1
|
Sun F, Fang M, Zhang H, Song Q, Li S, Li Y, Jiang S, Yang L. Drp1: Focus on Diseases Triggered by the Mitochondrial Pathway. Cell Biochem Biophys 2024; 82:435-455. [PMID: 38438751 DOI: 10.1007/s12013-024-01245-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 03/06/2024]
Abstract
Drp1 (Dynamin-Related Protein 1) is a cytoplasmic GTPase protein encoded by the DNM1L gene that influences mitochondrial dynamics by mediating mitochondrial fission processes. Drp1 has been demonstrated to play an important role in a variety of life activities such as cell survival, proliferation, migration, and death. Drp1 has been shown to play different physiological roles under different physiological conditions, such as normal and inflammation. Recently studies have revealed that Drp1 plays a critical role in the occurrence, development, and aggravation of a series of diseases, thereby it serves as a potential therapeutic target for them. In this paper, we review the structure and biological properties of Drp1, summarize the biological processes that occur in the inflammatory response to Drp1, discuss its role in various cancers triggered by the mitochondrial pathway and investigate effective methods for targeting Drp1 in cancer treatment. We also synthesized the phenomena of Drp1 involving in the triggering of other diseases. The results discussed herein contribute to our deeper understanding of mitochondrial kinetic pathway-induced diseases and their therapeutic applications. It is critical for advancing the understanding of the mechanisms of Drp1-induced mitochondrial diseases and preventive therapies.
Collapse
Affiliation(s)
- Fulin Sun
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
- Health Science Center, Qingdao University, Qingdao, China
| | - Min Fang
- Department of Gynaecology, Qingdao Women and Children's Hospital, Qingdao, 266021, Shandong, China
| | - Huhu Zhang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Qinghang Song
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
- Health Science Center, Qingdao University, Qingdao, China
| | - Shuang Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Ya Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Shuyao Jiang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
- Health Science Center, Qingdao University, Qingdao, China
| | - Lina Yang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China.
| |
Collapse
|
2
|
Deng J, Lai G, Zhang C, Li K, Zhu W, Xie B, Zhong X. A robust primary liver cancer subtype related to prognosis and drug response based on a multiple combined classifying strategy. Heliyon 2024; 10:e25570. [PMID: 38352751 PMCID: PMC10861988 DOI: 10.1016/j.heliyon.2024.e25570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 01/13/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
The recurrence or resistance to treatment of primary liver cancer (PLL) is significantly related to the heterogeneity present within the tumor. In this study, we integrated prognosis risk score, mRNAsi index, and immune characteristics clustering to classify patients. The four subtypes obtained from the combined classification are associated with PLC's prognosis and drug response. In these subtypes, we observed mRNAsiH_ICCA subtype, the intersection between high mRNAsi and immune characteristics clustering A, had the worst prognosis. Specifically, immune characteristics clustering B (ICC_B) had high drug sensitivity in most drugs regardless of the value of mRNAsi. On the other hand, patients with low mRNAsi responded better to ten drugs including KU-55933 and NU7441, while patients with high mRNAsi might benefit from drugs like Leflunomide. By matching the specific characteristics of each combined subtype with the drug-induced cell line expression profile, we identified a group of potential therapeutic drugs that might regulate the expression of disease signature genes. We developed a feasible multiple combined typing strategy, hoping to guide therapeutic selection and promote the development of precision medicine.
Collapse
Affiliation(s)
- Jielian Deng
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Chongqing, China
- Medical Department, Yidu Cloud (Beijing) Technology Co., Beijing, China
| | - Guichuan Lai
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Cong Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Kangjie Li
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Wenyan Zhu
- Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing, China
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Medical Department, Yidu Cloud (Beijing) Technology Co., Beijing, China
| | - Biao Xie
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Xiaoni Zhong
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Baidya AT, Deshwal S, Das B, Mathew AT, Devi B, Sandhir R, Kumar R. Catalyzing a Cure: Discovery and development of LRRK2 inhibitors for the treatment of Parkinson's disease. Bioorg Chem 2024; 143:106972. [PMID: 37995640 DOI: 10.1016/j.bioorg.2023.106972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023]
Abstract
Parkinson's disease (PD) is an age-related second most common progressive neurodegenerative disorder that affects millions of people worldwide. Despite decades of research, no effective disease modifying therapeutics have reached clinics for treatment/management of PD. Leucine-rich repeat kinase 2 (LRRK2) which controls membrane trafficking and lysosomal function and its variant LRRK2-G2019S are involved in the development of both familial and sporadic PD. LRRK2, is therefore considered as a legitimate target for the development of therapeutics against PD. During the last decade, efforts have been made to develop effective, safe and selective LRRK2 inhibitors and also our understanding about LRRK2 has progressed. However, there is an urge to learn from the previously designed and reported LRRK2 inhibitors in order to effectively approach designing of new LRRK2 inhibitors. In this review, we have aimed to cover the pre-clinical studies undertaken to develop small molecule LRRK2 inhibitors by screening the patents and other available literature in the last decade. We have highlighted LRRK2 as targets in the progress of PD and subsequently covered detailed design, synthesis and development of diverse scaffolds as LRRK2 inhibitors. Moreover, LRRK2 inhibitors under clinical development has also been discussed. LRRK2 inhibitors seem to be potential targets for future therapeutic interventions in the treatment and management of PD and this review can act as a cynosure for guiding discovery, design, and development of selective and non-toxic LRRK2 inhibitors. Although, there might be challenges in developing effective LRRK2 inhibitors, the opportunity to successfully develop novel therapeutics targeting LRRK2 against PD has never been greater.
Collapse
Affiliation(s)
- Anurag Tk Baidya
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005, UP, India
| | - Sonam Deshwal
- Department of Biochemistry, Panjab University, Chandigarh 160014, India
| | - Bhanuranjan Das
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005, UP, India
| | - Alen T Mathew
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005, UP, India
| | - Bharti Devi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005, UP, India
| | - Rajat Sandhir
- Department of Biochemistry, Panjab University, Chandigarh 160014, India
| | - Rajnish Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005, UP, India.
| |
Collapse
|
4
|
Obergasteiger J, Castonguay AM, Pizzi S, Magnabosco S, Frapporti G, Lobbestael E, Baekelandt V, Hicks AA, Pramstaller PP, Gravel C, Corti C, Lévesque M, Volta M. The small GTPase Rit2 modulates LRRK2 kinase activity, is required for lysosomal function and protects against alpha-synuclein neuropathology. NPJ Parkinsons Dis 2023; 9:44. [PMID: 36973269 PMCID: PMC10042831 DOI: 10.1038/s41531-023-00484-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Abstract
In Parkinson's disease (PD) misfolded alpha-synuclein (aSyn) accumulates in the substantia nigra, where dopaminergic neurons are progressively lost. The mechanisms underlying aSyn pathology are still unclear, but they are hypothesized to involve the autophagy-lysosome pathway (ALP). LRRK2 mutations are a major cause of familial and sporadic PD, and LRRK2 kinase activity has been shown to be involved in pS129-aSyn inclusion modulation. We observed selective downregulation of the novel PD risk factor RIT2 in vitro and in vivo. Rit2 overexpression in G2019S-LRRK2 cells rescued ALP abnormalities and diminished aSyn inclusions. In vivo, viral mediated overexpression of Rit2 operated neuroprotection against AAV-A53T-aSyn. Furthermore, Rit2 overexpression prevented the A53T-aSyn-dependent increase of LRRK2 kinase activity in vivo. On the other hand, reduction of Rit2 levels leads to defects in the ALP, similar to those induced by the G2019S-LRRK2 mutation. Our data indicate that Rit2 is required for correct lysosome function, inhibits overactive LRRK2 to ameliorate ALP impairment, and counteracts aSyn aggregation and related deficits. Targeting Rit2 could represent an effective strategy to combat neuropathology in familial and idiopathic PD.
Collapse
Affiliation(s)
- Julia Obergasteiger
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, CERVO Brain Research Centre, 2601 Chemin de la Canardiere, Quebec, QC, Canada
| | - Anne-Marie Castonguay
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, CERVO Brain Research Centre, 2601 Chemin de la Canardiere, Quebec, QC, Canada
| | - Sara Pizzi
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy
| | - Stefano Magnabosco
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy
| | - Giulia Frapporti
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Evy Lobbestael
- Department of Neurosciences, KU Leuven, Herestraat 49 bus 1023, 3000, Leuven, Belgium
| | - Veerle Baekelandt
- Department of Neurosciences, KU Leuven, Herestraat 49 bus 1023, 3000, Leuven, Belgium
| | - Andrew A Hicks
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy
| | - Peter P Pramstaller
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy
| | - Claude Gravel
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, CERVO Brain Research Centre, 2601 Chemin de la Canardiere, Quebec, QC, Canada
| | - Corrado Corti
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, CERVO Brain Research Centre, 2601 Chemin de la Canardiere, Quebec, QC, Canada.
| | - Mattia Volta
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Volta 21, 39100, Bolzano, Italy.
| |
Collapse
|
5
|
Fields M, Marcuzzi A, Gonelli A, Celeghini C, Maximova N, Rimondi E. Mitochondria-Targeted Antioxidants, an Innovative Class of Antioxidant Compounds for Neurodegenerative Diseases: Perspectives and Limitations. Int J Mol Sci 2023; 24:ijms24043739. [PMID: 36835150 PMCID: PMC9960436 DOI: 10.3390/ijms24043739] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/06/2023] [Accepted: 02/11/2023] [Indexed: 02/15/2023] Open
Abstract
Neurodegenerative diseases comprise a wide spectrum of pathologies characterized by progressive loss of neuronal functions and structures. Despite having different genetic backgrounds and etiology, in recent years, many studies have highlighted a point of convergence in the mechanisms leading to neurodegeneration: mitochondrial dysfunction and oxidative stress have been observed in different pathologies, and their detrimental effects on neurons contribute to the exacerbation of the pathological phenotype at various degrees. In this context, increasing relevance has been acquired by antioxidant therapies, with the purpose of restoring mitochondrial functions in order to revert the neuronal damage. However, conventional antioxidants were not able to specifically accumulate in diseased mitochondria, often eliciting harmful effects on the whole body. In the last decades, novel, precise, mitochondria-targeted antioxidant (MTA) compounds have been developed and studied, both in vitro and in vivo, to address the need to counter the oxidative stress in mitochondria and restore the energy supply and membrane potentials in neurons. In this review, we focus on the activity and therapeutic perspectives of MitoQ, SkQ1, MitoVitE and MitoTEMPO, the most studied compounds belonging to the class of MTA conjugated to lipophilic cations, in order to reach the mitochondrial compartment.
Collapse
Affiliation(s)
- Matteo Fields
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Annalisa Marcuzzi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Correspondence:
| | - Arianna Gonelli
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Claudio Celeghini
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Natalia Maximova
- Department of Pediatrics, Pediatrics, Bone Marrow Transplant Unit, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | - Erika Rimondi
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
6
|
Boecker CA. The Role of LRRK2 in Intracellular Organelle Dynamics. J Mol Biol 2023:167998. [PMID: 36764357 DOI: 10.1016/j.jmb.2023.167998] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/02/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
Pathogenic mutations in the leucine-rich repeat kinase 2 (LRRK2) gene hyperactivate LRRK2 kinase activity and lead to the development of Parkinson's disease (PD). Membrane recruitment of LRRK2 and the identification of RAB GTPases as bona fide LRRK2 substrates strongly indicate that LRRK2 regulates intracellular trafficking. This review highlights the current literature on the role of LRRK2 in intracellular organelle dynamics. With a focus on the effects of LRRK2 on microtubule function, mitochondrial dynamics, the autophagy-lysosomal pathway, and synaptic vesicle trafficking, it summarizes our current understanding of how intracellular dynamics are altered upon pathogenic LRRK2 hyperactivation.
Collapse
Affiliation(s)
- C Alexander Boecker
- Department of Neurology, University Medical Center Goettingen, Robert-Koch-Strasse 40, 37075 Goettingen, Germany.
| |
Collapse
|
7
|
Yu H, Sun T, He X, Wang Z, Zhao K, An J, Wen L, Li JY, Li W, Feng J. Association between Parkinson's Disease and Diabetes Mellitus: From Epidemiology, Pathophysiology and Prevention to Treatment. Aging Dis 2022; 13:1591-1605. [PMID: 36465171 PMCID: PMC9662283 DOI: 10.14336/ad.2022.0325] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/25/2022] [Indexed: 08/27/2023] Open
Abstract
Diabetes mellitus (DM) and Parkinson's disease (PD) are both age-related diseases of global concern being among the most common chronic metabolic and neurodegenerative diseases, respectively. While both diseases can be genetically inherited, environmental factors play a vital role in their pathogenesis. Moreover, DM and PD have common underlying molecular mechanisms, such as misfolded protein aggregation, mitochondrial dysfunction, oxidative stress, chronic inflammation, and microbial dysbiosis. Recently, epidemiological and experimental studies have reported that DM affects the incidence and progression of PD. Moreover, certain antidiabetic drugs have been proven to decrease the risk of PD and delay its progression. In this review, we elucidate the epidemiological and pathophysiological association between DM and PD and summarize the antidiabetic drugs used in animal models and clinical trials of PD, which may provide reference for the clinical translation of antidiabetic drugs in PD treatment.
