: 19799889 DOI: 10.1016/j.cca.2009.09.025] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Revised: 09/19/2009] [Accepted: 09/22/2009] [Indexed: 12/13/2022]
Abstract
BACKGROUND Paraoxonase enzyme was first identified as a protective barrier against organophosphorus poisoning. After painstaking research spanning the last three decades, the knowledge about this enzyme has increased immensely. The present review attempts to elaborate the role of paraoxonase enzyme in normal physiology as well as provide an overview of the various disorders in which the enzyme may have a role in etiopathogenesis. METHODS The literature was searched from the websites of the National Library of Medicine (http://www.ncbi.nlm.nih.gov/) and Pub Med Central, the U.S. National Library of Medicine's digital archive of life sciences journal literature. RESULTS Paraoxonase acts as an important antioxidant enzyme against oxidative stress. The enzyme has been implicated in the pathogenesis of a number of disorders including cardiovascular disorders, cancers etc. CONCLUSIONS A better understanding of the molecular mechanism of the enzyme along with the regulatory circuits will help us to utilize agonists to potentiate the anti oxidant actions of the enzyme.
Collapse
Affiliation(s)
- Binita Goswami
- Department of Biochemistry, G B Pant Hospital, New Delhi, India.
| | | | | | | |
Collapse
|
50
|
Yang L, Shi Q, Ho DJ, Starkov AA, Wille EJ, Xu H, Chen HL, Zhang S, Stack CM, Calingasan NY, Gibson GE, Beal MF. Mice deficient in dihydrolipoyl succinyl transferase show increased vulnerability to mitochondrial toxins. Neurobiol Dis 2009; 36:320-30. [PMID: 19660549 DOI: 10.1016/j.nbd.2009.07.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 07/08/2009] [Accepted: 07/28/2009] [Indexed: 11/24/2022] Open
Abstract
The activity of a key mitochondrial tricarboxylic acid cycle enzyme, alpha-ketoglutarate dehydrogenase complex (KGDHC), declines in many neurodegenerative diseases. KGDHC consists of three subunits. The dihydrolipoyl succinyl transferase (DLST) component is unique to KGDHC. DLST(+/-) mice showed reduced mRNA and protein levels and decreased brain mitochondrial KGDHC activity. Neurotoxic effects of mitochondrial toxins were exacerbated in DLST(+/-) mice. MPTP produced a significantly greater reduction of striatal dopamine and tyrosine hydroxylase-positive neurons in the substantia nigra pars compacta of DLST(+/-) mice. DLST deficiency enhanced the severity of lipid peroxidation in the substantia nigra after MPTP treatment. Striatal lesions induced by either malonate or 3-nitropropionic acid (3-NP) were significantly larger in DLST(+/-) mice than in wildtype controls. DLST deficiency enhanced the 3-NP inhibition of mitochondria enzymes, and 3-NP induced protein and DNA oxidations. These observations support the hypothesis that reductions in KGDHC may impair the adaptability of the brain and contribute to the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lichuan Yang
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|