1
|
Saadh MJ, Faisal A, Adil M, Zabibah RS, Mamadaliev AM, Jawad MJ, Alsaikhan F, Farhood B. Parkinson's Disease and MicroRNAs: A Duel Between Inhibition and Stimulation of Apoptosis in Neuronal Cells. Mol Neurobiol 2024; 61:8552-8574. [PMID: 38520611 DOI: 10.1007/s12035-024-04111-w] [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: 12/14/2022] [Revised: 02/03/2024] [Accepted: 02/27/2024] [Indexed: 03/25/2024]
Abstract
Parkinson's disease (PD) is one of the most prevalent diseases of central nervous system that is caused by degeneration of the substantia nigra's dopamine-producing neurons through apoptosis. Apoptosis is regulated by initiators' and executioners' caspases both in intrinsic and extrinsic pathways, further resulting in neuronal damage. In that context, targeting apoptosis appears as a promising therapeutic approach for treating neurodegenerative diseases. Non-coding RNAs-more especially, microRNAs, or miRNAs-are a promising target for the therapy of neurodegenerative diseases because they are essential for a number of cellular processes, including signaling, apoptosis, cell proliferation, and gene regulation. It is estimated that a substantial portion of coding genes (more than 60%) are regulated by miRNAs. These small regulatory molecules can have wide-reaching consequences on cellular processes like apoptosis, both in terms of intrinsic and extrinsic pathways. Furthermore, it was recommended that a disruption in miRNA expression levels could also result in perturbation of typical apoptosis pathways, which may be a factor in certain diseases like PD. The latest research on miRNAs and their impact on neural cell injury in PD models by regulating the apoptosis pathway is summarized in this review article. Furthermore, the importance of lncRNA/circRNA-miRNA-mRNA network for regulating apoptosis pathways in PD models and treatment is explored. These results can be utilized for developing new strategies in PD treatment.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Ahmed Faisal
- Department of Pharmacy, Al-Noor University College, Nineveh, Iraq
| | - Mohaned Adil
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | | | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
- School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
2
|
Valizadeh M, Derafsh E, Abdi Abyaneh F, Parsamatin SK, Noshabad FZR, Alinaghipour A, Yaghoobi Z, Taheri AT, Dadgostar E, Aschner M, Mirzaei H, Tamtaji OR, Nabavizadeh F. Non-Coding RNAs and Neurodegenerative Diseases: Information of their Roles in Apoptosis. Mol Neurobiol 2024; 61:4508-4537. [PMID: 38102518 DOI: 10.1007/s12035-023-03849-z] [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: 03/22/2023] [Accepted: 11/16/2023] [Indexed: 12/17/2023]
Abstract
Apoptosis can be known as a key factor in the pathogenesis of neurodegenerative disorders. In disease conditions, the rate of apoptosis expands and tissue damage may become apparent. Recently, the scientific studies of the non-coding RNAs (ncRNAs) has provided new information of the molecular mechanisms that contribute to neurodegenerative disorders. Numerous reports have documented that ncRNAs have important contributions to several biological processes associated with the increase of neurodegenerative disorders. In addition, microRNAs (miRNAs), circular RNAs (circRNAs), as well as, long ncRNAs (lncRNAs) represent ncRNAs subtypes with the usual dysregulation in neurodegenerative disorders. Dysregulating ncRNAs has been associated with inhibiting or stimulating apoptosis in neurodegenerative disorders. Therefore, this review highlighted several ncRNAs linked to apoptosis in neurodegenerative disorders. CircRNAs, lncRNAs, and miRNAs were also illustrated completely regarding the respective signaling pathways of apoptosis.
Collapse
Affiliation(s)
| | - Ehsan Derafsh
- Windsor University School of Medicine, Cayon, Canada
| | | | - Sayedeh Kiana Parsamatin
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Azam Alinaghipour
- School of Medical Sciences, Yazd Branch, Islamic Azad University, Yazd, Iran
| | - Zahra Yaghoobi
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, IR, Iran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, IR, Iran
| | - Abdolkarim Talebi Taheri
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsan Dadgostar
- Behavioral Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, IR, Iran
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, IR, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, IR, Iran.
| | - Omid Reza Tamtaji
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, IR, Iran.
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, IR, Iran.
| | - Fatemeh Nabavizadeh
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, IR, Iran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, IR, Iran
| |
Collapse
|
3
|
Hwang JA, Choi SK, Kim SH, Kim DW. Pharmacological Inhibition of LRRK2 Exhibits Neuroprotective Activity in Mouse Photothrombotic Stroke Model. Exp Neurobiol 2024; 33:36-45. [PMID: 38471803 PMCID: PMC10938073 DOI: 10.5607/en23023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) mutations are the most common cause of Parkinson's disease (PD). Interestingly, recent studies have reported an increased risk of stroke in patients with PD harboring LRRK2 mutations, but there is no evidence showing the functional involvement of LRRK2 in stroke. Here, we found that LRRK2 kinase activity was significantly induced in the Rose-Bengal (RB) photothrombosis-induced stroke mouse model. Interestingly, stroke infarct volumes were significantly reduced, and neurological deficits were diminished by pharmacological inhibition of LRRK2 kinase activity using MLi-2, a brain-penetrant LRRK2 kinase inhibitor. Immunohistochemical analysis showed p-LRRK2 level in stroke lesions, co-localizing with mitophagy-related proteins (PINK, Parkin, LC3B, cytochrome c), suggesting their involvement in stroke progression. Overlapping p-LRRK2 with cytochrome c/TUNEL/JC-1 (an indicator of mitochondrial membrane potential) puncta in RB photothrombosis indicated LRRK2-induced mitochondrial apoptosis, which was blocked by MLi-2. These results suggest that pharmacological inhibition of LRRK2 kinase activity could attenuate mitochondrial apoptosis, ultimately leading to neuroprotective potential in stroke progression. In conclusion, LRRK2 kinase activity might be neuro-pathogenic due to impaired mitophagy in stroke progression, and pharmacological inhibition of LRRK2 kinase activity could be beneficial in reducing the risk of stroke in patients with LRRK2 mutations.
Collapse
Affiliation(s)
- Jeong-Ah Hwang
- Center for Rare Disease Therapeutic Technology, Therapeutic & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea
| | - Seung Kyu Choi
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon 35015, Korea
| | - Seong Hwan Kim
- Center for Rare Disease Therapeutic Technology, Therapeutic & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
- Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon 35015, Korea
- Department of Anatomy & Developmental Biology, Kyung Hee University School of Dentistry, Seoul 02447, Korea
| |
Collapse
|
4
|
Lashgari NA, Roudsari NM, Niknejad A, Shamsnia HS, Shayan M, Shalmani LM, Momtaz S, Rezaei N, Abdolghaffari AH. LRRK2; Communicative Role in the Treatment of Parkinson's Disease and Ulcerative Colitis Overlapping. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1177-1188. [PMID: 38279762 DOI: 10.2174/0118715273270874231205050727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/12/2023] [Accepted: 10/23/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Involvement of gastrointestinal inflammation in Parkinson's disease (PD) pathogenesis and movement have progressively emerged. Inflammation is involved in the etiology of both PD and inflammatory bowel disease (IBD). Transformations in leucine-rich recurrent kinase 2 (LRRK2) are among the best hereditary supporters of IBD and PD. Elevated levels of LRRK2 have been reported in stimulated colonic tissue from IBD patients and peripheral invulnerable cells from irregular PD patients; thus, it is thought that LRRK2 directs inflammatory cycles. OBJECTIVE Since its revelation, LRRK2 has been seriously linked in neurons, albeit various lines of proof affirmed that LRRK2 is profoundly communicated in invulnerable cells. Subsequently, LRRK2 might sit at a junction by which stomach inflammation and higher LRRK2 levels in IBD might be a biomarker of expanded risk for inconsistent PD or potentially may address a manageable helpful objective in incendiary sicknesses that increment the risk of PD. Here, we discuss how PD and IBD share covering aggregates, especially regarding LRRK2 and present inhibitors, which could be a helpful objective in ongoing treatments. METHOD English data were obtained from Google Scholar, PubMed, Scopus, and Cochrane library studies published between 1990-December 2022. RESULT Inhibitors of the LRRK2 pathway can be considered as the novel treatment approaches for IBD and PD treatment. CONCLUSION Common mediators and pathways are involved in the pathophysiology of IBD and PD, which are majorly correlated with inflammatory situations. Such diseases could be used for further clinical investigations.