Collapse
Affiliation(s)
- Haiyang Yu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Tong Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Xin He
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Zhen Wang
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Health Sciences Institute, China Medical University, Shenyang, Liaoning, China.
| | - Kaidong Zhao
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Health Sciences Institute, China Medical University, Shenyang, Liaoning, China.
| | - Jing An
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Lulu Wen
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Jia-Yi Li
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Health Sciences Institute, China Medical University, Shenyang, Liaoning, China.
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Wen Li
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Health Sciences Institute, China Medical University, Shenyang, Liaoning, China.
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
8
|
Unraveling Pathophysiological Link Between Mitophagy Pathway and Vascular Dementia. ARCHIVES OF NEUROSCIENCE 2022. [DOI: 10.5812/ans-124588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Context: Vascular dementia (VaD) is the second most common type of dementia after Alzheimer’s disease worldwide. Vascular dementia is a neurodegenerative disorder characterized by gradual cognitive impairment. Ischemic and hemorrhagic strokes result in VaD, markedly distributing cerebral blood flow and decreasing patients’ cognitive and memory performance. Due to their high energy demands, neurons are more sensitive to cellular architecture changes and exposed to mitochondrial stress than other cell types. Mitochondrial dysfunction and selective autophagy of mitochondria, known as mitophagy, are associated with VaD. This review aims to elucidate the association between mitophagy and VaD. Evidence Acquisition: This review was conducted independently by at least two researchers dominant in various VaD studies. We searched databases including Elsevier, Google Scholar, and PubMed using the terms ‘vascular dementia’, ‘vascular cognitive impairment’, and ‘mitophagy’. We evaluated 70 articles on the relationship between VaD and mitophagy and interpreted the results. Adobe Photoshop 2022 was used for drawing figures by researchers. Results: The autophagy process plays a protective role in experimental VaD models via preserving vascular integrity and the structure of the blood-brain barrier, upregulating occludin and claudin protein expressions, reducing oxidative stress, and decreasing cognitive dysfunction. Some studies claim that autophagy could have adverse effects in a time-dependent manner against neuronal injury. Prolonged autophagy and overexpressed autophagic proteins induce ischemic injury and cause neuronal cells to undergo apoptotic cell death. Conclusions: Although there are limited studies on the activation of mitophagy-related pathways in VaD, and the definitive role of mitophagy in neuronal healing is unclear, further research is needed to elucidate mitophagy pathways in neurons.
Collapse
|
9
|
Pan ZN, Liu JC, Ju JQ, Wang Y, Sun SC. LRRK2 regulates actin assembly for spindle migration and mitochondrial function in mouse oocyte meiosis. J Mol Cell Biol 2021; 14:6464148. [PMID: 34918122 PMCID: PMC8962687 DOI: 10.1093/jmcb/mjab079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 11/16/2022] Open
Abstract
Leucine-rich-repeat kinase 2 (LRRK2) belongs to the Roco GTPase family and is a large multidomain protein harboring both GTPase and kinase activities. LRRK2 plays indispensable roles in many processes, such as autophagy and vesicle trafficking in mitosis. In this study, we showed the critical roles of LRRK2 in mammalian oocyte meiosis. LRRK2 is mainly accumulated at the meiotic spindle periphery during oocyte maturation. Depleting LRRK2 led to the polar body extrusion defects and also induced large polar bodies in mouse oocytes. Mass spectrometry analysis and co-immunoprecipitation results showed that LRRK2 was associated with several actin-regulating factors, such as Fascin and Rho-kinase (ROCK), and depletion of LRRK2 affected the expression of ROCK, phosphorylated cofilin, and Fascin. Further analysis showed that LRRK2 depletion did not affect spindle organization but caused the failure of spindle migration, which was largely due to the decrease of cytoplasmic actin filaments. Moreover, LRRK2 showed a similar localization pattern to mitochondria, and LRRK2 was associated with several mitochondria-related proteins. Indeed, mitochondrial distribution and function were both disrupted in LRRK2-depleted oocytes. In summary, our results indicated the critical roles of LRRK2 in actin assembly for spindle migration and mitochondrial function in mouse oocyte meiosis.
Collapse
Affiliation(s)
- Zhen-Nan Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jing-Cai Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jia-Qian Ju
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yue Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
10
|
Huang H, Zheng S, Lu M. Downregulation of lncRNA MEG3 is involved in Parkinson's disease. Metab Brain Dis 2021; 36:2323-2328. [PMID: 34643842 DOI: 10.1007/s11011-021-00835-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/28/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Long non-coding RNA (lncRNA) MEG3 regulates human cancers, while its role in Parkinson's disease (PD) is unknown. The present study explored the involvement of MEG3 in PD. METHODS This study enrolled PD patients (n = 79) and healthy controls (n = 62) who were admitted to the Second Affiliated Hospital of Nanchang University from May 2016 to March 2018. RT-qPCR was performed to measure the expression of MEG3 and LRRK2. ROC curve analysis was performed for diagnostic analysis. Cell transfections were performed to analyze the interaction between MEG3 and LRRK2. Cell apoptosis and MTT assays were performed to evaluate the effect of cell transfections on cell apoptosis and viability. RESULTS MEG3 was downregulated in PD patients compared to that in the healthy controls. ROC curve analysis showed altered expression of MEG3 in PD patients. MEG3 was also down-regulated in SH-SY5Y cells treated with MPP + . Overexpression of MEG3 increased the expression levels of leucine-rich repeat kinase 2 (LRRK2) in SH-SY5Y cells. In contrast, overexpression of LRRK2 showed no significant effects on MEG3. Overexpression of MEG3 improved the viability and inhibited the apoptosis of SH-SY5Y cells pretreated with MPP + . CONCLUSIONS In conclusion, lncRNA MEG3 is downregulated in PD and may affect the expression of LRRK2 to regulate cell viability and apoptosis involved in PD.
Collapse
Affiliation(s)
- Hui Huang
- Department of Neurosurgery, the Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, PR, 330006, China
| | - Suyue Zheng
- Department of Neurosurgery, the First Affiliated Hospital of Nanchang University, Nanchang, PR, 330006, China
| | - Mingwei Lu
- Department of Neurosurgery, the Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, PR, 330006, China.
| |
Collapse
|
11
|
Helton LG, Soliman A, von Zweydorf F, Kentros M, Manschwetus JT, Hall S, Gilsbach B, Ho FY, Athanasopoulos PS, Singh RK, LeClair TJ, Versées W, Raimondi F, Herberg FW, Gloeckner CJ, Rideout H, Kortholt A, Kennedy EJ. Allosteric Inhibition of Parkinson's-Linked LRRK2 by Constrained Peptides. ACS Chem Biol 2021; 16:2326-2338. [PMID: 34496561 DOI: 10.1021/acschembio.1c00487] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Leucine-Rich Repeat Kinase 2 (LRRK2) is a large, multidomain protein with dual kinase and GTPase function that is commonly mutated in both familial and idiopathic Parkinson's Disease (PD). While dimerization of LRRK2 is commonly detected in PD models, it remains unclear whether inhibition of dimerization can regulate catalytic activity and pathogenesis. Here, we show constrained peptides that are cell-penetrant, bind LRRK2, and inhibit LRRK2 activation by downregulating dimerization. We further show that inhibited dimerization decreases kinase activity and inhibits ROS production and PD-linked apoptosis in primary cortical neurons. While many ATP-competitive LRRK2 inhibitors induce toxicity and mislocalization of the protein in cells, these constrained peptides were found to not affect LRRK2 localization. The ability of these peptides to inhibit pathogenic LRRK2 kinase activity suggests that disruption of dimerization may serve as a new allosteric strategy to downregulate PD-related signaling pathways.
Collapse
Affiliation(s)
- Leah G. Helton
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - Ahmed Soliman
- Department of Cell Biochemistry, University of Groningen, 9747 Groningen, The Netherlands
| | - Felix von Zweydorf
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tübingen, Germany
| | - Michalis Kentros
- Center for Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, 115 27 Athens, Greece
| | - Jascha T. Manschwetus
- Department of Biochemistry, Institute for Biology, University of Kassel, 34132, Kassel, Germany
| | - Scotty Hall
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - Bernd Gilsbach
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tübingen, Germany
| | - Franz Y. Ho
- Department of Cell Biochemistry, University of Groningen, 9747 Groningen, The Netherlands
| | | | - Ranjan K. Singh
- VIB-VUB Center for Structural Biology, Pleinlaan 2, 1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Timothy J. LeClair
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - Wim Versées
- VIB-VUB Center for Structural Biology, Pleinlaan 2, 1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Francesco Raimondi
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, 56126, Pisa, Italy
| | - Friedrich W. Herberg
- Department of Biochemistry, Institute for Biology, University of Kassel, 34132, Kassel, Germany
| | - Christian Johannes Gloeckner
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tübingen, Germany
- Core Facility for Medical Bioanalytics, Center for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany
| | - Hardy Rideout
- Center for Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, 115 27 Athens, Greece
| | - Arjan Kortholt
- Department of Cell Biochemistry, University of Groningen, 9747 Groningen, The Netherlands
- Department of Pharmacology, Innovative Technologies Application and Research Center, Suleyman Demirel University, 32260 Isparta, Turkey
| | - Eileen J. Kennedy
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
12
|
Lee CY, Menozzi E, Chau KY, Schapira AHV. Glucocerebrosidase 1 and leucine-rich repeat kinase 2 in Parkinson disease and interplay between the two genes. J Neurochem 2021; 159:826-839. [PMID: 34618942 DOI: 10.1111/jnc.15524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 01/24/2023]
Abstract
The glucocerebrosidase 1 gene (GBA1), bi-allelic variants of which cause Gaucher disease (GD), encodes the lysosomal enzyme glucocerebrosidase (GCase) and is a risk factor for Parkinson Disease (PD). GBA1 variants are linked to a reduction in GCase activity in the brain. Variants in Leucine-Rich Repeat Kinase 2 (LRRK2), such as the gain-of-kinase-function variant G2019S, cause the most common familial form of PD. In patients without GBA1 and LRRK2 mutations, GCase and LRRK2 activity are also altered, suggesting that these two genes are implicated in all forms of PD and that they may play a broader role in PD pathogenesis. In this review, we review the proposed roles of GBA1 and LRRK2 in PD, focussing on the endolysosomal pathway. In particular, we highlight the discovery of Ras-related in brain (Rab) guanosine triphosphatases (GTPases) as LRRK2 kinase substrates and explore the links between increased LRRK2 activity and Rab protein function, lysosomal dysfunction, alpha-synuclein accumulation and GCase activity. We also discuss the discovery of RAB10 as a potential mediator of LRRK2 and GBA1 interaction in PD. Finally, we discuss the therapeutic implications of these findings, including current approaches and future perspectives related to novel drugs targeting LRRK2 and GBA1.
Collapse
Affiliation(s)
- Chiao-Yin Lee
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Elisa Menozzi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Kai-Yin Chau
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| |
Collapse
|
13
|
Zhang K, Zhu S, Li J, Jiang T, Feng L, Pei J, Wang G, Ouyang L, Liu B. Targeting autophagy using small-molecule compounds to improve potential therapy of Parkinson's disease. Acta Pharm Sin B 2021; 11:3015-3034. [PMID: 34729301 PMCID: PMC8546670 DOI: 10.1016/j.apsb.2021.02.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/28/2021] [Accepted: 02/19/2021] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD), known as one of the most universal neurodegenerative diseases, is a serious threat to the health of the elderly. The current treatment has been demonstrated to relieve symptoms, and the discovery of new small-molecule compounds has been regarded as a promising strategy. Of note, the homeostasis of the autolysosome pathway (ALP) is closely associated with PD, and impaired autophagy may cause the death of neurons and thereby accelerating the progress of PD. Thus, pharmacological targeting autophagy with small-molecule compounds has been drawn a rising attention so far. In this review, we focus on summarizing several autophagy-associated targets, such as AMPK, mTORC1, ULK1, IMPase, LRRK2, beclin-1, TFEB, GCase, ERRα, C-Abelson, and as well as their relevant small-molecule compounds in PD models, which will shed light on a clue on exploiting more potential targeted small-molecule drugs tracking PD treatment in the near future.