Collapse
Affiliation(s)
- Naser-Aldin Lashgari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nazanin Momeni Roudsari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amirhossein Niknejad
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hedieh Sadat Shamsnia
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maryam Shayan
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Mohaghegh Shalmani
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
5
|
Yao L, Lu F, Koc S, Zheng Z, Wang B, Zhang S, Skutella T, Lu G. LRRK2 Gly2019Ser Mutation Promotes ER Stress via Interacting with THBS1/TGF-β1 in Parkinson's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303711. [PMID: 37672887 PMCID: PMC10602550 DOI: 10.1002/advs.202303711] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/29/2023] [Indexed: 09/08/2023]
Abstract
The gene mutations of LRRK2, which encodes leucine-rich repeat kinase 2 (LRRK2), are associated with one of the most prevalent monogenic forms of Parkinson's disease (PD). However, the potential effectors of the Gly2019Ser (G2019S) mutation remain unknown. In this study, the authors investigate the effects of LRRK2 G2019S on endoplasmic reticulum (ER) stress in induced pluripotent stem cell (iPSC)-induced dopamine neurons and explore potential therapeutic targets in mice model. These findings demonstrate that LRRK2 G2019S significantly promotes ER stress in neurons and mice. Interestingly, inhibiting LRRK2 activity can ameliorate ER stress induced by the mutation. Moreover, LRRK2 mutation can induce ER stress by directly interacting with thrombospondin-1/transforming growth factor beta1 (THBS1/TGF-β1). Inhibition of LRRK2 kinase activity can effectively suppress ER stress and the expression of THBS1/TGF-β1. Knocking down THBS1 can rescue ER stress by interacting with TGF-β1 and behavior burden caused by the LRRK2 mutation, while suppression of TGF-β1 has a similar effect. Overall, it is demonstrated that the LRRK2 mutation promotes ER stress by directly interacting with THBS1/TGF-β1, leading to neural death in PD. These findings provide valuable insights into the pathogenesis of PD, highlighting potential diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Longping Yao
- Department of NeurosurgeryFirst Affiliated Hospital of Nanchang UniversityNanchang330209P. R. China
- Department of NeurosurgeryZhujiang HospitalSouthern Medical UniversityGuangzhou510282P. R. China
- Institute for Anatomy and Cell BiologyMedical FacultyHeidelberg University69120HeidelbergGermany
| | - Fengfei Lu
- Department of NeurosurgeryZhujiang HospitalSouthern Medical UniversityGuangzhou510282P. R. China
| | - Sumeyye Koc
- Department of NeuroscienceInstitute of Health SciencesOndokuz Mayıs UniversitySamsun55139Turkey
| | - Zijian Zheng
- Department of NeurosurgeryFirst Affiliated Hospital of Nanchang UniversityNanchang330209P. R. China
| | - Baoyan Wang
- Department of NeurosurgeryZhujiang HospitalSouthern Medical UniversityGuangzhou510282P. R. China
| | - Shizhong Zhang
- Department of NeurosurgeryZhujiang HospitalSouthern Medical UniversityGuangzhou510282P. R. China
| | - Thomas Skutella
- Institute for Anatomy and Cell BiologyMedical FacultyHeidelberg University69120HeidelbergGermany
| | - Guohui Lu
- Department of NeurosurgeryFirst Affiliated Hospital of Nanchang UniversityNanchang330209P. R. China
| |
Collapse
|
6
|
Kania E, Long JS, McEwan DG, Welkenhuyzen K, La Rovere R, Luyten T, Halpin J, Lobbestael E, Baekelandt V, Bultynck G, Ryan KM, Parys JB. LRRK2 phosphorylation status and kinase activity regulate (macro)autophagy in a Rab8a/Rab10-dependent manner. Cell Death Dis 2023; 14:436. [PMID: 37454104 PMCID: PMC10349885 DOI: 10.1038/s41419-023-05964-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/27/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common genetic cause of Parkinson's disease (PD), with growing importance also for Crohn's disease and cancer. LRRK2 is a large and complex protein possessing both GTPase and kinase activity. Moreover, LRRK2 activity and function can be influenced by its phosphorylation status. In this regard, many LRRK2 PD-associated mutants display decreased phosphorylation of the constitutive phosphorylation cluster S910/S935/S955/S973, but the role of these changes in phosphorylation status with respect to LRRK2 physiological functions remains unknown. Here, we propose that the S910/S935/S955/S973 phosphorylation sites act as key regulators of LRRK2-mediated autophagy under both basal and starvation conditions. We show that quadruple LRRK2 phosphomutant cells (4xSA; S910A/S935A/S955A/S973A) have impaired lysosomal functionality and fail to induce and proceed with autophagy during starvation. In contrast, treatment with the specific LRRK2 kinase inhibitors MLi-2 (100 nM) or PF-06447475 (150 nM), which also led to decreased LRRK2 phosphorylation of S910/S935/S955/S973, did not affect autophagy. In explanation, we demonstrate that the autophagy impairment due to the 4xSA LRRK2 phospho-dead mutant is driven by its enhanced LRRK2 kinase activity. We show mechanistically that this involves increased phosphorylation of LRRK2 downstream targets Rab8a and Rab10, as the autophagy impairment in 4xSA LRRK2 cells is counteracted by expression of phosphorylation-deficient mutants T72A Rab8a and T73A Rab10. Similarly, reduced autophagy and decreased LRRK2 phosphorylation at the constitutive sites were observed in cells expressing the pathological R1441C LRRK2 PD mutant, which also displays increased kinase activity. These data underscore the relation between LRRK2 phosphorylation at its constitutive sites and the importance of increased LRRK2 kinase activity in autophagy regulation and PD pathology.
Collapse
Affiliation(s)
- Elżbieta Kania
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Herestraat 49, Campus Gasthuisberg O&NI - B802, 3000, Leuven, Belgium
| | - Jaclyn S Long
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - David G McEwan
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Kirsten Welkenhuyzen
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Herestraat 49, Campus Gasthuisberg O&NI - B802, 3000, Leuven, Belgium
| | - Rita La Rovere
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Herestraat 49, Campus Gasthuisberg O&NI - B802, 3000, Leuven, Belgium
| | - Tomas Luyten
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Herestraat 49, Campus Gasthuisberg O&NI - B802, 3000, Leuven, Belgium
| | - John Halpin
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Evy Lobbestael
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences & Leuven Brain Institute, KU Leuven, Herestraat 49, Campus Gasthuisberg B1023, 3000, Leuven, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences & Leuven Brain Institute, KU Leuven, Herestraat 49, Campus Gasthuisberg B1023, 3000, Leuven, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Herestraat 49, Campus Gasthuisberg O&NI - B802, 3000, Leuven, Belgium
| | - Kevin M Ryan
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK.
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK.
| | - Jan B Parys
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Herestraat 49, Campus Gasthuisberg O&NI - B802, 3000, Leuven, Belgium.
| |
Collapse
|
7
|
Leem YH, Kim DY, Park JE, Kim HS. Necrosulfonamide exerts neuroprotective effect by inhibiting necroptosis, neuroinflammation, and α-synuclein oligomerization in a subacute MPTP mouse model of Parkinson's disease. Sci Rep 2023; 13:8783. [PMID: 37258791 DOI: 10.1038/s41598-023-35975-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/26/2023] [Indexed: 06/02/2023] Open
Abstract
Parkinson's disease (PD) is an incurable movement disorder characterized by dopaminergic cell loss, neuroinflammation, and α-synuclein pathology. Herein, we investigated the therapeutic effects of necrosulfonamide (NSA), a specific inhibitor of mixed lineage kinase domain-like protein (MLKL), in a subacute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. MLKL is an executor of necroptosis, a programmed cell death pathway that causes inflammation. Repeated administration of NSA resulted in the recovery of impaired motor performance and dopaminergic degeneration. Furthermore, NSA inhibited the phosphorylation, ubiquitylation, and oligomerization of MLKL, all of which are associated with MLKL cell death-inducing activity in dopaminergic cells in the substantia nigra (SN). NSA also inhibited microglial activation and reactive astrogliosis as well as the MPTP-induced expression of proinflammatory molecules such as tumor necrosis factor-α, interleukin-1β, inducible nitric oxide synthase, and cystatin F. Furthermore, NSA inhibited α-synuclein oligomerization and phosphorylation in the SN of MPTP-treated mice by inhibiting the activity of glycogen synthase kinase 3β and matrix metalloproteinase-3. In conclusion, NSA has anti-necroptotic, anti-inflammatory, and anti-synucleinopathic effects on PD pathology. Therefore, NSA is a potential therapeutic candidate for PD.
Collapse
Affiliation(s)
- Yea-Hyun Leem
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, 808-1 Magok-Dong, Gangseo-gu, Seoul, 07804, South Korea
| | - Do-Yeon Kim
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, 808-1 Magok-Dong, Gangseo-gu, Seoul, 07804, South Korea
| | - Jung-Eun Park
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, 808-1 Magok-Dong, Gangseo-gu, Seoul, 07804, South Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, 808-1 Magok-Dong, Gangseo-gu, Seoul, 07804, South Korea.
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
8
|
Chang KH, Liu CH, Wang YR, Lo YS, Chang CW, Wu HC, Chen CM. Upregulation of APAF1 and CSF1R in Peripheral Blood Mononuclear Cells of Parkinson's Disease. Int J Mol Sci 2023; 24:ijms24087095. [PMID: 37108258 PMCID: PMC10139006 DOI: 10.3390/ijms24087095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/08/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Increased oxidative stress and neuroinflammation play a crucial role in the pathogenesis of Parkinson's disease (PD). In this study, the expression levels of 52 genes related to oxidative stress and inflammation were measured in peripheral blood mononuclear cells of the discovery cohort including 48 PD patients and 25 healthy controls. Four genes, including ALDH1A, APAF1, CR1, and CSF1R, were found to be upregulated in PD patients. The expression patterns of these genes were validated in a second cohort of 101 PD patients and 61 healthy controls. The results confirmed the upregulation of APAF1 (PD: 0.34 ± 0.18, control: 0.26 ± 0.11, p < 0.001) and CSF1R (PD: 0.38 ± 0.12, control: 0.33 ± 0.10, p = 0.005) in PD patients. The expression level of APAF1 was correlated with the scores of the Unified Parkinson's Disease Rating Scale (UPDRS, r = 0.235, p = 0.018) and 39-item PD questionnaire (PDQ-39, r = 0.250, p = 0.012). The expression level of CSF1R was negatively correlated with the scores of the mini-mental status examination (MMSE, r = -0.200, p = 0.047) and Montréal Cognitive Assessment (MoCA, r = -0.226, p = 0.023). These results highly suggest that oxidative stress biomarkers in peripheral blood may be useful in monitoring the progression of motor disabilities and cognitive decline in PD patients.
Collapse
Affiliation(s)
- Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chia-Hsin Liu
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yi-Ru Wang
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yen-Shi Lo
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chun-Wei Chang
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Hsiu-Chuan Wu
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| |
Collapse
|
9
|
Dallmann-Sauer M, Xu YZ, da Costa ALF, Tao S, Gomes TA, Prata RBDS, Correa-Macedo W, Manry J, Alcaïs A, Abel L, Cobat A, Fava VM, Pinheiro RO, Lara FA, Probst CM, Mira MT, Schurr E. Allele-dependent interaction of LRRK2 and NOD2 in leprosy. PLoS Pathog 2023; 19:e1011260. [PMID: 36972292 PMCID: PMC10079233 DOI: 10.1371/journal.ppat.1011260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/06/2023] [Accepted: 03/02/2023] [Indexed: 03/29/2023] Open
Abstract
Leprosy, caused by Mycobacterium leprae, rarely affects children younger than 5 years. Here, we studied a multiplex leprosy family that included monozygotic twins aged 22 months suffering from paucibacillary leprosy. Whole genome sequencing identified three amino acid mutations previously associated with Crohn’s disease and Parkinson’s disease as candidate variants for early onset leprosy: LRRK2 N551K, R1398H and NOD2 R702W. In genome-edited macrophages, we demonstrated that cells expressing the LRRK2 mutations displayed reduced apoptosis activity following mycobacterial challenge independently of NOD2. However, employing co-immunoprecipitation and confocal microscopy we showed that LRRK2 and NOD2 proteins interacted in RAW cells and monocyte-derived macrophages, and that this interaction was substantially reduced for the NOD2 R702W mutation. Moreover, we observed a joint effect of LRRK2 and NOD2 variants on Bacillus Calmette-Guérin (BCG)-induced respiratory burst, NF-κB activation and cytokine/chemokine secretion with a strong impact for the genotypes found in the twins consistent with a role of the identified mutations in the development of early onset leprosy.
Collapse
Affiliation(s)
- Monica Dallmann-Sauer
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre; Montreal, Canada
- McGill International TB Centre, McGill University; Montreal, Canada
- Departments of Human Genetics and Medicine, Faculty of Medicine and Health Science, McGill University; Montreal, Canada
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná; Curitiba, Brazil
| | - Yong Zhong Xu
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre; Montreal, Canada
- McGill International TB Centre, McGill University; Montreal, Canada
| | - Ana Lúcia França da Costa
- Department of Specialized Medicine, Health Sciences Center, Federal University of Piauí; Teresina, Brazil
| | - Shao Tao
- Division of Experimental Medicine, Faculty of Medicine, McGill University; Montreal, Canada
- The Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre; Montreal, Canada
| | - Tiago Araujo Gomes
- Laboratory of Cellular Microbiology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation; Rio de Janeiro, Brazil
| | | | - Wilian Correa-Macedo
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre; Montreal, Canada
- McGill International TB Centre, McGill University; Montreal, Canada
- Department of Biochemistry, Faculty of Medicine and Health Science, McGill University; Montreal, Canada
| | - Jérémy Manry
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre; Montreal, Canada
- McGill International TB Centre, McGill University; Montreal, Canada
| | - Alexandre Alcaïs
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U.1163, Paris, France
- Université Paris Cité, Imagine Institute, Paris, France
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U.1163, Paris, France
- Université Paris Cité, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, United States of America
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U.1163, Paris, France
- Université Paris Cité, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, United States of America
| | - Vinicius M. Fava
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre; Montreal, Canada
- McGill International TB Centre, McGill University; Montreal, Canada
| | - Roberta Olmo Pinheiro
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation; Rio de Janeiro, Brazil
| | - Flavio Alves Lara
- Laboratory of Cellular Microbiology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation; Rio de Janeiro, Brazil
| | - Christian M. Probst
- Laboratory of Systems and Molecular Biology of Trypanosomatids, Instituto Carlos Chagas; FIOCRUZ, Curitiba, Brazil
| | - Marcelo T. Mira
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná; Curitiba, Brazil
- * E-mail: (M.T.M); (E.S.)
| | - Erwin Schurr
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre; Montreal, Canada
- McGill International TB Centre, McGill University; Montreal, Canada
- Departments of Human Genetics and Medicine, Faculty of Medicine and Health Science, McGill University; Montreal, Canada
- Department of Biochemistry, Faculty of Medicine and Health Science, McGill University; Montreal, Canada
- * E-mail: (M.T.M); (E.S.)