Collapse
Key Words
- 3-MA, 3-methyladenine
- 5-HT2A, Serotonin 2A
- 5-HT2C, serotonin 2C
- A2A, adenosine 2A
- AADC, aromatic amino acid decarboxylase
- ALP, autophagy-lysosomal pathway
- AMPK, 5ʹAMP-activated protein kinase
- ATG, autophagy related protein
- ATP13A2, ATPase cation transporting 13A2
- ATTEC, autophagosome-tethering compound
- AUC, the area under the curve
- AUTAC, autophagy targeting chimera
- Autophagy
- BAF, bafilomycinA1
- BBB, blood−brain barrier
- CL, clearance rate
- CMA, chaperone-mediated autophagy
- CNS, central nervous system
- COMT, catechol-O-methyltransferase
- DA, dopamine
- DAT, dopamine transporter
- DJ-1, Parkinson protein 7
- DR, dopamine receptor
- ER, endoplasmic reticulum
- ERRα, estrogen-related receptor alpha
- F, oral bioavailability
- GAPDH, glyceraldehyde 3-phosphate dehydrogenase
- GBA, glucocerebrosidase β acid
- GWAS, genome-wide association study
- HDAC6, histone deacetylase 6
- HSC70, heat shock cognate 71 kDa protein
- HSPA8, heat shock 70 kDa protein 8
- IMPase, inositol monophosphatase
- IPPase, inositol polyphosphate 1-phosphatase
- KI, knockin
- LAMP2A, lysosome-associated membrane protein 2 A
- LC3, light chain 3
- LIMP-2, lysosomal integrated membrane protein-2
- LRRK2, leucine-rich repeat sequence kinase 2
- LRS, leucyl-tRNA synthetase
- LUHMES, lund human mesencephalic
- Lamp2a, type 2A lysosomal-associated membrane protein
- MAO-B, monoamine oxidase B
- MPP+, 1-methyl-4-phenylpyridinium
- MPTP, 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine
- MYCBP2, MYC-binding protein 2
- NMDA, N-methyl-d-aspartic acid
- ONRs, orphan nuclear receptors
- PD therapy
- PD, Parkinson's disease
- PDE4, phosphodiesterase 4
- PI3K, phosphatidylinositol 3-kinase
- PI3P, phosphatidylinositol 3-phosphate
- PINK1, PTEN-induced kinase 1
- PLC, phospholipase C
- PREP, prolyl oligopeptidase
- Parkin, parkin RBR E3 ubiquitin−protein ligase
- Parkinson's disease (PD)
- ROS, reactive oxygen species
- SAR, structure–activity relationship
- SAS, solvent accessible surface
- SN, substantia nigra
- SNCA, α-synuclein gene
- SYT11, synaptotagmin 11
- Small-molecule compound
- TFEB, transcription factor EB
- TSC2, tuberous sclerosis complex 2
- Target
- ULK1, UNC-51-like kinase 1
- UPS, ubiquitin−proteasome system
- mAChR, muscarinic acetylcholine receptor
- mTOR, the mammalian target of rapamycin
- α-syn, α-synuclein
Collapse
|
14
|
Constitutive silencing of LRRK2 kinase activity leads to early glucocerebrosidase deregulation and late impairment of autophagy in vivo. Neurobiol Dis 2021; 159:105487. [PMID: 34419621 DOI: 10.1016/j.nbd.2021.105487] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 01/18/2023] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are associated with Parkinson's disease. LRRK2 modulates the autophagy-lysosome pathway (ALP), a clearance process subserving the quality control of cellular proteins and organelles. Since dysfunctional ALP might lead to α-synuclein accumulation and, hence, Parkinson's disease, LRRK2 kinase modulation of ALP, its age-dependence and relation with pSer129 α-synuclein inclusions were investigated in vivo. Striatal ALP markers were analyzed by Western blotting in 3, 12 and 20-month-old LRRK2 G2019S knock-in mice (bearing enhanced kinase activity), LRRK2 knock-out mice, LRRK2 D1994S knock-in (kinase-dead) mice and wild-type controls. The lysosomotropic agent chloroquine was used to investigate the autophagic flux in vivo. Quantitative Real-time PCR was used to quantify the transcript levels of key ALP genes. The activity of the lysosomal enzyme glucocerebrosidase was measured using enzymatic assay. Immunohistochemistry was used to co-localize LC3B puncta with pSer129 α-synuclein inclusion in striatal and nigral neurons. No genotype differences in ALP markers were observed at 3 months. Conversely, increase of LC3-I, p62, LAMP2 and GAPDH levels, decrease of p-mTOR levels and downregulation of mTOR and TFEB expression was observed in 12-month-old kinase-dead mice. The LC3-II/I ratio was reduced following administration of chloroquine, suggesting a defective autophagic flux. G2019S knock-in mice showed LAMP2 accumulation and downregulation of ALP key genes MAP1LC3B, LAMP2, mTOR, TFEB and GBA1. Subacute administration of the LRRK2 kinase inhibitor MLi-2 in wild-type and G2019S knock-in mice did not replicate the pattern of kinase-dead mice. Lysosomal glucocerebrosidase activity was increased in 3 and 12-month-old knock-out and kinase-dead mice. LC3B puncta accumulation and pSer129 α-synuclein inclusions were dissociated in striatal neurons of kinase-dead and G2019S knock-in mice. We conclude that constitutive LRRK2 kinase silencing results in early deregulation of GCase activity followed by late impairment of macroautophagy and chaperone-mediated autophagy.
Collapse
|
15
|
Yao L, Wu J, Koc S, Lu G. Genetic Imaging of Neuroinflammation in Parkinson's Disease: Recent Advancements. Front Cell Dev Biol 2021; 9:655819. [PMID: 34336822 PMCID: PMC8320775 DOI: 10.3389/fcell.2021.655819] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/14/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is one of the most prevalent neurodegenerative aging disorders characterized by motor and non-motor symptoms due to the selective loss of midbrain dopaminergic (DA) neurons. The decreased viability of DA neurons slowly results in the appearance of motor symptoms such as rigidity, bradykinesia, resting tremor, and postural instability. These symptoms largely depend on DA nigrostriatal denervation. Pharmacological and surgical interventions are the main treatment for improving clinical symptoms, but it has not been possible to cure PD. Furthermore, the cause of neurodegeneration remains unclear. One of the possible neurodegeneration mechanisms is a chronic inflammation of the central nervous system, which is mediated by microglial cells. Impaired or dead DA neurons can directly lead to microglia activation, producing a large number of reactive oxygen species and pro-inflammatory cytokines. These cytotoxic factors contribute to the apoptosis and death of DA neurons, and the pathological process of neuroinflammation aggravates the primary morbid process and exacerbates ongoing neurodegeneration. Therefore, anti-inflammatory treatment exerts a robust neuroprotective effect in a mouse model of PD. Since discovering the first mutation in the α-synuclein gene (SNCA), which can cause disease-causing, PD has involved many genes and loci such as LRRK2, Parkin, SNCA, and PINK1. In this article, we summarize the critical descriptions of the genetic factors involved in PD's occurrence and development (such as LRRK2, SNCA, Parkin, PINK1, and inflammasome), and these factors play a crucial role in neuroinflammation. Regulation of these signaling pathways and molecular factors related to these genetic factors can vastly improve the neuroinflammation of PD.
Collapse
Affiliation(s)
- Longping Yao
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiayu Wu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Sumeyye Koc
- Department of Neuroscience, Institute of Health Sciences, Ondokuz Mayıs University, Samsun, Turkey
| | - Guohui Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
16
|
Custodia A, Aramburu-Núñez M, Correa-Paz C, Posado-Fernández A, Gómez-Larrauri A, Castillo J, Gómez-Muñoz A, Sobrino T, Ouro A. Ceramide Metabolism and Parkinson's Disease-Therapeutic Targets. Biomolecules 2021; 11:945. [PMID: 34202192 PMCID: PMC8301871 DOI: 10.3390/biom11070945] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Ceramide is a bioactive sphingolipid involved in numerous cellular processes. In addition to being the precursor of complex sphingolipids, ceramides can act as second messengers, especially when they are generated at the plasma membrane of cells. Its metabolic dysfunction may lead to or be a consequence of an underlying disease. Recent reports on transcriptomics and electrospray ionization mass spectrometry analysis have demonstrated the variation of specific levels of sphingolipids and enzymes involved in their metabolism in different neurodegenerative diseases. In the present review, we highlight the most relevant discoveries related to ceramide and neurodegeneration, with a special focus on Parkinson's disease.
Collapse
Affiliation(s)
- Antía Custodia
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Marta Aramburu-Núñez
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Clara Correa-Paz
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Adrián Posado-Fernández
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Ana Gómez-Larrauri
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country, P.O. Box 644, 48980 Bilbao, Spain; (A.G.-L.); (A.G.-M.)
- Respiratory Department, Cruces University Hospital, Barakaldo, 48903 Bizkaia, Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Antonio Gómez-Muñoz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country, P.O. Box 644, 48980 Bilbao, Spain; (A.G.-L.); (A.G.-M.)
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Alberto Ouro
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| |
Collapse
|
17
|
Abe T, Kuwahara T. Targeting of Lysosomal Pathway Genes for Parkinson's Disease Modification: Insights From Cellular and Animal Models. Front Neurol 2021; 12:681369. [PMID: 34194386 PMCID: PMC8236816 DOI: 10.3389/fneur.2021.681369] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/20/2021] [Indexed: 01/01/2023] Open
Abstract
Previous genetic studies on hereditary Parkinson's disease (PD) have identified a set of pathogenic gene mutations that have strong impacts on the pathogenicity of PD. In addition, genome-wide association studies (GWAS) targeted to sporadic PD have nominated an increasing number of genetic variants that influence PD susceptibility. Although the clinical and pathological characteristics in hereditary PD are not identical to those in sporadic PD, α-synuclein, and LRRK2 are definitely associated with both types of PD, with LRRK2 mutations being the most frequent cause of autosomal-dominant PD. On the other hand, a significant portion of risk genes identified from GWAS have been associated with lysosomal functions, pointing to a critical role of lysosomes in PD pathogenesis. Experimental studies have suggested that the maintenance or upregulation of lysosomal activity may protect against neuronal dysfunction or degeneration. Here we focus on the roles of representative PD gene products that are implicated in lysosomal pathway, namely LRRK2, VPS35, ATP13A2, and glucocerebrosidase, and provide an overview of their disease-associated functions as well as their cooperative actions in the pathogenesis of PD, based on the evidence from cellular and animal models. We also discuss future perspectives of targeting lysosomal activation as a possible strategy to treat neurodegeneration.
Collapse
Affiliation(s)
- Tetsuro Abe
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoki Kuwahara
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
18
|
Yakhine-Diop SMS, Rodríguez-Arribas M, Canales-Cortés S, Martínez-Chacón G, Uribe-Carretero E, Blanco-Benítez M, Duque-González G, Paredes-Barquero M, Alegre-Cortés E, Climent V, Aiastui A, López de Munain A, Bravo-San Pedro JM, Niso-Santano M, Fuentes JM, González-Polo RA. The parkinsonian LRRK2 R1441G mutation shows macroautophagy-mitophagy dysregulation concomitant with endoplasmic reticulum stress. Cell Biol Toxicol 2021; 38:889-911. [PMID: 34060004 DOI: 10.1007/s10565-021-09617-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 05/12/2021] [Indexed: 12/15/2022]
Abstract
Autophagy is a mechanism responsible for the degradation of cellular components to maintain their homeostasis. However, autophagy is commonly altered and compromised in several diseases, including neurodegenerative disorders. Parkinson's disease (PD) can be considered a multifactorial disease because environmental factors, genetic factors, and aging are involved. Several genes are involved in PD pathology, among which the LRRK2 gene and its mutations, inherited in an autosomal dominant manner, are responsible for most genetic PD cases. The R1441G LRRK2 mutation is, after G2019S, the most important in PD pathogenesis. Our results demonstrate a relationship between the R1441G LRRK2 mutation and a mechanistic dysregulation of autophagy that compromises cell viability. This altered autophagy mechanism is associated with organellar stress including mitochondrial (which induces mitophagy) and endoplasmic reticulum (ER) stress, consistent with the fact that patients with this mutation are more vulnerable to toxins related to PD, such as MPP+.