| |
Collapse
|
10
|
Olufunmilayo EO, Gerke-Duncan MB, Holsinger RMD. Oxidative Stress and Antioxidants in Neurodegenerative Disorders. Antioxidants (Basel) 2023; 12:antiox12020517. [PMID: 36830075 PMCID: PMC9952099 DOI: 10.3390/antiox12020517] [Citation(s) in RCA: 181] [Impact Index Per Article: 90.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Neurodegenerative disorders constitute a substantial proportion of neurological diseases with significant public health importance. The pathophysiology of neurodegenerative diseases is characterized by a complex interplay of various general and disease-specific factors that lead to the end point of neuronal degeneration and loss, and the eventual clinical manifestations. Oxidative stress is the result of an imbalance between pro-oxidant species and antioxidant systems, characterized by an elevation in the levels of reactive oxygen and reactive nitrogen species, and a reduction in the levels of endogenous antioxidants. Recent studies have increasingly highlighted oxidative stress and associated mitochondrial dysfunction to be important players in the pathophysiologic processes involved in neurodegenerative conditions. In this article, we review the current knowledge of the general effects of oxidative stress on the central nervous system, the different specific routes by which oxidative stress influences the pathophysiologic processes involved in Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis and Huntington's disease, and how oxidative stress may be therapeutically reversed/mitigated in order to stall the pathological progression of these neurodegenerative disorders to bring about clinical benefits.
Collapse
Affiliation(s)
- Edward O. Olufunmilayo
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Department of Medicine, University College Hospital, Queen Elizabeth Road, Oritamefa, Ibadan 5116, PMB, Nigeria
| | - Michelle B. Gerke-Duncan
- Education Innovation, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - R. M. Damian Holsinger
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Correspondence:
| |
Collapse
|
11
|
Pathophysiological evaluation of the LRRK2 G2385R risk variant for Parkinson’s disease. NPJ Parkinsons Dis 2022; 8:97. [PMID: 35931783 PMCID: PMC9355991 DOI: 10.1038/s41531-022-00367-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Missense variants in leucine-rich repeat kinase 2 (LRRK2) lead to familial and sporadic Parkinson’s disease (PD). The pathological features of PD patients with LRRK2 variants differ. Here, we report an autopsy case harboring the LRRK2 G2385R, a risk variant for PD occurring mainly in Asian populations. The patient exhibited levodopa-responsive parkinsonism at the early stage and visual hallucinations at the advanced stage. The pathological study revealed diffuse Lewy bodies with neurofibrillary tangles, amyloid plaques, and mild signs of neuroinflammation. Biochemically, detergent-insoluble phospho-α-synuclein was accumulated in the frontal, temporal, entorhinal cortexes, and putamen, consistent with the pathological observations. Elevated phosphorylation of Rab10, a substrate of LRRK2, was also prominent in various brain regions. In conclusion, G2385R appears to increase LRRK2 kinase activity in the human brain, inducing a deleterious brain environment that causes Lewy body pathology.
Collapse
|
12
|
Wal P, Dwivedi J, Wal A, Vig H, Singh Y. Detailed insight into the pathophysiology and the behavioral complications associated with the Parkinson's disease and its medications. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2022. [DOI: 10.1186/s43094-022-00425-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The loss of dopamine neurons in the substantia nigra, as well as other mostly catecholaminergic neurons, causes many of the motor symptoms that define Parkinson's disease. Parkinson's disease is commonly thought of as a movement disorder, the significant prevalence of psychiatric complications such as cognitive impairment, and psychosis suggests it should be considered a neuropsychiatric illness, and all behavioral complications are linked to growing disability and the medication.
Main body
Apart from the disease-induced abnormalities, there are several other side effects of the disease and also from the medication used to prevent the disease. This article focuses on the pathogenesis of Parkinson’s disease and also the behavioral abnormalities caused by the disease and its medication. The study's data were gathered by searching several review articles and research papers from a variety of sources, including Elsevier, PubMed, Research Gate, Journal of Pharmaceutical Science, etc., from the year 1985 to 2021. Parkinson's disease is a neurodegenerative disease caused by a variety of complex processes. It is responsible not just for motor symptoms, but also for a variety of behavioral symptoms that can arise as a result of the disease and/or medication.
Conclusion
Only symptomatic drugs are available; thus, finding treatments that directly address the disease mechanisms causing Parkinson’s disease is essential. To alleviate the disease's burden on patients and their families, better treatments for the neuropsychiatric repercussions of Parkinson's disease are required.
Graphical Abstract
Collapse
|
13
|
Thomas C, Wurzer L, Malle E, Ristow M, Madreiter-Sokolowski CT. Modulation of Reactive Oxygen Species Homeostasis as a Pleiotropic Effect of Commonly Used Drugs. FRONTIERS IN AGING 2022; 3:905261. [PMID: 35821802 PMCID: PMC9261327 DOI: 10.3389/fragi.2022.905261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/18/2022] [Indexed: 01/17/2023]
Abstract
Age-associated diseases represent a growing burden for global health systems in our aging society. Consequently, we urgently need innovative strategies to counteract these pathological disturbances. Overwhelming generation of reactive oxygen species (ROS) is associated with age-related damage, leading to cellular dysfunction and, ultimately, diseases. However, low-dose ROS act as crucial signaling molecules and inducers of a vaccination-like response to boost antioxidant defense mechanisms, known as mitohormesis. Consequently, modulation of ROS homeostasis by nutrition, exercise, or pharmacological interventions is critical in aging. Numerous nutrients and approved drugs exhibit pleiotropic effects on ROS homeostasis. In the current review, we provide an overview of drugs affecting ROS generation and ROS detoxification and evaluate the potential of these effects to counteract the development and progression of age-related diseases. In case of inflammation-related dysfunctions, cardiovascular- and neurodegenerative diseases, it might be essential to strengthen antioxidant defense mechanisms in advance by low ROS level rises to boost the individual ROS defense mechanisms. In contrast, induction of overwhelming ROS production might be helpful to fight pathogens and kill cancer cells. While we outline the potential of ROS manipulation to counteract age-related dysfunction and diseases, we also raise the question about the proper intervention time and dosage.
Collapse
Affiliation(s)
- Carolin Thomas
- Laboratory of Energy Metabolism Institute of Translational Medicine Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Lia Wurzer
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Ernst Malle
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Michael Ristow
- Laboratory of Energy Metabolism Institute of Translational Medicine Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | | |
Collapse
|
14
|
Lima IS, Pêgo AC, Barros JT, Prada AR, Gozzelino R. Cell Death-Osis of Dopaminergic Neurons and the Role of Iron in Parkinson's Disease. Antioxid Redox Signal 2021; 35:453-473. [PMID: 33233941 DOI: 10.1089/ars.2020.8229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Significance: There is still no cure for neurodegenerative diseases, such as Parkinson's disease (PD). Current treatments are based on the attempt to reduce dopaminergic neuronal loss, and multidisciplinary approaches have been used to provide only a temporary symptoms' relief. In addition to the difficulties of drugs developed against PD to access the brain, the specificity of those inhibitory compounds could be a concern. This because neurons might degenerate by activating distinct signaling pathways, which are often initiated by the same stimulus. Recent Advances: Apoptosis, necroptosis, and ferroptosis were shown to significantly contribute to PD progression and, so far, are the main death programs described as capable to alter brain homeostasis. Their activation is characterized by different biochemical and morphological features, some of which might even share the same molecular players. Critical Issues: If there is a pathological need to engage, in PD, multiple death programs, sequentially or simultaneously, is not clear yet. Possibly the activation of apoptosis, necroptosis, and/or ferroptosis correlates to different PD stages and symptom severities. This would imply that the efficacy of therapeutic approaches against neuronal death might depend on the death program they target and the relevance of this death pathway on a specific PD phase. Future Directions: In this review, we describe the molecular mechanisms underlying the activation of apoptosis, necroptosis, and ferroptosis in PD. Understanding the interrelationship between different death pathways' activation in PD is of utmost importance for the development of therapeutic approaches against disease progression. Antioxid. Redox Signal. 35, 453-473.