Collapse
Affiliation(s)
- Sokhna M S Yakhine-Diop
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain.,Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain.,Instituto de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Mario Rodríguez-Arribas
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain.,Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain.,Instituto de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Saray Canales-Cortés
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain
| | - Guadalupe Martínez-Chacón
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain.,Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain.,Instituto de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Elisabet Uribe-Carretero
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain.,Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain.,Instituto de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Mercedes Blanco-Benítez
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain
| | - Gema Duque-González
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain
| | - Marta Paredes-Barquero
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain
| | - Eva Alegre-Cortés
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain
| | - Vicente Climent
- Departamento de Anatomía Y Embriología Humana, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - Ana Aiastui
- Cell Culture Platform, Donostia University Hospital, San Sebastián, Spain.,Neuroscience Area of Biodonostia Health Research Institute, Donostia University Hospital, San Sebastián, Spain
| | - Adolfo López de Munain
- Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain.,Department of Neurology, Donostia University Hospital, San Sebastian, Spain.,Ilundain Foundation, San Sebastian, Spain.,Department of Neurosciences, University of the Basque Country UPV-EHU, San Sebastián, Spain
| | - José M Bravo-San Pedro
- Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain.,Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Mireia Niso-Santano
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain. .,Instituto de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain.
| | - José M Fuentes
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain. .,Instituto de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain.
| | - Rosa A González-Polo
- Departamento de Bioquímica Y Biología Molecular Y Genética, Facultad de Enfermería Y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades (CIBERNED), Madrid, Spain. .,Instituto de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain.
| |
Collapse
|
19
|
Wang X, Guo G, Zhang J, Aebez N, Liu Z, Liu CF, Ross CA, Smith WW. Mutant-TMEM230-induced neurodegeneration and impaired axonal mitochondrial transport. Hum Mol Genet 2021; 30:1535-1542. [PMID: 34002226 DOI: 10.1093/hmg/ddab128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with movement disorders including resting tremor, rigidity, bradykinesia and postural instability. Recent studies have identified a new PD associated gene, TMEM230 (transmembrane protein 230). However, the pathological roles of TMEM230 and its variants are not fully understood. TMEM230 gene encodes two protein isoforms. Isoform2 is the major protein form (~95%) in human. In this study, we overexpress isoform2 TMEM230 variants (WT or PD-linked *184Wext*5 mutant) or knockdown endogenous protein in cultured SH-5Y5Y cells and mouse primary hippocampus neurons to study their pathological roles. We found that overexpression of WT and mutant TMEM230 or knockdown of endogenous TMEM230-induced neurodegeneration and impaired mitochondria transport at the retrograde direction in axons. Mutant TMEM230 caused more severe neurotoxicity and mitochondrial transport impairment than WT-TMEM230 did. Our results demonstrate that maintaining TMEM230 protein levels is critical for neuron survival and axon transport. These findings suggest that mutant-TMEM230-induced mitochondrial transport impairment could be the early event leading to neurite injury and neurodegeneration in PD development.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Institute of Neuroscience, Soochow University School of Medicine, Suzhou, Jiangsu 215123, China
| | - Gongbo Guo
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jinru Zhang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Nicolas Aebez
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Zhaohui Liu
- Department of Human Anatomy and Cytoneurobiology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chun-Feng Liu
- Institute of Neuroscience, Soochow University School of Medicine, Suzhou, Jiangsu 215123, China.,Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Christopher A Ross
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Wanli W Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
20
|
Inhibition of LRRK2 restores parkin-mediated mitophagy and attenuates intervertebral disc degeneration. Osteoarthritis Cartilage 2021; 29:579-591. [PMID: 33434630 DOI: 10.1016/j.joca.2021.01.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 11/24/2020] [Accepted: 01/02/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To elucidate the role of LRRK2 in intervertebral disc degeneration (IDD) as well as its mitophagy regulation mechanism. METHODS The expression of LRRK2 in human degenerative nucleus pulposus tissues as well as in oxidative stress-induced rat nucleus pulposus cells (NPCs) was detected by western blot. LRRK2 was knocked down in NPCs by lentivirus (LV)-shLRRK2 transfection; apoptosis and mitophagy were assessed by western blot, TUNEL assay, immunofluorescence staining and mitophagy detection assay in LRRK2-deficient NPCs under oxidative stress. After knockdown of Parkin in NPCs with siRNA transfection, apoptosis and mitophagy were further assessed. In puncture-induced rat IDD model, X-ray, MRI, hematoxylin-eosin (HE) and Safranin O-Fast green (SO) staining were performed to evaluate the therapeutic effects of LV-shLRRK2 on IDD. RESULTS We found that the expression of LRRK2 was increased in degenerative NPCs both in vivo and in vitro. LRRK2 deficiency significantly suppressed oxidative stress-induced mitochondria-dependent apoptosis in NPCs; meanwhile, mitophagy was promoted. However, these effects were abolished by the mitophagy inhibitor, suggesting the effect of LRRK2 on apoptosis in NPCs is mitophagy-dependent. Furthermore, Parkin knockdown study showed that LRRK2 deficiency activated mitophagy by recruiting Parkin. In vivo study demonstrated that LRRK2 inhibition ameliorated IDD in rats. CONCLUSIONS The results revealed that LRRK2 is involved in the pathogenesis of IDD, while knockdown of LRRK2 inhibits oxidative stress-induced apoptosis through mitophagy. Thus, inhibition of LRRK2 may be a promising therapeutic strategy for IDD.
Collapse
|
21
|
Benn CL, Dawson LA. Clinically Precedented Protein Kinases: Rationale for Their Use in Neurodegenerative Disease. Front Aging Neurosci 2020; 12:242. [PMID: 33117143 PMCID: PMC7494159 DOI: 10.3389/fnagi.2020.00242] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Kinases are an intensively studied drug target class in current pharmacological research as evidenced by the large number of kinase inhibitors being assessed in clinical trials. Kinase-targeted therapies have potential for treatment of a broad array of indications including central nervous system (CNS) disorders. In addition to the many variables which contribute to identification of a successful therapeutic molecule, drug discovery for CNS-related disorders also requires significant consideration of access to the target organ and specifically crossing the blood-brain barrier (BBB). To date, only a small number of kinase inhibitors have been reported that are specifically designed to be BBB permeable, which nonetheless demonstrates the potential for success. This review considers the potential for kinase inhibitors in the context of unmet medical need for neurodegenerative disease. A subset of kinases that have been the focus of clinical investigations over a 10-year period have been identified and discussed individually. For each kinase target, the data underpinning the validity of each in the context of neurodegenerative disease is critically evaluated. Selected molecules for each kinase are identified with information on modality, binding site and CNS penetrance, if known. Current clinical development in neurodegenerative disease are summarized. Collectively, the review indicates that kinase targets with sufficient rationale warrant careful design approaches with an emphasis on improving brain penetrance and selectivity.
Collapse
|
22
|
Choi SY, Lee JH, Chung AY, Jo Y, Shin JH, Park HC, Kim H, Lopez-Gonzalez R, Ryu JR, Sun W. Prevention of mitochondrial impairment by inhibition of protein phosphatase 1 activity in amyotrophic lateral sclerosis. Cell Death Dis 2020; 11:888. [PMID: 33087694 PMCID: PMC7578657 DOI: 10.1038/s41419-020-03102-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease caused by progressive loss of motor neurons (MNs) and subsequent muscle weakness. These pathological features are associated with numerous cellular changes, including alteration in mitochondrial morphology and function. However, the molecular mechanisms associating mitochondrial structure with ALS pathology are poorly understood. In this study, we found that Dynamin-related protein 1 (Drp1) was dephosphorylated in several ALS models, including those with SOD1 and TDP-43 mutations, and the dephosphorylation was mediated by the pathological induction of protein phosphatase 1 (PP1) activity in these models. Suppression of the PP1-Drp1 cascade effectively prevented ALS-related symptoms, including mitochondrial fragmentation, mitochondrial complex I impairment, axonal degeneration, and cell death, in primary neuronal culture models, iPSC-derived human MNs, and zebrafish models in vivo. These results suggest that modulation of PP1-Drp1 activity may be a therapeutic target for multiple pathological features of ALS.
Collapse
Affiliation(s)
- So Yoen Choi
- Department of Anatomy, Korea University College of Medicine, Brain Korea 21 plus, Seoul, 02841, Republic of Korea
- Department of Neurology, University of Massachusetts Medical school, Worcester, MA, USA
| | - Ju-Hyun Lee
- Department of Anatomy, Korea University College of Medicine, Brain Korea 21 plus, Seoul, 02841, Republic of Korea
| | - Ah-Young Chung
- Graduate School of Medicine, Korea University, Ansan, Gyeonggido, Republic of Korea
| | - Youhwa Jo
- Department of Anatomy, Korea University College of Medicine, Brain Korea 21 plus, Seoul, 02841, Republic of Korea
| | - Joo-Ho Shin
- Division of Pharmacology, Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do, 440-746, Republic of Korea
| | - Hae-Chul Park
- Graduate School of Medicine, Korea University, Ansan, Gyeonggido, Republic of Korea
| | - Hyun Kim
- Department of Anatomy, Korea University College of Medicine, Brain Korea 21 plus, Seoul, 02841, Republic of Korea
| | | | - Jae Ryun Ryu
- Department of Anatomy, Korea University College of Medicine, Brain Korea 21 plus, Seoul, 02841, Republic of Korea
| | - Woong Sun
- Department of Anatomy, Korea University College of Medicine, Brain Korea 21 plus, Seoul, 02841, Republic of Korea.
| |
Collapse
|
23
|
Palese F, Pontis S, Realini N, Piomelli D. NAPE-specific phospholipase D regulates LRRK2 association with neuronal membranes. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 90:217-238. [PMID: 33706934 DOI: 10.1016/bs.apha.2020.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
N-acylphosphatidylethanolamines (NAPEs) are glycerophospholipid precursors for bioactive lipid amides and potential regulators of membrane function. They are hydrolyzed by NAPE-specific phospholipase D (NAPE-PLD) and have been implicated in neurodegenerative disorders such as Parkinson's disease. Here, we used siRNA-mediated silencing of NAPE-PLD in human SH-SY5Y cells and NAPE-PLD-/- mice to determine whether NAPEs influence the membrane association of LRRK2, a multifunctional protein kinase that is frequently mutated in persons with sporadic Parkinson's disease. NAPE-PLD deletion caused a significant accumulation of non-metabolized NAPEs, which was accompanied by a shift of LRRK2 from membrane to cytosol and a reduction in total LRRK2 content. Conversely, exposure of intact SH-SY5Y cells to bacterial PLD lowered NAPE levels and enhanced LRRK2 association with membranes. The results suggest that NAPE-PLD activity may contribute to the control of LRRK2 localization by regulating membrane NAPE levels.
Collapse
Affiliation(s)
- Francesca Palese
- Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy; Departments of Anatomy and Neurobiology, Pharmacology and Biological Chemistry, University of California, Irvine, CA, United States
| | - Silvia Pontis
- Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Natalia Realini
- Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Daniele Piomelli
- Departments of Anatomy and Neurobiology, Pharmacology and Biological Chemistry, University of California, Irvine, CA, United States.
| |
Collapse
|
24
|
Zhao Y, Keshiya S, Perera G, Schramko L, Halliday GM, Dzamko N. LRRK2 kinase inhibitors reduce alpha-synuclein in human neuronal cell lines with the G2019S mutation. Neurobiol Dis 2020; 144:105049. [PMID: 32800998 DOI: 10.1016/j.nbd.2020.105049] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 07/31/2020] [Accepted: 08/08/2020] [Indexed: 11/28/2022] Open
Abstract
Kinase activating missense mutations in leucine-rich repeat kinase 2 (LRRK2) predispose to Parkinson's disease. Consequently, there is much interest in delineating LRRK2 biology, both in terms of gaining further insight into disease causes, and also determining whether or not LRRK2 is a potential Parkinson's disease therapeutic target. Indeed, many potent and selective small molecule inhibitors of LRRK2 have been developed and are currently being used for pre-clinical testing in cell and animal models. In the current study, we have obtained fibroblasts from four subjects with the common LRRK2 mutation, G2019S. Fibroblasts were reprogrammed to induced pluripotent stem cells and then to neural stem cells and ultimately neurons. Two clones for each of the human neural cell lines were then chronically treated with and without either of two distinct inhibitors of LRRK2 and effects on toxicity and Parkinson's disease related phenotypes were assessed. Cells with the G2019S mutation had a propensity to accumulate the pathological Parkinson's disease protein α-synuclein. Moreover, α-synuclein accumulation in the G2019S cells was significantly reduced with both LRRK2 inhibitors in seven of the eight cell lines studied. LRRK2 inhibitors also improved the nuclear morphology of G2019S cells and impacted on measures of autophagy and endoplasmic reticulum stress. Lastly, we did not find evidence of inhibitor toxicity under the chronic treatment conditions. These results add to evidence that LRRK2 inhibitors may have utility in the treatment of Parkinson's disease via reducing α-synuclein.