Collapse
Affiliation(s)
- Illyane Sofia Lima
- Inflammation and Neurodegeneration Laboratory, Centro de Estudos de Doenças Crónicas (CEDOC)/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Ana Catarina Pêgo
- Inflammation and Neurodegeneration Laboratory, Centro de Estudos de Doenças Crónicas (CEDOC)/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - João Tomas Barros
- Inflammation and Neurodegeneration Laboratory, Centro de Estudos de Doenças Crónicas (CEDOC)/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Ana Rita Prada
- Inflammation and Neurodegeneration Laboratory, Centro de Estudos de Doenças Crónicas (CEDOC)/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Raffaella Gozzelino
- Inflammation and Neurodegeneration Laboratory, Centro de Estudos de Doenças Crónicas (CEDOC)/NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Universidade Técnica do Atlântico (UTA), São Vicente, Cabo Verde
| |
Collapse
|
15
|
Verma A, Ebanks K, Fok CY, Lewis PA, Bettencourt C, Bandopadhyay R. In silico comparative analysis of LRRK2 interactomes from brain, kidney and lung. Brain Res 2021; 1765:147503. [PMID: 33915162 PMCID: PMC8212912 DOI: 10.1016/j.brainres.2021.147503] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/06/2021] [Accepted: 04/23/2021] [Indexed: 01/11/2023]
Abstract
Mutations in LRRK2 are the most frequent cause of familial Parkinson's disease (PD), with common LRRK2 non-coding variants also acting as risk factors for idiopathic PD. Currently, therapeutic agents targeting LRRK2 are undergoing advanced clinical trials in humans, however, it is important to understand the wider implications of LRRK2 targeted treatments given that LRRK2 is expressed in diverse tissues including the brain, kidney and lungs. This presents challenges to treatment in terms of effects on peripheral organ functioning, thus, protein interactors of LRRK2 could be targeted in lieu to optimize therapeutic effects. Herein an in-silico analysis of LRRK2 direct interactors in brain tissue from various brain regionswas conducted along with a comparative analysis of the LRRK2 interactome in the brain, kidney, and lung tissues. This was carried out based on curated protein-protein interaction (PPI) data from protein interaction databases such as HIPPIE, human gene/protein expression databases and Gene ontology (GO) enrichment analysis using Bingo. Seven targets (MAP2K6, MATK, MAPT, PAK6, SH3GL2, CDC42EP3 and CHGB) were found to be viable objectives for LRRK2 based investigations for PD that would have minimal impact on optimal functioning within peripheral organs. Specifically, MAPT, CHGB, PAK6, and SH3GL2 interacted with LRRK2 in the brain and kidney but not in lung tissue whilst LRRK2-MAP2K6 interacted only in the cerebellum and MATK-LRRK2 interaction was absent in kidney tissues. CDC42EP3 expression levels were low in brain tissues compared to kidney/lung. The results of this computational analysis suggest new avenues for experimental investigations towards LRRK2-targeted therapeutics.
Collapse
Affiliation(s)
- Amrita Verma
- Reta Lila Weston Institute of Neurological Studies, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom
| | - Kirsten Ebanks
- Reta Lila Weston Institute of Neurological Studies, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom
| | - Chi-Yee Fok
- Reta Lila Weston Institute of Neurological Studies, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom
| | - Patrick A Lewis
- Royal Veterinary College, Royal College Street, London NW10TV, United Kingdom; Department of Neurodegenerative Disease and Queen Square Brain Bank, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Conceicao Bettencourt
- Department of Neurodegenerative Disease and Queen Square Brain Bank, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom
| | - Rina Bandopadhyay
- Reta Lila Weston Institute of Neurological Studies, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom.
| |
Collapse
|
16
|
Moujalled D, Strasser A, Liddell JR. Molecular mechanisms of cell death in neurological diseases. Cell Death Differ 2021; 28:2029-2044. [PMID: 34099897 PMCID: PMC8257776 DOI: 10.1038/s41418-021-00814-y] [Citation(s) in RCA: 404] [Impact Index Per Article: 101.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
Tightly orchestrated programmed cell death (PCD) signalling events occur during normal neuronal development in a spatially and temporally restricted manner to establish the neural architecture and shaping the CNS. Abnormalities in PCD signalling cascades, such as apoptosis, necroptosis, pyroptosis, ferroptosis, and cell death associated with autophagy as well as in unprogrammed necrosis can be observed in the pathogenesis of various neurological diseases. These cell deaths can be activated in response to various forms of cellular stress (exerted by intracellular or extracellular stimuli) and inflammatory processes. Aberrant activation of PCD pathways is a common feature in neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS), Alzheimer's disease, Parkinson's disease, and Huntington's disease, resulting in unwanted loss of neuronal cells and function. Conversely, inactivation of PCD is thought to contribute to the development of brain cancers and to impact their response to therapy. For many neurodegenerative diseases and brain cancers current treatment strategies have only modest effect, engendering the need for investigations into the origins of these diseases. With many diseases of the brain displaying aberrations in PCD pathways, it appears that agents that can either inhibit or induce PCD may be critical components of future therapeutic strategies. The development of such therapies will have to be guided by preclinical studies in animal models that faithfully mimic the human disease. In this review, we briefly describe PCD and unprogrammed cell death processes and the roles they play in contributing to neurodegenerative diseases or tumorigenesis in the brain. We also discuss the interplay between distinct cell death signalling cascades and disease pathogenesis and describe pharmacological agents targeting key players in the cell death signalling pathways that have progressed through to clinical trials.
Collapse
Affiliation(s)
- Diane Moujalled
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia.
| | - Andreas Strasser
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia.
| | - Jeffrey R Liddell
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
17
|
A Novel LRRK2 Variant p.G2294R in the WD40 Domain Identified in Familial Parkinson's Disease Affects LRRK2 Protein Levels. Int J Mol Sci 2021; 22:ijms22073708. [PMID: 33918221 PMCID: PMC8038167 DOI: 10.3390/ijms22073708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/14/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a major causative gene of late-onset familial Parkinson’s disease (PD). The suppression of kinase activity is believed to confer neuroprotection, as most pathogenic variants of LRRK2 associated with PD exhibit increased kinase activity. We herein report a novel LRRK2 variant—p.G2294R—located in the WD40 domain, detected through targeted gene-panel screening in a patient with familial PD. The proband showed late-onset Parkinsonism with dysautonomia and a good response to levodopa, without cognitive decline or psychosis. Cultured cell experiments revealed that p.G2294R is highly destabilized at the protein level. The LRRK2 p.G2294R protein expression was upregulated in the patient’s peripheral blood lymphocytes. However, macrophages differentiated from the same peripheral blood showed decreased LRRK2 protein levels. Moreover, our experiment indicated reduced phagocytic activity in the pathogenic yeasts and α-synuclein fibrils. This PD case presents an example wherein the decrease in LRRK2 activity did not act in a neuroprotective manner. Further investigations are needed in order to elucidate the relationship between LRRK2 expression in the central nervous system and the pathogenesis caused by altered LRRK2 activity.
Collapse
|
18
|
Fais M, Sanna G, Galioto M, Nguyen TTD, Trần MUT, Sini P, Carta F, Turrini F, Xiong Y, Dawson TM, Dawson VL, Crosio C, Iaccarino C. LRRK2 Modulates the Exocyst Complex Assembly by Interacting with Sec8. Cells 2021; 10:203. [PMID: 33498474 PMCID: PMC7909581 DOI: 10.3390/cells10020203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 11/17/2022] Open
Abstract
Mutations in LRRK2 play a critical role in both familial and sporadic Parkinson's disease (PD). Up to date, the role of LRRK2 in PD onset and progression remains largely unknown. However, experimental evidence highlights a critical role of LRRK2 in the control of vesicle trafficking, likely by Rab phosphorylation, that in turn may regulate different aspects of neuronal physiology. Here we show that LRRK2 interacts with Sec8, one of eight subunits of the exocyst complex. The exocyst complex is an evolutionarily conserved multisubunit protein complex mainly involved in tethering secretory vesicles to the plasma membrane and implicated in the regulation of multiple biological processes modulated by vesicle trafficking. Interestingly, Rabs and exocyst complex belong to the same protein network. Our experimental evidence indicates that LRRK2 kinase activity or the presence of the LRRK2 kinase domain regulate the assembly of exocyst subunits and that the over-expression of Sec8 significantly rescues the LRRK2 G2019S mutant pathological effect. Our findings strongly suggest an interesting molecular mechanism by which LRRK2 could modulate vesicle trafficking and may have important implications to decode the complex role that LRRK2 plays in neuronal physiology.
Collapse
Affiliation(s)
- Milena Fais
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (M.F.); (G.S.); (M.G.); (T.T.D.N.); (M.U.T.T.); (P.S.); (C.C.)
| | - Giovanna Sanna
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (M.F.); (G.S.); (M.G.); (T.T.D.N.); (M.U.T.T.); (P.S.); (C.C.)
| | - Manuela Galioto
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (M.F.); (G.S.); (M.G.); (T.T.D.N.); (M.U.T.T.); (P.S.); (C.C.)
| | - Thi Thanh Duyen Nguyen
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (M.F.); (G.S.); (M.G.); (T.T.D.N.); (M.U.T.T.); (P.S.); (C.C.)
| | - Mai Uyên Thi Trần
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (M.F.); (G.S.); (M.G.); (T.T.D.N.); (M.U.T.T.); (P.S.); (C.C.)
| | - Paola Sini
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (M.F.); (G.S.); (M.G.); (T.T.D.N.); (M.U.T.T.); (P.S.); (C.C.)
| | | | - Franco Turrini
- Nurex Srl, 07100 Sassari, Italy; (F.C.); (F.T.)
- Department of Oncology, University of Turin, 10126 Turin, Italy
| | - Yulan Xiong
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.X.); (T.M.D.); (V.L.D.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.X.); (T.M.D.); (V.L.D.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Valina L. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.X.); (T.M.D.); (V.L.D.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Claudia Crosio
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (M.F.); (G.S.); (M.G.); (T.T.D.N.); (M.U.T.T.); (P.S.); (C.C.)
| | - Ciro Iaccarino
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (M.F.); (G.S.); (M.G.); (T.T.D.N.); (M.U.T.T.); (P.S.); (C.C.)
| |
Collapse
|
19
|
Novel PET Biomarkers to Disentangle Molecular Pathways across Age-Related Neurodegenerative Diseases. Cells 2020; 9:cells9122581. [PMID: 33276490 PMCID: PMC7761606 DOI: 10.3390/cells9122581] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/25/2020] [Accepted: 11/28/2020] [Indexed: 12/11/2022] Open
Abstract
There is a need to disentangle the etiological puzzle of age-related neurodegenerative diseases, whose clinical phenotypes arise from known, and as yet unknown, pathways that can act distinctly or in concert. Enhanced sub-phenotyping and the identification of in vivo biomarker-driven signature profiles could improve the stratification of patients into clinical trials and, potentially, help to drive the treatment landscape towards the precision medicine paradigm. The rapidly growing field of neuroimaging offers valuable tools to investigate disease pathophysiology and molecular pathways in humans, with the potential to capture the whole disease course starting from preclinical stages. Positron emission tomography (PET) combines the advantages of a versatile imaging technique with the ability to quantify, to nanomolar sensitivity, molecular targets in vivo. This review will discuss current research and available imaging biomarkers evaluating dysregulation of the main molecular pathways across age-related neurodegenerative diseases. The molecular pathways focused on in this review involve mitochondrial dysfunction and energy dysregulation; neuroinflammation; protein misfolding; aggregation and the concepts of pathobiology, synaptic dysfunction, neurotransmitter dysregulation and dysfunction of the glymphatic system. The use of PET imaging to dissect these molecular pathways and the potential to aid sub-phenotyping will be discussed, with a focus on novel PET biomarkers.
Collapse
|
20
|
Chang KH, Chen CM. The Role of Oxidative Stress in Parkinson's Disease. Antioxidants (Basel) 2020; 9:antiox9070597. [PMID: 32650609 PMCID: PMC7402083 DOI: 10.3390/antiox9070597] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022] Open
Abstract
Parkinson’s disease (PD) is caused by progressive neurodegeneration of dopaminergic (DAergic) neurons with abnormal accumulation of α-synuclein in substantia nigra (SN). Studies have suggested the potential involvement of dopamine, iron, calcium, mitochondria and neuroinflammation in contributing to overwhelmed oxidative stress and neurodegeneration in PD. Function studies on PD-causative mutations of SNCA, PRKN, PINK1, DJ-1, LRRK2, FBXO7 and ATP13A2 further indicate the role of oxidative stress in the pathogenesis of PD. Therefore, it is reasonable that molecules involved in oxidative stress, such as DJ-1, coenzyme Q10, uric acid, 8-hydroxy-2’-deoxyguanosin, homocysteine, retinoic acid/carotenes, vitamin E, glutathione peroxidase, superoxide dismutase, xanthine oxidase and products of lipid peroxidation, could be candidate biomarkers for PD. Applications of antioxidants to modulate oxidative stress could be a strategy in treating PD. Although a number of antioxidants, such as creatine, vitamin E, coenzyme Q10, pioglitazone, melatonin and desferrioxamine, have been tested in clinical trials, none of them have demonstrated conclusive evidence to ameliorate the neurodegeneration in PD patients. Difficulties in clinical studies may be caused by the long-standing progression of neurodegeneration, lack of biomarkers for premotor stage of PD and inadequate drug delivery across blood–brain barrier. Solutions for these challenges will be warranted for future studies with novel antioxidative treatment in PD patients.
Collapse
Affiliation(s)
| | - Chiung-Mei Chen
- Correspondence: ; Tel.: +886-3-3281200 (ext. 8347); Fax: +886-3-3288849
| |
Collapse
|
21
|
Rivero-Ríos P, Romo-Lozano M, Fasiczka R, Naaldijk Y, Hilfiker S. LRRK2-Related Parkinson's Disease Due to Altered Endolysosomal Biology With Variable Lewy Body Pathology: A Hypothesis. Front Neurosci 2020; 14:556. [PMID: 32581693 PMCID: PMC7287096 DOI: 10.3389/fnins.2020.00556] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Mutations in the gene encoding for leucine-rich repeat kinase 2 (LRRK2) are associated with both familial and sporadic Parkinson's disease (PD). LRRK2 encodes a large protein comprised of a GTPase and a kinase domain. All pathogenic variants converge on enhancing LRRK2 kinase substrate phosphorylation, and distinct LRRK2 kinase inhibitors are currently in various stages of clinical trials. Although the precise pathophysiological functions of LRRK2 remain largely unknown, PD-associated mutants have been shown to alter various intracellular vesicular trafficking pathways, especially those related to endolysosomal protein degradation events. In addition, biochemical studies have identified a subset of Rab proteins, small GTPases required for all vesicular trafficking steps, as substrate proteins for the LRRK2 kinase activity in vitro and in vivo. Therefore, it is crucial to evaluate the impact of such phosphorylation on neurodegenerative mechanisms underlying LRRK2-related PD, especially with respect to deregulated Rab-mediated endolysosomal membrane trafficking and protein degradation events. Surprisingly, a significant proportion of PD patients due to LRRK2 mutations display neuronal cell loss in the substantia nigra pars compacta in the absence of any apparent α-synuclein-containing Lewy body neuropathology. These findings suggest that endolysosomal alterations mediated by pathogenic LRRK2 per se are not sufficient to cause α-synuclein aggregation. Here, we will review current knowledge about the link between pathogenic LRRK2, Rab protein phosphorylation and endolysosomal trafficking alterations, and we will propose a testable working model whereby LRRK2-related PD may present with variable LB pathology.