Collapse
Affiliation(s)
- Ye Zhao
- Neuroscience Research Australia, Sydney NSW 2031 & School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, Sydney, NSW 2052, Australia; Brain and Mind Centre & Central Clinical School, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Shikara Keshiya
- Brain and Mind Centre & Central Clinical School, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Gayathri Perera
- Brain and Mind Centre & Central Clinical School, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Lauren Schramko
- Neuroscience Research Australia, Sydney NSW 2031 & School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, Sydney, NSW 2052, Australia
| | - Glenda M Halliday
- Neuroscience Research Australia, Sydney NSW 2031 & School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, Sydney, NSW 2052, Australia; Brain and Mind Centre & Central Clinical School, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Nicolas Dzamko
- Neuroscience Research Australia, Sydney NSW 2031 & School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, Sydney, NSW 2052, Australia; Brain and Mind Centre & Central Clinical School, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia.
| |
Collapse
|
25
|
Abstract
Parkinson's disease (PD) is a leading cause of neurodegeneration that is defined by the selective loss of dopaminergic neurons and the accumulation of protein aggregates called Lewy bodies (LBs). The unequivocal identification of Mendelian inherited mutations in 13 genes in PD has provided transforming insights into the pathogenesis of this disease. The mechanistic analysis of several PD genes, including α-synuclein (α-syn), leucine-rich repeat kinase 2 (LRRK2), PTEN-induced kinase 1 (PINK1), and Parkin, has revealed central roles for protein aggregation, mitochondrial damage, and defects in endolysosomal trafficking in PD neurodegeneration. In this review, we outline recent advances in our understanding of these gene pathways with a focus on the emergent role of Rab (Ras analog in brain) GTPases and vesicular trafficking as a common mechanism that underpins how mutations in PD genes lead to neuronal loss. These advances have led to previously distinct genes such as vacuolar protein-sorting-associated protein 35 (VPS35) and LRRK2 being implicated in a common signaling pathway. A greater understanding of these common nodes of vesicular trafficking will be crucial for linking other PD genes and improving patient stratification in clinical trials underway against α-syn and LRRK2 targets.
Collapse
Affiliation(s)
- Pawan Kishor Singh
- MRC Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom;
| | - Miratul M K Muqit
- MRC Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom;
| |
Collapse
|
26
|
Gypenoside Inhibits Endothelial Cell Apoptosis in Atherosclerosis by Modulating Mitochondria through PI3K/Akt/Bad Pathway. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2819658. [PMID: 32685460 PMCID: PMC7327587 DOI: 10.1155/2020/2819658] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/13/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022]
Abstract
Atherosclerosis remains the most common cause of deaths worldwide. Endothelial cell apoptosis is an important process in the progress of atherosclerosis, as it can cause the endothelium to lose their capability in regulating the lipid homeostasis, inflammation, and immunity. Endothelial cell injury can disrupt the integrity and barrier function of an endothelium and facilitate lipid deposition, leading to atherogenesis. Chinese medicine techniques for preventing and treating atherosclerosis are gaining attention, especially natural products. In this study, we demonstrated that gypenoside could decrease the levels of serum lipid, alleviate the formation of atherosclerotic plaque, and lessen aortic intima thickening. Gypenoside potentially activates the PI3K/Akt/Bad signal pathway to modulate the apoptosis-related protein expression in the aorta. Moreover, gypenoside downregulated mitochondrial fission and fusion proteins, mitochondrial energy-related proteins in the mouse aorta. In conclusion, this study demonstrated a new function of gypenoside in endothelial apoptosis and suggested a therapeutic potential of gypenoside in atherosclerosis associated with apoptosis by modulating mitochondrial function through the PI3K/Akt/Bad pathway.
Collapse
|
27
|
Obergasteiger J, Frapporti G, Lamonaca G, Pizzi S, Picard A, Lavdas AA, Pischedda F, Piccoli G, Hilfiker S, Lobbestael E, Baekelandt V, Hicks AA, Corti C, Pramstaller PP, Volta M. Kinase inhibition of G2019S-LRRK2 enhances autolysosome formation and function to reduce endogenous alpha-synuclein intracellular inclusions. Cell Death Discov 2020; 6:45. [PMID: 32550012 PMCID: PMC7280235 DOI: 10.1038/s41420-020-0279-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/07/2020] [Accepted: 05/16/2020] [Indexed: 12/15/2022] Open
Abstract
The Parkinson's disease (PD)-associated kinase Leucine-Rich Repeat Kinase 2 (LRRK2) is a crucial modulator of the autophagy-lysosome pathway, but unclarity exists on the precise mechanics of its role and the direction of this modulation. In particular, LRRK2 is involved in the degradation of pathological alpha-synuclein, with pathogenic mutations precipitating neuropathology in cellular and animal models of PD, and a significant proportion of LRRK2 patients presenting Lewy neuropathology. Defects in autophagic processing and lysosomal degradation of alpha-synuclein have been postulated to underlie its accumulation and onset of neuropathology. Thus, it is critical to obtain a comprehensive knowledge on LRRK2-associated pathology. Here, we investigated a G2019S-LRRK2 recombinant cell line exhibiting accumulation of endogenous, phosphorylated alpha-synuclein. We found that G2019S-LRRK2 leads to accumulation of LC3 and abnormalities in lysosome morphology and proteolytic activity in a kinase-dependent fashion, but independent from constitutively active Rab10. Notably, LRRK2 inhibition was ineffective upon upstream blockade of autophagosome-lysosome fusion events, highlighting this step as critical for alpha-synuclein clearance.
Collapse
Affiliation(s)
- Julia Obergasteiger
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck - Via Galvani 31, 39100 Bolzano, Italy
| | - Giulia Frapporti
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck - Via Galvani 31, 39100 Bolzano, Italy
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Via Sommarive 9, 38123 Povo, TN Italy
| | - Giulia Lamonaca
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck - Via Galvani 31, 39100 Bolzano, Italy
| | - Sara Pizzi
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck - Via Galvani 31, 39100 Bolzano, Italy
| | - Anne Picard
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck - Via Galvani 31, 39100 Bolzano, Italy
| | - Alexandros A. Lavdas
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck - Via Galvani 31, 39100 Bolzano, Italy
| | - Francesca Pischedda
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Via Sommarive 9, 38123 Povo, TN Italy
| | - Giovanni Piccoli
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Via Sommarive 9, 38123 Povo, TN Italy
| | - Sabine Hilfiker
- Department of Anesthesiology, Rutgers University - New Jersey Medical School, Medical Science Building, 185 South Orange Avenue, Newark, NJ 07103 USA
| | - Evy Lobbestael
- Department of Neurosciences, KU Leuven, Herestraat 49 bus 1023, 3000 Leuven, Belgium
| | - Veerle Baekelandt
- Department of Neurosciences, KU Leuven, Herestraat 49 bus 1023, 3000 Leuven, Belgium
| | - Andrew A. Hicks
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck - Via Galvani 31, 39100 Bolzano, Italy
| | - Corrado Corti
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck - Via Galvani 31, 39100 Bolzano, Italy
| | - Peter P. Pramstaller
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck - Via Galvani 31, 39100 Bolzano, Italy
- Department of Neurology, General Central Hospital, Via Böhler 5, 39100 Bolzano, Italy
- Department of Neurology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Mattia Volta
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck - Via Galvani 31, 39100 Bolzano, Italy
| |
Collapse
|
28
|
Madureira M, Connor-Robson N, Wade-Martins R. "LRRK2: Autophagy and Lysosomal Activity". Front Neurosci 2020; 14:498. [PMID: 32523507 PMCID: PMC7262160 DOI: 10.3389/fnins.2020.00498] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/21/2020] [Indexed: 01/07/2023] Open
Abstract
It has been 15 years since the Leucine-rich repeat kinase 2 (LRRK2) gene was identified as the most common genetic cause for Parkinson's disease (PD). The two most common mutations are the LRRK2-G2019S, located in the kinase domain, and the LRRK2-R1441C, located in the ROC-COR domain. While the LRRK2-G2019S mutation is associated with increased kinase activity, the LRRK2-R1441C exhibits a decreased GTPase activity and altered kinase activity. Multiple lines of evidence have linked the LRRK2 protein with a role in the autophagy pathway and with lysosomal activity in neurons. Neurons rely heavily on autophagy to recycle proteins and process cellular waste due to their post-mitotic state. Additionally, lysosomal activity decreases with age which can potentiate the accumulation of α-synuclein, the pathological hallmark of PD, and subsequently lead to the build-up of Lewy bodies (LBs) observed in this disorder. This review provides an up to date summary of the LRRK2 field to understand its physiological role in the autophagy pathway in neurons and related cells. Careful assessment of how LRRK2 participates in the regulation of phagophore and autophagosome formation, autophagosome and lysosome fusion, lysosomal maturation, maintenance of lysosomal pH and calcium levels, and lysosomal protein degradation are addressed. The autophagy pathway is a complex cellular process and due to the variety of LRRK2 models studied in the field, associated phenotypes have been reported to be seemingly conflicting. This review provides an in-depth discussion of different models to assess the normal and disease-associated role of the LRRK2 protein on autophagic function. Given the importance of the autophagy pathway in Parkinson's pathogenesis it is particularly relevant to focus on the role of LRRK2 to discover novel therapeutic approaches that restore lysosomal protein degradation homeostasis.
Collapse
Affiliation(s)
- Marta Madureira
- Department of Physiology, Anatomy and Genetics, Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Graduate Program in Areas of Basic and Applied Biology (GABBA), Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Natalie Connor-Robson
- Department of Physiology, Anatomy and Genetics, Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
| | - Richard Wade-Martins
- Department of Physiology, Anatomy and Genetics, Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
29
|
Xiang H, Zhang J, Lin C, Zhang L, Liu B, Ouyang L. Targeting autophagy-related protein kinases for potential therapeutic purpose. Acta Pharm Sin B 2020; 10:569-581. [PMID: 32322463 PMCID: PMC7161711 DOI: 10.1016/j.apsb.2019.10.003] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/06/2019] [Accepted: 09/09/2019] [Indexed: 02/08/2023] Open
Abstract
Autophagy, defined as a scavenging process of protein aggregates and damaged organelles mediated by lysosomes, plays a significant role in the quality control of macromolecules and organelles. Since protein kinases are integral to the autophagy process, it is critically important to understand the role of kinases in autophagic regulation. At present, intervention of autophagic processes by small-molecule modulators targeting specific kinases has becoming a reasonable and prevalent strategy for treating several varieties of human disease, especially cancer. In this review, we describe the role of some autophagy-related kinase targets and kinase-mediated phosphorylation mechanisms in autophagy regulation. We also summarize the small-molecule kinase inhibitors/activators of these targets, highlighting the opportunities of these new therapeutic agents.
Collapse
Key Words
- 4E-BP1, eukaryotic translation initiation factor 4E-binding protein
- AKT1, AKT serine/threonine kinase 1
- AMBRA1, autophagy/beclin-1 regulator 1
- AMPK, AMP-activated protein kinase
- ARF, auxin response factor gene
- ATG, autophagy-related protein
- Autophagy
- Autophagy-related kinase
- CaMKK2, calcium/calmodulin-dependent protein kinase kinase 2
- DAPK, death associated protein kinase
- FIP200, FAK family kinase-interacting protein of 200 kDa
- GAP, GTPase-activating protein
- GO, gene ontology
- GSK3α, glycogen synthase kinase 3 alpha
- HMGB1, high mobility group protein B1
- Human disease therapy
- JNK1, C-Jun N-terminal kinase
- LC3, microtubule-associated protein 1 light chain 3
- LKB1, serine/threonine-protein kinase stk11
- LPS, lipopolysaccharide
- LRRK2, leucine rich repeat kinase 2
- PD, Parkinson's disease
- PI, phosphatidylinositol
- PI3 kinase, phosphoinositide 3-kinase
- PI3P, phosphatidylinositol triphosphate
- PIM2, proviral insertion in murine lymphomas 2
- PINK1, PTEN-induced putative kinase 1
- PIP2, phosphatidylinositol-4,5-bisphosphate
- PKACα, a protein kinase cAMP-activated catalytic subunit alpha
- PKCα, protein kinase C alpha type
- PKD1, polycystin-1
- PPIs, protein–protein interactions
- PROTAC, proteolysis targeting chimeras
- PTMs, post-translational modifications
- Phosphorylation
- Protein kinases
- Rheb, the RAS homolog enriched in brain
- Small-molecule kinase inhibitors/activators
- TAK1, transforming growth factor activated kinase-1
- TFEB, transcription factor EB
- TNBC, triple-negative breast cancer
- TSC1/2, tuberous sclerosis complex proteins 1/2
- ULK complex, ULK1–mATG13–FIP200–ATG101 complex
- ULK1, unc-51-like kinase 1
- UVRAG, ultraviolet resistance-associated gene
- mTOR, mammalian target of rapamycin
- mTORC1, mammalian target of rapamycin complex 1
Collapse
Affiliation(s)
- Honggang Xiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Jifa Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Congcong Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lan Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
| |
Collapse
|
30
|
LRRK2 regulation of immune-pathways and inflammatory disease. Biochem Soc Trans 2020; 47:1581-1595. [PMID: 31769472 PMCID: PMC6925522 DOI: 10.1042/bst20180463] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022]
Abstract
Mutations in the leucine-rich-repeat kinase 2 (LRRK2) gene are associated with familial and sporadic cases of Parkinson's disease but are also found in immune-related disorders such as inflammatory bowel disease, tuberculosis and leprosy. LRRK2 is highly expressed in immune cells and has been functionally linked to pathways pertinent to immune cell function, such as cytokine release, autophagy and phagocytosis. Here, we examine the current understanding of the role of LRRK2 kinase activity in pathway regulation in immune cells, drawing upon data from multiple diseases associated with LRRK2 to highlight the pleiotropic effects of LRRK2 in different cell types. We discuss the role of the bona fide LRRK2 substrate, Rab GTPases, in LRRK2 pathway regulation as well as downstream events in the autophagy and inflammatory pathways.