Collapse
Affiliation(s)
- Pilar Rivero-Ríos
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
| | - María Romo-Lozano
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Rachel Fasiczka
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Yahaira Naaldijk
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Sabine Hilfiker
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
22
|
Diana A, Collu M, Casu MA, Mocci I, Aguilar-Santelises M, Setzu MD. Improvements of Motor Performances in the Drosophila LRRK2 Loss-of-Function Model of Parkinson’s Disease: Effects of Dialyzed Leucocyte Extracts from Human Serum. Brain Sci 2020; 10:brainsci10010045. [PMID: 31947539 PMCID: PMC7017078 DOI: 10.3390/brainsci10010045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/05/2020] [Accepted: 01/09/2020] [Indexed: 11/16/2022] Open
Abstract
Within neurodegenerative syndromes, Parkinson’s disease (PD) is typically associated with its locomotor defects, sleep disturbances and related dopaminergic (DA) neuron loss. The fruit fly, Drosophila melanogaster (D. melanogaster), with leucine-rich repeat kinase 2 mutants (LRRK2) loss-of-function in the WD40 domain, provides mechanistic insights into corresponding human behaviour, possibly disclosing some physiopathologic features of PD in both genetic and sporadic forms. Moreover, several data support the boosting impact of innate and adaptive immunity pathways for driving the progression of PD. In this context, human dialyzable leukocyte extracts (DLE) have been extensively used to transfer antigen-specific information that influences the activity of various immune components, including inflammatory cytokines. Hence, the main goal of our study was to ascertain the therapeutic potential of DLE from male and female donors on D. melanogaster LRRK2 loss-of-function, as compared to D. melanogaster wild-type (WT), in terms of rescuing physiological parameters, such as motor and climbing activities, which are severely compromised in the mutant flies. Finally, in search of the anatomical structures responsible for restored functions in parkinsonian-like mutant flies, we found a topographical correlation between improvement of locomotor performances and an increased number of dopaminergic neurons in selective areas of LRRK2 mutant brains.
Collapse
Affiliation(s)
- Andrea Diana
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy;
- Correspondence: (A.D.); (M.D.S.); Tel.: +39-070-6754054/56 (A.D.); +39-070-6754183 (M.D.S.)
| | - Maria Collu
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy;
| | - Maria Antonietta Casu
- CNR Institute of Translational Pharmacology, 09010 Pula/Cagliari, Italy; (M.A.C.); (I.M.)
| | - Ignazia Mocci
- CNR Institute of Translational Pharmacology, 09010 Pula/Cagliari, Italy; (M.A.C.); (I.M.)
| | | | - Maria Dolores Setzu
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy;
- Correspondence: (A.D.); (M.D.S.); Tel.: +39-070-6754054/56 (A.D.); +39-070-6754183 (M.D.S.)
| |
Collapse
|
23
|
Tamtaji OR, Reiter RJ, Alipoor R, Dadgostar E, Kouchaki E, Asemi Z. Melatonin and Parkinson Disease: Current Status and Future Perspectives for Molecular Mechanisms. Cell Mol Neurobiol 2020; 40:15-23. [PMID: 31388798 PMCID: PMC11448849 DOI: 10.1007/s10571-019-00720-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 07/31/2019] [Indexed: 12/29/2022]
Abstract
Parkinson disease (PD) is a chronic and neurodegenerative disease with motor and nonmotor symptoms. Multiple pathways are involved in the pathophysiology of PD, including apoptosis, autophagy, oxidative stress, inflammation, α-synuclein aggregation, and changes in the neurotransmitters. Preclinical and clinical studies have shown that melatonin supplementation is an appropriate therapy for PD. Administration of melatonin leads to inhibition of some pathways related to apoptosis, autophagy, oxidative stress, inflammation, α-synuclein aggregation, and dopamine loss in PD. In addition, melatonin improves some nonmotor symptom in patients with PD. Limited studies, however, have evaluated the role of melatonin on molecular mechanisms and clinical symptoms in PD. This review summarizes what is known regarding the impact of melatonin on PD in preclinical and clinical studies.
Collapse
Affiliation(s)
- Omid Reza Tamtaji
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health San Antonio, San Antonio, TX, USA
| | - Reza Alipoor
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Islamic Republic of Iran
| | | | - Ebrahim Kouchaki
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| |
Collapse
|
24
|
Trist BG, Hare DJ, Double KL. Oxidative stress in the aging substantia nigra and the etiology of Parkinson's disease. Aging Cell 2019; 18:e13031. [PMID: 31432604 PMCID: PMC6826160 DOI: 10.1111/acel.13031] [Citation(s) in RCA: 440] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/05/2019] [Accepted: 08/07/2019] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease prevalence is rapidly increasing in an aging global population. With this increase comes exponentially rising social and economic costs, emphasizing the immediate need for effective disease‐modifying treatments. Motor dysfunction results from the loss of dopaminergic neurons in the substantia nigra pars compacta and depletion of dopamine in the nigrostriatal pathway. While a specific biochemical mechanism remains elusive, oxidative stress plays an undeniable role in a complex and progressive neurodegenerative cascade. This review will explore the molecular factors that contribute to the high steady‐state of oxidative stress in the healthy substantia nigra during aging, and how this chemical environment renders neurons susceptible to oxidative damage in Parkinson's disease. Contributing factors to oxidative stress during aging and as a pathogenic mechanism for Parkinson's disease will be discussed within the context of how and why therapeutic approaches targeting cellular redox activity in this disorder have, to date, yielded little therapeutic benefit. We present a contemporary perspective on the central biochemical contribution of redox imbalance to Parkinson's disease etiology and argue that improving our ability to accurately measure oxidative stress, dopaminergic neurotransmission and cell death pathways in vivo is crucial for both the development of new therapies and the identification of novel disease biomarkers.
Collapse
Affiliation(s)
- Benjamin G. Trist
- Brain and Mind Centre and Discipline of Pharmacology, Faculty of Medical and Health The University of Sydney Sydney NSW Australia
| | - Dominic J. Hare
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Vic. Australia
- Elemental Bio‐imaging Facility University of Technology Sydney Broadway NSW Australia
| | - Kay L. Double
- Brain and Mind Centre and Discipline of Pharmacology, Faculty of Medical and Health The University of Sydney Sydney NSW Australia
| |
Collapse
|
25
|
Kim J, Daadi MM. Non-cell autonomous mechanism of Parkinson's disease pathology caused by G2019S LRRK2 mutation in Ashkenazi Jewish patient: Single cell analysis. Brain Res 2019; 1722:146342. [PMID: 31330122 PMCID: PMC8152577 DOI: 10.1016/j.brainres.2019.146342] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 07/17/2019] [Accepted: 07/18/2019] [Indexed: 10/26/2022]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, characterized by the loss of the midbrain dopaminergic neurons, which leads to impaired motor and cognitive functions. PD is predominantly an idiopathic disease, however about 5% of cases are linked to hereditary mutations. The most common mutation in both familial and sporadic PD is the G2019S mutation of leucine-rich repeat kinase 2 (LRRK2) with high prevalence in Ashkenazi Jewish patients and in North African Berber and Arab patients. It is still not fully understood how this mutation leads to PD pathology. In this study, we derived induced pluripotent stem cells (iPSCs) from an Ashkenazi Jewish patient with G2019S LRRK2 mutation to isolate self-renewable multipotent neural stem cells (NSCs) and to model this form of PD in vitro. To investigate the cellular diversity and disease pathology in the NSCs, we used single cell RNA-seq transcriptomic profiling. The evidence suggests there are three subpopulations within the NSCs: a committed neuronal population, intermediate stage population and undifferentiated stage population. Unbiased single-cell transcriptomic analysis revealed differential expression and dysregulation of genes involved in PD pathology. The significantly affected genes were involved in mitochondrial function, DNA repair, protein degradation, oxidative stress, lysosome biogenesis, ubiquitination, endosome function, autophagy and mitochondrial quality control. The results suggest that G2019S LRRK2 mutation may affect multiple cell types in a non-cell autonomous mechanism of PD pathology and that unbiased single-cell transcriptomics holds promise for personalized medicine.