Collapse
|
31
|
Feng ST, Wang ZZ, Yuan YH, Sun HM, Chen NH, Zhang Y. Update on the association between alpha-synuclein and tau with mitochondrial dysfunction: Implications for Parkinson's disease. Eur J Neurosci 2020; 53:2946-2959. [PMID: 32031280 DOI: 10.1111/ejn.14699] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 01/13/2020] [Accepted: 01/23/2020] [Indexed: 12/26/2022]
Abstract
The critical role of mitochondrial dysfunction in the pathological mechanisms of neurodegenerative disorders, particularly Parkinson's disease (PD), is well established. Compelling evidence indicates that Parkinson's proteins (e.g., α-synuclein, Parkin, PINK1, DJ-1, and LRRK2) are associated with mitochondrial dysfunction and oxidative stress in PD. Significantly, there is a possible central role of alpha-synuclein (α-Syn) in the occurrence of mitochondrial dysfunction and oxidative stress by the mediation of different signaling pathways. Also, tau, traditionally considered as the main component of neurofibrillary tangles, aggregates and amplifies the neurotoxic effects on mitochondria by interacting with α-Syn. Moreover, oxidative stress caused by mitochondrial dysfunction favors assembly of both α-Syn and tau and also plays a key role in the formation of protein aggregates. In this review, we provide an overview of the relationship between these two pathological proteins and mitochondrial dysfunction in PD, and also summarize the underlying mechanisms in the interplay of α-Syn aggregation and phosphorylated tau targeting the mitochondria, to find new strategies to prevent PD processing.
Collapse
Affiliation(s)
- Si-Tong Feng
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhen-Zhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu-He Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong-Mei Sun
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
32
|
Bonello F, Hassoun SM, Mouton-Liger F, Shin YS, Muscat A, Tesson C, Lesage S, Beart PM, Brice A, Krupp J, Corvol JC, Corti O. LRRK2 impairs PINK1/Parkin-dependent mitophagy via its kinase activity: pathologic insights into Parkinson's disease. Hum Mol Genet 2020; 28:1645-1660. [PMID: 30629163 DOI: 10.1093/hmg/ddz004] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/05/2018] [Accepted: 01/01/2019] [Indexed: 11/12/2022] Open
Abstract
Mutations of LRRK2, encoding leucine-rich repeat kinase 2 (LRRK2), are the leading cause of autosomal dominant Parkinson's disease (PD). The most frequent of these mutations, G2019S substitution, increases kinase activity, but it remains unclear how it causes PD. Recent studies suggest that LRRK2 modulates mitochondrial homeostasis. Mitochondrial dysfunction plays a key role in the pathogenesis of autosomal recessive PD forms linked to PARK2 and PINK1, encoding the cytosolic E3 ubiquitin-protein ligase Parkin and the mitochondrial kinase PINK1, which jointly regulate mitophagy. We explored the role of LRRK2 and its kinase activity in PINK1/Parkin-dependent mitophagy. LRRK2 increased mitochondrial aggregation and attenuated mitochondrial clearance in cells coexpressing Parkin and exposed to the protonophore carbonylcyanide m-chlorophenylhydrazone. Förster resonance energy transfer imaging microscopy showed that LRRK2 impaired the interactions between Parkin and Drp1 and their mitochondrial targets early in mitophagy. The inhibition of LRRK2 kinase activity by a 'kinase-dead' LRRK2 mutation or with a pharmacological inhibitor (LRRK2-IN-1) restored these interactions. The monitoring of mitophagy in human primary fibroblasts with the novel dual-fluorescence mtRosella reporter and a new hypothermic shock paradigm revealed similar defects in PD patients with the G2019S LRRK2 substitution or PARK2 mutations relative to healthy subjects. This defect was restored by LRRK2-IN-1 treatment in LRRK2 patients only. Our results suggest that PD forms due to LRRK2 and PARK2 mutations involve pathogenic mechanisms converging on PINK1/Parkin-dependent mitophagy.
Collapse
Affiliation(s)
- Fiona Bonello
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France
| | - Sidi-Mohamed Hassoun
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France
| | - François Mouton-Liger
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France
| | - Yea Seul Shin
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Adeline Muscat
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France
| | - Christelle Tesson
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France
| | - Suzanne Lesage
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France
| | - Philip M Beart
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Alexis Brice
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France.,AP-HP, Hôpital de la Pitié-Salpêtrière, Department of Genetics, Paris, France
| | | | - Jean-Christophe Corvol
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France.,AP-HP, Hôpital de la Pitié-Salpêtrière, Clinical Investigation Center for Neurology (CIC), Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Olga Corti
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France
| |
Collapse
|
33
|
Cogo S, Manzoni C, Lewis PA, Greggio E. Leucine-rich repeat kinase 2 and lysosomal dyshomeostasis in Parkinson disease. J Neurochem 2020; 152:273-283. [PMID: 31693760 DOI: 10.1111/jnc.14908] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/26/2019] [Accepted: 10/31/2019] [Indexed: 12/24/2022]
Abstract
Over the last two decades, a number of studies have underlined the importance of lysosomal-based degradative pathways in maintaining the homeostasis of post-mitotic cells, and revealed the remarkable contribution of a functional autophagic machinery in the promotion of longevity. In contrast, defects in the clearance of organelles and aberrant protein aggregates have been linked to accelerated neuronal loss and neurological dysfunction. Several neurodegenerative disorders, among which Alzheimer disease (AD), Frontotemporal dementia, and Amyotrophic Lateral Sclerosis to name a few, are associated with alterations of the autophagy and endo-lysosomal pathways. In Parkinson disease (PD), the most prevalent genetic determinant, Leucine-rich repeat kinase 2 (LRRK2), is believed to be involved in the regulation of intracellular vesicle traffic, autophagy and lysosomal function. Here, we review the current understanding of the mechanisms by which LRRK2 regulates lysosomal-based degradative pathways in neuronal and non-neuronal cells and discuss the impact of pathogenic PD mutations in contributing to lysosomal dyshomeostasis.
Collapse
Affiliation(s)
- Susanna Cogo
- Department of Biology, University of Padova, Padova, Italy
| | - Claudia Manzoni
- School of Pharmacy, University of Reading, Reading, UK
- Department of Neurodegenerative Diseases, University College London, London, UK
| | - Patrick A Lewis
- School of Pharmacy, University of Reading, Reading, UK
- Department of Neurodegenerative Diseases, University College London, London, UK
| | - Elisa Greggio
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
34
|
Stott SRW, Wyse RK, Brundin P. Novel approaches to counter protein aggregation pathology in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2020; 252:451-492. [PMID: 32247372 PMCID: PMC10019778 DOI: 10.1016/bs.pbr.2019.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The primary neuropathological characteristics of the Parkinsonian brain are the loss of nigral dopamine neurons and the aggregation of alpha synuclein protein. Efforts to development potentially disease-modifying treatments have largely focused on correcting these aspects of the condition. In the last decade treatments targeting protein aggregation have entered the clinical pipeline. In this chapter we provide an overview of ongoing clinical trial programs for different therapies attempting to reduce protein aggregation pathology in Parkinson's disease. We will also briefly consider various novel approaches being proposed-and being developed preclinically-to inhibit/reduce aggregated protein pathology in Parkinson's.
Collapse
Affiliation(s)
| | | | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States.
| |
Collapse
|
35
|
Feng ST, Wang ZZ, Yuan YH, Wang XL, Sun HM, Chen NH, Zhang Y. Dynamin-related protein 1: A protein critical for mitochondrial fission, mitophagy, and neuronal death in Parkinson’s disease. Pharmacol Res 2020; 151:104553. [DOI: 10.1016/j.phrs.2019.104553] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/14/2019] [Accepted: 11/16/2019] [Indexed: 01/14/2023]
|
36
|
Berwick DC, Heaton GR, Azeggagh S, Harvey K. LRRK2 Biology from structure to dysfunction: research progresses, but the themes remain the same. Mol Neurodegener 2019; 14:49. [PMID: 31864390 PMCID: PMC6925518 DOI: 10.1186/s13024-019-0344-2] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
Since the discovery of leucine-rich repeat kinase 2 (LRRK2) as a protein that is likely central to the aetiology of Parkinson’s disease, a considerable amount of work has gone into uncovering its basic cellular function. This effort has led to the implication of LRRK2 in a bewildering range of cell biological processes and pathways, and probable roles in a number of seemingly unrelated medical conditions. In this review we summarise current knowledge of the basic biochemistry and cellular function of LRRK2. Topics covered include the identification of phosphorylation substrates of LRRK2 kinase activity, in particular Rab proteins, and advances in understanding the activation of LRRK2 kinase activity via dimerisation and association with membranes, especially via interaction with Rab29. We also discuss biochemical studies that shed light on the complex LRRK2 GTPase activity, evidence of roles for LRRK2 in a range of cell signalling pathways that are likely cell type specific, and studies linking LRRK2 to the cell biology of organelles. The latter includes the involvement of LRRK2 in autophagy, endocytosis, and processes at the trans-Golgi network, the endoplasmic reticulum and also key microtubule-based cellular structures. We further propose a mechanism linking LRRK2 dimerisation, GTPase function and membrane recruitment with LRRK2 kinase activation by Rab29. Together these data paint a picture of a research field that in many ways is moving forward with great momentum, but in other ways has not changed fundamentally. Many key advances have been made, but very often they seem to lead back to the same places.
Collapse
Affiliation(s)
- Daniel C Berwick
- School of Health, Life and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK.
| | - George R Heaton
- Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Sonia Azeggagh
- School of Health, Life and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Kirsten Harvey
- Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK.
| |
Collapse
|
37
|
Albanese F, Novello S, Morari M. Autophagy and LRRK2 in the Aging Brain. Front Neurosci 2019; 13:1352. [PMID: 31920513 PMCID: PMC6928047 DOI: 10.3389/fnins.2019.01352] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/02/2019] [Indexed: 12/20/2022] Open
Abstract
Autophagy is a highly conserved process by which long-lived macromolecules, protein aggregates and dysfunctional/damaged organelles are delivered to lysosomes for degradation. Autophagy plays a crucial role in regulating protein quality control and cell homeostasis in response to energetic needs and environmental challenges. Indeed, activation of autophagy increases the life-span of living organisms, and impairment of autophagy is associated with several human disorders, among which neurodegenerative disorders of aging, such as Parkinson’s disease. These disorders are characterized by the accumulation of aggregates of aberrant or misfolded proteins that are toxic for neurons. Since aging is associated with impaired autophagy, autophagy inducers have been viewed as a strategy to counteract the age-related physiological decline in brain functions and emergence of neurodegenerative disorders. Parkinson’s disease is a hypokinetic, multisystemic disorder characterized by age-related, progressive degeneration of central and peripheral neuronal populations, associated with intraneuronal accumulation of proteinaceous aggregates mainly composed by the presynaptic protein α-synuclein. α-synuclein is a substrate of macroautophagy and chaperone-mediated autophagy (two major forms of autophagy), thus impairment of its clearance might favor the process of α-synuclein seeding and spreading that trigger and sustain the progression of this disorder. Genetic factors causing Parkinson’s disease have been identified, among which mutations in the LRRK2 gene, which encodes for a multidomain protein encompassing central GTPase and kinase domains, surrounded by protein-protein interaction domains. Six LRRK2 mutations have been pathogenically linked to Parkinson’s disease, the most frequent being the G2019S in the kinase domain. LRRK2-associated Parkinson’s disease is clinically and neuropathologically similar to idiopathic Parkinson’s disease, also showing age-dependency and incomplete penetrance. Several mechanisms have been proposed through which LRRK2 mutations can lead to Parkinson’s disease. The present article will focus on the evidence that LRRK2 and its mutants are associated with autophagy dysregulation. Studies in cell lines and neurons in vitro and in LRRK2 knock-out, knock-in, kinase-dead and transgenic animals in vivo will be reviewed. The role of aging in LRRK2-induced synucleinopathy will be discussed. Possible mechanisms underlying the LRRK2-mediated control over autophagy will be analyzed, and the contribution of autophagy dysregulation to the neurotoxic actions of LRRK2 will be examined.
Collapse
Affiliation(s)
- Federica Albanese
- Section of Pharmacology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Salvatore Novello
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Michele Morari
- Section of Pharmacology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
38
|
Vidyadhara DJ, Lee JE, Chandra SS. Role of the endolysosomal system in Parkinson's disease. J Neurochem 2019; 150:487-506. [PMID: 31287913 PMCID: PMC6707858 DOI: 10.1111/jnc.14820] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/01/2019] [Accepted: 07/03/2019] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders, affecting 1-1.5% of the total population. While progress has been made in understanding the neurodegenerative mechanisms that lead to cell death in late stages of PD, mechanisms for early, causal pathogenic events are still elusive. Recent developments in PD genetics increasingly point at endolysosomal (E-L) system dysfunction as the early pathomechanism and key pathway affected in PD. Clathrin-mediated synaptic endocytosis, an integral part of the neuronal E-L system, is probably the main early target as evident in auxilin, RME-8, and synaptojanin-1 mutations that cause PD. Autophagy, another important pathway in the E-L system, is crucial in maintaining proteostasis and a healthy mitochondrial pool, especially in neurons considering their inability to divide and requirement to function an entire life-time. PINK1 and Parkin mutations severely perturb autophagy of dysfunctional mitochondria (mitophagy), both in the cell body and synaptic terminals of dopaminergic neurons, leading to PD. Endolysosomal sorting and trafficking is also crucial, which is complex in multi-compartmentalized neurons. VPS35 and VPS13C mutations noted in PD target these mechanisms. Mutations in GBA comprise the most common risk factor for PD and initiate pathology by compromising lysosomal function. This is also the case for ATP13A2 mutations. Interestingly, α-synuclein and LRRK2, key proteins involved in PD, function in different steps of the E-L pathway and target their components to induce disease pathogenesis. In this review, we discuss these E-L system genes that are linked to PD and how their dysfunction results in PD pathogenesis. This article is part of the Special Issue "Synuclein".
Collapse
Affiliation(s)
- D J Vidyadhara
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - John E Lee
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sreeganga S Chandra
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
39
|
Zilocchi M, Fasano M, Alberio T. Mitochondrial Proteins in the Development of Parkinson’s Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1158:17-44. [DOI: 10.1007/978-981-13-8367-0_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
40
|
Zhang X, Huang W, Fan Y, Sun Y, Ge X. Role of GTPases in the regulation of mitochondrial dynamics in Parkinson's disease. Exp Cell Res 2019; 382:111460. [PMID: 31194975 DOI: 10.1016/j.yexcr.2019.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/27/2019] [Accepted: 06/08/2019] [Indexed: 12/11/2022]
Abstract
Mitochondria are highly dynamic organelle that undergo frequent fusion and division, and the balance of these opposing processes regulates mitochondrial morphology, distribution, and function. Mitochondrial fission facilitates the replication and distribution of mitochondria during cell division, whereas the fusion process including inner and outer mitochondrial membrane fusion allows the exchange of intramitochondrial material between adjacent mitochondria. Despite several GTPase family proteins have been implicated as key modulators of mitochondrial dynamics, the mechanisms by which these proteins regulate mitochondrial homeostasis and function remain not clearly understood. Neuronal function and survival are closely related to mitochondria dynamics, and disturbed mitochondrial fission/fusion may influence neurotransmission, synaptic maintenance, neuronal survival and function. Recent studies have shown that mitochondrial dysfunction caused by aberrant mitochondrial dynamics plays an essential role in the pathogenesis of both sporadic and familial Parkinson's disease (PD). Collectively, we review the molecular mechanism of known GTPase proteins in regulating mitochondrial fission and fusion, but also highlight the causal role for mitochondrial dynamics in PD pathogenesis.
Collapse
Affiliation(s)
- Xiaoling Zhang
- Department of Pharmacology, Medical College, Yangzhou University, Yangzhou, 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Wenmin Huang
- Department of Pharmacology, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Yiyun Fan
- Department of Pharmacology, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Ying Sun
- Department of Pharmacology, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Xiaoqun Ge
- Department of Pharmacology, Medical College, Yangzhou University, Yangzhou, 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| |
Collapse
|
41
|
Kluss JH, Mamais A, Cookson MR. LRRK2 links genetic and sporadic Parkinson's disease. Biochem Soc Trans 2019; 47:651-661. [PMID: 30837320 PMCID: PMC6563926 DOI: 10.1042/bst20180462] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/05/2019] [Accepted: 02/07/2019] [Indexed: 12/21/2022]
Abstract
The past two decades in research has revealed the importance of leucine-rich repeat kinase 2 (LRRK2) in both monogenic and sporadic forms of Parkinson's disease (PD). In families, mutations in LRRK2 can cause PD with age-dependent but variable penetrance and genome-wide association studies have found variants of the gene that are risk factors for sporadic PD. Functional studies have suggested that the common mechanism that links all disease-associated variants is that they increase LRRK2 kinase activity, albeit in different ways. Here, we will discuss the roles of LRRK2 in areas of inflammation and vesicular trafficking in the context of monogenic and sporadic PD. We will also provide a hypothetical model that links inflammation and vesicular trafficking together in an effort to outline how these pathways might interact and eventually lead to neuronal cell death. We will also highlight the translational potential of LRRK2-specific kinase inhibitors for the treatment of PD.
Collapse
Affiliation(s)
- Jillian H Kluss
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707, U.S.A
| | - Adamantios Mamais
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707, U.S.A
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707, U.S.A.
| |
Collapse
|
42
|
Lee W, Kim SH. Autophagy at synapses in neurodegenerative diseases. Arch Pharm Res 2019; 42:407-415. [DOI: 10.1007/s12272-019-01148-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/21/2019] [Indexed: 12/31/2022]
|
43
|
Ho DH, Lee H, Son I, Seol W. G2019s LRRK2 promotes mitochondrial fission and increases TNFα-mediated neuroinflammation responses. Anim Cells Syst (Seoul) 2019; 23:106-111. [PMID: 30949397 PMCID: PMC6440522 DOI: 10.1080/19768354.2019.1585948] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/31/2019] [Accepted: 02/18/2019] [Indexed: 11/02/2022] Open
Abstract
Leucine rich-repeat kinase 2 (LRRK2) is involved in the pathogenesis of Parkinson's disease (PD). LRRK2 has kinase and GTPase activities, and mediates several cell functions, including vesicle trafficking, apoptosis, autophagy, mitochondrial dynamics, and neuroinflammation. G2019S (GS) is the most prevalent mutation of LRRK2. The mutation increases kinase activity, suggesting that this activity is crucial for PD pathogenesis. The activation and inhibition of LRRK2 kinase increases and reduces the levels of proinflammatory cytokines, respectively suggesting that the role of LRRK2 in neuroinflammation is critical for the pathology of PD. Previously, we demonstrated that microglial activation by lipopolysaccharide (LPS) increases mitochondrial fission via the activation of LRRK2 kinase, while LRRK2 kinase inhibition diminishes the fission morphology and release of tumor necrosis factor-alpha (TNFα) in BV2 or rat primary microglia and the brains of GS transgenic mice. In this study, the ectopic expression of GS LRRK2 in BV2 cells significantly elevated the expression of Drp1 along the fragmented mitochondria and decreased mitochondria size compared with controls. GS LRRK2-transfected BV2 cells displayed significantly increased TNFα release and neuronal death. Inhibition of LRRK2 kinase alleviated these features. TNFα levels in brains of GS mice were significantly increased compared to those in their littermates. These data further support our previous findings concerning LPS-induced neuroinflammation and mitochondrial fission in microglia via LRRK2 kinase activation.
Collapse
Affiliation(s)
- Dong Hwan Ho
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo-si, Republic of Korea
| | - Heajin Lee
- Electron Microscopy Research Center, Korea Basic Science Institute (KBSI), Daejeon-si, Republic of Korea
| | - Ilhong Son
- Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo-si, Republic of Korea
| | - Wongi Seol
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo-si, Republic of Korea
| |
Collapse
|
44
|
Cresto N, Gardier C, Gubinelli F, Gaillard MC, Liot G, West AB, Brouillet E. The unlikely partnership between LRRK2 and α-synuclein in Parkinson's disease. Eur J Neurosci 2019; 49:339-363. [PMID: 30269383 PMCID: PMC6391223 DOI: 10.1111/ejn.14182] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 09/11/2018] [Accepted: 09/17/2018] [Indexed: 12/19/2022]
Abstract
Our understanding of the mechanisms underlying Parkinson's disease, the once archetypical nongenetic neurogenerative disorder, has dramatically increased with the identification of α-synuclein and LRRK2 pathogenic mutations. While α-synuclein protein composes the aggregates that can spread through much of the brain in disease, LRRK2 encodes a multidomain dual-enzyme distinct from any other protein linked to neurodegeneration. In this review, we discuss emergent datasets from multiple model systems that suggest these unlikely partners do interact in important ways in disease, both within cells that express both LRRK2 and α-synuclein as well as through more indirect pathways that might involve neuroinflammation. Although the link between LRRK2 and disease can be understood in part through LRRK2 kinase activity (phosphotransferase activity), α-synuclein toxicity is multilayered and plausibly interacts with LRRK2 kinase activity in several ways. We discuss common protein interactors like 14-3-3s that may regulate α-synuclein and LRRK2 in disease. Finally, we examine cellular pathways and outcomes common to both mutant α-synuclein expression and LRRK2 activity and points of intersection. Understanding the interplay between these two unlikely partners in disease may provide new therapeutic avenues for PD.
Collapse
Affiliation(s)
- Noémie Cresto
- Neurodegenerative Diseases Laboratory, UMR9199, CEA, CNRS, Université Paris Sud, Université Paris-Saclay, and MIRCen (Molecular Imaging Research Centre), Institut François Jacob, DRF, CEA, Fontenay-aux-Roses, France
| | - Camille Gardier
- Neurodegenerative Diseases Laboratory, UMR9199, CEA, CNRS, Université Paris Sud, Université Paris-Saclay, and MIRCen (Molecular Imaging Research Centre), Institut François Jacob, DRF, CEA, Fontenay-aux-Roses, France
| | - Francesco Gubinelli
- Neurodegenerative Diseases Laboratory, UMR9199, CEA, CNRS, Université Paris Sud, Université Paris-Saclay, and MIRCen (Molecular Imaging Research Centre), Institut François Jacob, DRF, CEA, Fontenay-aux-Roses, France
| | - Marie-Claude Gaillard
- Neurodegenerative Diseases Laboratory, UMR9199, CEA, CNRS, Université Paris Sud, Université Paris-Saclay, and MIRCen (Molecular Imaging Research Centre), Institut François Jacob, DRF, CEA, Fontenay-aux-Roses, France
| | - Géraldine Liot
- Neurodegenerative Diseases Laboratory, UMR9199, CEA, CNRS, Université Paris Sud, Université Paris-Saclay, and MIRCen (Molecular Imaging Research Centre), Institut François Jacob, DRF, CEA, Fontenay-aux-Roses, France
| | - Andrew B. West
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama, United States 35294
| | - Emmanuel Brouillet
- Neurodegenerative Diseases Laboratory, UMR9199, CEA, CNRS, Université Paris Sud, Université Paris-Saclay, and MIRCen (Molecular Imaging Research Centre), Institut François Jacob, DRF, CEA, Fontenay-aux-Roses, France
| |
Collapse
|
45
|
Singh A, Zhi L, Zhang H. LRRK2 and mitochondria: Recent advances and current views. Brain Res 2019; 1702:96-104. [PMID: 29894679 PMCID: PMC6281802 DOI: 10.1016/j.brainres.2018.06.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/17/2018] [Accepted: 06/07/2018] [Indexed: 12/21/2022]
Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene account for most common causes of familial and sporadic Parkinson's disease (PD) and are one of the strongest genetic risk factors in sporadic PD. Pathways implicated in LRRK2-dependent neurodegeneration include cytoskeletal dynamics, vesicular trafficking, autophagy, mitochondria, and calcium homeostasis. However, the exact molecular mechanisms still need to be elucidated. Both genetic and environmental causes of PD have highlighted the importance of mitochondrial dysfunction in the pathogenesis of PD. Mitochondrial impairment has been observed in fibroblasts and iPSC-derived neural cells from PD patients with LRRK2 mutations, and LRRK2 has been shown to localize to mitochondria and to regulate its function. In this review we discuss recent discoveries relating to LRRK2 mutations and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Alpana Singh
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, United States
| | - Lianteng Zhi
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, United States
| | - Hui Zhang
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, United States.
| |
Collapse
|
46
|
The emerging interrelation between ROCO and related kinases, intracellular Ca 2+ signaling, and autophagy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:1054-1067. [PMID: 30582936 DOI: 10.1016/j.bbamcr.2018.12.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/13/2018] [Accepted: 12/17/2018] [Indexed: 12/12/2022]
Abstract
ROCO kinases form a family of proteins characterized by kinase activity in addition to the presence of the so-called ROC (Ras of complex proteins)/COR (C-terminal of ROC) domains having a role in their GTPase activity. These are the death-associated protein kinase (DAPK) 1 and the leucine-rich repeat kinases (LRRK) 1 and 2. These kinases all play roles in cellular life and death decisions and in autophagy in particular. Related to the ROCO kinases is DAPK 2 that however cannot be classified as a ROCO protein due to the absence of the ROC/COR domains. This review aims to bring together what is known about the relation between these proteins and intracellular Ca2+ signals in the induction and regulation of autophagy. Interestingly, DAPK 1 and 2 and LRRK2 are all linked to Ca2+ signaling in their effects on autophagy, though in various ways. Present evidence supports an upstream role for LRRK2 that via lysosomal and endoplasmic reticulum Ca2+ release can trigger autophagy induction. In contrast herewith, DAPK1 and 2 react on existing Ca2+ signals to stimulate the autophagic pathway. Further research will be needed for obtaining a full understanding of the role of these various kinases in autophagy and to assess their exact relation with intracellular Ca2+ signaling as this would be helpful in the development of novel therapeutic strategies against neurodegenerative disorders, cancer and auto-immune diseases. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
|
47
|
Mamais A, Manzoni C, Nazish I, Arber C, Sonustun B, Wray S, Warner TT, Cookson MR, Lewis PA, Bandopadhyay R. Analysis of macroautophagy related proteins in G2019S LRRK2 Parkinson's disease brains with Lewy body pathology. Brain Res 2018; 1701:75-84. [PMID: 30055128 PMCID: PMC6361106 DOI: 10.1016/j.brainres.2018.07.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/04/2018] [Accepted: 07/24/2018] [Indexed: 12/12/2022]
Abstract
LRRK2, the gene encoding the multidomain kinase Leucine-Rich Repeat Kinase 2 (LRRK2), has been linked to familial and sporadic forms of Parkinson's disease (PD), as well as cancer, leprosy and Crohn's disease, establishing it as a target for discovery therapeutics. LRRK2 has been associated with a range of cellular processes, however its physiological and pathological functions remain unclear. The most prevalent LRRK2 mutations in PD have been shown to affect macroautophagy in various cellular models while a role in autophagy signalling has been recapitulated in vivo. Dysregulation of autophagy has been implicated in PD pathology, and this raises the possibility that differential autophagic activity is relevant to disease progression in PD patients carrying LRRK2 mutations. To examine the relevance of LRRK2 to the regulation of macroautophagy in a disease setting we examined the levels of autophagic markers in the basal ganglia of G2019S LRRK2 PD post-mortem tissue, in comparison to pathology-matched idiopathic PD (iPD), using immunoblotting (IB). Significantly lower levels of p62 and LAMP1 were observed in G2019S LRRK2 PD compared to iPD cases. Similarly, an increase in ULK1 was observed in iPD but was not reflected in G2019S LRRK2 PD cases. Furthermore, examination of p62 by immunohistochemistry (IH) recapitulated a distinct signature for G2019S PD. IH of LAMP1, LC3 and ULK1 broadly correlated with the IB results. Our data from a small but pathologically well-characterized cases highlights a divergence of G2019S PD carriers in terms of autophagic response in alpha-synuclein pathology affected brain regions compared to iPD.
Collapse
Affiliation(s)
- Adamantios Mamais
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, United Kingdom; Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Building 35, 35 Convent Drive, Bethesda, MD 20892-3707, USA.
| | - Claudia Manzoni
- School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AP, United Kingdom; Department of Neurodegenerative Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Iqra Nazish
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, United Kingdom; Department of Clinical and Movement Neuroscience, UCL Institute of Neurology, WC1N 3BG, United Kingdom
| | - Charles Arber
- Department of Neurodegenerative Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Berkiye Sonustun
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, United Kingdom
| | - Selina Wray
- Department of Neurodegenerative Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Thomas T Warner
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, United Kingdom; Department of Clinical and Movement Neuroscience, UCL Institute of Neurology, WC1N 3BG, United Kingdom
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Building 35, 35 Convent Drive, Bethesda, MD 20892-3707, USA
| | - Patrick A Lewis
- School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AP, United Kingdom; Department of Neurodegenerative Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Rina Bandopadhyay
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, United Kingdom; Department of Clinical and Movement Neuroscience, UCL Institute of Neurology, WC1N 3BG, United Kingdom.
| |
Collapse
|
48
|
Ho DH, Kim H, Nam D, Sim H, Kim J, Kim HG, Son I, Seol W. LRRK2 impairs autophagy by mediating phosphorylation of leucyl-tRNA synthetase. Cell Biochem Funct 2018; 36:431-442. [PMID: 30411383 DOI: 10.1002/cbf.3364] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 09/19/2018] [Accepted: 10/06/2018] [Indexed: 02/05/2023]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a causal gene of Parkinson disease. G2019S pathogenic mutation increases its kinase activity. LRRK2 regulates various phenotypes including autophagy, neurite outgrowth, and vesicle trafficking. Leucyl-tRNA synthetase (LRS) attaches leucine to tRNALeu and activates mTORC1. Down-regulation of LRS induces autophagy. We investigated the relationship between LRRK2 and LRS in regulating autophagy and observed interaction between endogenous LRRK2 and LRS proteins and LRS phosphorylation by LRRK2. Mutation studies implicated that T293 in the LRS editing domain was a putative phosphorylation site. Phospho-Thr in LRS was increased in cells overexpressing G2019S and dopaminergic neurons differentiated from induced pluripotent stem (iPS) cells of a G2019S carrier. It was decreased by treatment with an LRRK2 kinase inhibitor (GSK2578215A). Phosphomimetic T293D displayed lower leucine bindings than wild type (WT), suggesting its defective editing function. Cellular expression of T293D increased expression of GRP78/BiP, LC3B-II, and p62 proteins and number of LC3 puncta. Increase of GRP78 and phosphorylated LRS was diminished by treatment with GSK2578215A. Levels of LC3B, GRP78/BiP, p62, and α-synuclein proteins were also increased in G2019S transgenic (TG) mice. These data suggest that LRRK2-mediated LRS phosphorylation impairs autophagy by increasing protein misfolding and endoplasmic reticulum stress mediated by LRS editing defect. SIGNIFICANCE OF THE STUDY: Leucine-rich repeat kinase 2 (LRRK2) is the most common genetic cause of Parkinson disease (PD), and the most prevalent pathogenic mutation, G2019S, increases its kinase activity. In this study, we elucidated that leucyl-tRNA synthetase (LRS) was an LRRK2 kinase substrate and identified T293 as an LRRK2 phosphorylation site. LRRK2-meidated LRS phosphorylation or G2019S can lead to impairment of LRS editing, increased ER stress, and accumulation of autophagy markers. These results demonstrate that LRRK2 kinase activity can facilitate accumulation of misfolded protein, suggesting that LRRK2 kinase might be a potential PD therapeutic target along with previous studies.
Collapse
Affiliation(s)
- Dong Hwan Ho
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo, Republic of Korea
| | - Hyejung Kim
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo, Republic of Korea
| | - Daleum Nam
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo, Republic of Korea
| | - Hyuna Sim
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Janghwan Kim
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Hyung Gun Kim
- Department of Pharmacology, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Ilhong Son
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo, Republic of Korea.,Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo, Republic of Korea
| | - Wongi Seol
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo, Republic of Korea
| |
Collapse
|
49
|
Jang J, Oh H, Nam D, Seol W, Seo MK, Park SW, Kim HG, Seo H, Son I, Ho DH. Increase in anti-apoptotic molecules, nucleolin, and heat shock protein 70, against upregulated LRRK2 kinase activity. Anim Cells Syst (Seoul) 2018; 22:273-280. [PMID: 30460108 PMCID: PMC6171436 DOI: 10.1080/19768354.2018.1518262] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/22/2018] [Accepted: 08/27/2018] [Indexed: 12/30/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is involved in Parkinson’s disease (PD) pathology. A previous study showed that rotenone treatment induced apoptosis, mitochondrial damage, and nucleolar disruption via up-regulated LRRK2 kinase activity, and these effects were rescued by an LRRK2 kinase inhibitor. Heat-shock protein 70 (Hsp70) is an anti-oxidative stress chaperone, and overexpression of Hsp70 enhanced tolerance to rotenone. Nucleolin (NCL) is a component of the nucleolus; overexpression of NCL reduced cellular vulnerability to rotenone. Thus, we hypothesized that rotenone-induced LRRK2 activity would promote changes in neuronal Hsp70 and NCL expressions. Moreover, LRRK2 G2019S, the most prevalent LRRK2 pathogenic mutant with increased kinase activity, could induce changes in Hsp70 and NCL expression. Rotenone treatment of differentiated SH-SY5Y (dSY5Y) cells increased LRKK2 levels and kinase activity, including phospho-S935-LRRK2, phospho-S1292-LRRK2, and the phospho-moesin/moesin ratio, in a dose-dependent manner. Neuronal toxicity and the elevation of cleaved poly (ADP-ribose) polymerase, NCL, and Hsp70 were increased by rotenone. To validate the induction of NCL and Hsp70 expression in response to rotenone, cycloheximide (CHX), a protein synthesis blocker, was administered with rotenone. Post-rotenone increased NCL and Hsp70 expression was repressed by CHX; whereas, rotenone-induced kinase activity and apoptotic toxicity remained unchanged. Transient expression of G2019S in dSY5Y increased the NCL and Hsp70 levels, while administration of a kinase inhibitor diminished these changes. Similar results were observed in rat primary neurons after rotenone treatment or G2019S transfection. Brains from G2019S-transgenic mice also showed increased NCL and Hsp70 levels. Accordingly, LRRK2 kinase inhibition might prevent oxidative stress-mediated PD progression. Abbreviations: 6-OHDA: 6-hydroxydopamine; CHX: cycloheximide; dSY5Y: differentiated SH-SY5Y; g2019S tg: g2019S transgenic mouse; GSK/A-KI: GSK2578215A kinase inhibitor; HSP70: heat shock protein 70; LDH: lactose dehydrogenase; LRRK2: leucine rich-repeat kinase 2; MPTP: 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; myc-GS LRRK2: myc-tagged g2019S LRRK2; NCL: nucleolin; PARP: poly(ADP-ribose) polymerase; PD: Parkinson’s disease; PINK1: PTEN-induced putative kinase 1; pmoesin: phosphorylated moesin at t558; ROS: reactive oxygen species
Collapse
Affiliation(s)
- Jihoon Jang
- Department of Molecular and Life Sciences, Hanyang University, Ansan-si, Republic of Korea.,InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo-si, Republic of Korea
| | - Hakjin Oh
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo-si, Republic of Korea
| | - Daleum Nam
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo-si, Republic of Korea
| | - Wongi Seol
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo-si, Republic of Korea
| | - Mi Kyoung Seo
- Paik Institute for Clinical Research, Inje University College of Medicine, Busan, Republic of Korea
| | - Sung Woo Park
- Paik Institute for Clinical Research, Inje University College of Medicine, Busan, Republic of Korea.,Department of Health Science and Technology, Graduate School of Inje University, Busan, Republic of Korea
| | - Hyung Gun Kim
- Department of Pharmacology, College of Medicine, Dankook University, Cheonan-si, Republic of Korea
| | - Hyemyung Seo
- Department of Molecular and Life Sciences, Hanyang University, Ansan-si, Republic of Korea
| | - Ilhong Son
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo-si, Republic of Korea.,Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo-si, Republic of Korea
| | - Dong Hwan Ho
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo-si, Republic of Korea
| |
Collapse
|
50
|
Joshi AU, Mochly-Rosen D. Mortal engines: Mitochondrial bioenergetics and dysfunction in neurodegenerative diseases. Pharmacol Res 2018; 138:2-15. [PMID: 30144530 DOI: 10.1016/j.phrs.2018.08.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 08/06/2018] [Accepted: 08/13/2018] [Indexed: 12/14/2022]
Abstract
Mitochondria are best known for their role in ATP generation. However, studies over the past two decades have shown that mitochondria do much more than that. Mitochondria regulate both necrotic and apoptotic cell death pathways, they store and therefore coordinate cellular Ca2+ signaling, they generate and metabolize important building blocks, by-products and signaling molecules, and they also generate and are targets of free radical species that modulate many aspects of cell physiology and pathology. Most estimates suggest that although the brain makes up only 2 percent of body weight, utilizes about 20 percent of the body's total ATP. Thus, mitochondrial dysfunction greatly impacts brain functions and is indeed associated with numerous neurodegenerative diseases. Furthermore, a number of abnormal disease-associated proteins have been shown to interact directly with mitochondria, leading to mitochondrial dysfunction and subsequent neuronal cell death. Here, we discuss the role of mitochondrial dynamics impairment in the pathological processes associated with neurodegeneration and suggest that a therapy targeting mitochondrialdysfunction holds a great promise.
Collapse
Affiliation(s)
- Amit U Joshi
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, CA, 94305-5174, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, CA, 94305-5174, USA.
| |
Collapse
|