Collapse
Affiliation(s)
- Jeffrey Kim
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States; Department of Cell Systems & Anatomy, TX, United States
| | - Marcel M Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States; Department of Cell Systems & Anatomy, TX, United States; Department of Radiology, University of Texas Health Science Center at San Antonio, TX, United States.
| |
Collapse
|
26
|
Mohajeri M, Martín-Jiménez C, Barreto GE, Sahebkar A. Effects of estrogens and androgens on mitochondria under normal and pathological conditions. Prog Neurobiol 2019; 176:54-72. [DOI: 10.1016/j.pneurobio.2019.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 02/23/2019] [Accepted: 03/05/2019] [Indexed: 02/06/2023]
|
27
|
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a large multidomain protein with both a Ras of complex (ROC) domain and a kinase domain (KD) and, therefore, exhibits both GTPase and kinase activities. Human genetics studies have linked LRRK2 as a major genetic contributor to familial and sporadic Parkinson's disease (PD), a neurodegenerative movement disorder that inflicts millions worldwide. The C-terminal region of LRRK2 is a Trp-Asp-40 (WD40) domain with poorly defined biological functions but has been implicated in microtubule interaction. Here, we present the crystal structure of the WD40 domain of human LRRK2 at 2.6-Å resolution, which reveals a seven-bladed WD40 fold. The structure displays a dimeric assembly in the crystal, which we further confirm by measurements in solution. We find that structure-based and PD-associated disease mutations in the WD40 domain including the common G2385R polymorphism mainly compromise dimer formation. Assessment of full-length LRRK2 kinase activity by measuring phosphorylation of Rab10, a member of the family of Rab GTPases known to be important kinase substrates of LRRK2, shows enhancement of kinase activity by several dimerization-defective mutants including G2385R, although dimerization impairment does not always result in kinase activation. Furthermore, mapping of phylogenetically conserved residues onto the WD40 domain structure reveals surface patches that may be important for additional functions of LRRK2. Collectively, our analyses provide insights for understanding the structures and functions of LRRK2 and suggest the potential utility of LRRK2 kinase inhibitors in treating PD patients with WD40 domain mutations.
Collapse
|
28
|
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: 92] [Impact Index Per Article: 15.3] [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
|
29
|
Kishore A, Ashok Kumar Sreelatha A, Sturm M, von-Zweydorf F, Pihlstrøm L, Raimondi F, Russell R, Lichtner P, Banerjee M, Krishnan S, Rajan R, Puthenveedu DK, Chung SJ, Bauer P, Riess O, Gloeckner CJ, Kruger R, Gasser T, Sharma M. Understanding the role of genetic variability in LRRK2 in Indian population. Mov Disord 2018; 34:496-505. [PMID: 30485545 DOI: 10.1002/mds.27558] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/24/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Genetic variability in LRRK2 has been unequivocally established as a major risk factor for familial and sporadic forms of PD in ethnically diverse populations. OBJECTIVES To resolve the role of LRRK2 in the Indian population. METHODS We performed targeted resequencing of the LRRK2 locus in 288 cases and 298 controls and resolved the haplotypic structure of LRRK2 in a combined cohort of 800 cases and 402 controls in the Indian population. We assessed the frequency of novel missense variants in the white and East Asian population by leveraging exome sequencing and densely genotype data, respectively. We did computational modeling and biochemical approach to infer the potential role of novel variants impacting the LRRK2 protein function. Finally, we assessed the phosphorylation activity of identified novel coding variants in the LRRK2 gene. RESULTS We identified four novel missense variants with frequency ranging from 0.0008% to 0.002% specific for the Indian population, encompassing armadillo and kinase domains of the LRRK2 protein. A common genetic variability within LRRK2 may contribute to increased risk, but it was nonsignificant after correcting for multiple testing, because of small cohort size. The computational modeling showed destabilizing effect on the LRRK2 function. In comparison to the wild-type, the kinase domain variant showed 4-fold increase in the kinase activity. CONCLUSIONS Our study, for the first time, identified novel missense variants for LRRK2, specific for the Indian population, and showed that a novel missense variant in the kinase domain modifies kinase activity in vitro. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Asha Kishore
- Sree Chitra Tirunal Institute for Medical Science and Technology, Kerala, India
| | - Ashwin Ashok Kumar Sreelatha
- Centre for Genetic Epidemiology, Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Tübingen, Germany
| | - Marc Sturm
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Felix von-Zweydorf
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | | | - Rob Russell
- Cell Networks, University of Heidelberg, Heidelberg, Germany
| | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | | | - Syam Krishnan
- Sree Chitra Tirunal Institute for Medical Science and Technology, Kerala, India
| | - Roopa Rajan
- Sree Chitra Tirunal Institute for Medical Science and Technology, Kerala, India.,All India Institute for Medical Sciences, New Delhi, India
| | | | - Sun Ju Chung
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | - Peter Bauer
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Christian Johannes Gloeckner
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Rejko Kruger
- Center of Neurology, and Hertie Institute for Clinical Brain Research, University Hospital, Tübingen, Germany.,LCSB, Luxembourg Centre for Systems Biology, University of Luxembourg, and Centre Hospitalier de Luxembourg (CHL), Luxembourg
| | - Thomas Gasser
- Center of Neurology, and Hertie Institute for Clinical Brain Research, University Hospital, Tübingen, Germany
| | - Manu Sharma
- Centre for Genetic Epidemiology, Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Tübingen, Germany
| |
Collapse
|
30
|
Cao J, Zhuang Y, Zhang J, Zhang Z, Yuan S, Zhang P, Li H, Li X, Shen H, Wang Z, Chen G. Leucine-rich repeat kinase 2 aggravates secondary brain injury induced by intracerebral hemorrhage in rats by regulating the P38 MAPK/Drosha pathway. Neurobiol Dis 2018; 119:53-64. [DOI: 10.1016/j.nbd.2018.07.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/03/2018] [Accepted: 07/20/2018] [Indexed: 12/28/2022] Open
|
31
|
Abstract
LRRK2 mutations are associated with the loss of neurons, that is to say toxicity, in patients and in experimental model systems. However, the mechanisms by which mutations can be linked to neurodegeneration are not fully defined. Here I will argue that mechanism in this context encompasses a variety of levels of information. Mutations or alterations in gene expression at a genetic level are one set of mechanisms that are reflected at the biochemical level likely in enhanced or persistent function of LRRK2. By impacting cellular pathways, prominently including changes in autophagy but also microtubule function, mitochondria and protein synthesis, in neurons and immune cells, the LRRK2 brain is primed for neurodegeneration in an age-dependent manner. These concepts have implications for not only modeling LRRK2 disease but also perhaps for Parkinson's disease more generally, including the development of therapeutic modalities.
Collapse
|
32
|
Chen YH, Huang EYK, Kuo TT, Miller J, Chiang YH, Hoffer BJ. Impact of Traumatic Brain Injury on Dopaminergic Transmission. Cell Transplant 2018; 26:1156-1168. [PMID: 28933212 PMCID: PMC5657731 DOI: 10.1177/0963689717714105] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Brain trauma is often associated with severe morbidity and is a major public health concern. Even when injury is mild and no obvious anatomic disruption is seen, many individuals suffer disabling neuropsychological impairments such as memory loss, mood dysfunction, substance abuse, and adjustment disorder. These changes may be related to subtle disruption of neural circuits as well as functional changes at the neurotransmitter level. In particular, there is considerable evidence that dopamine (DA) physiology in the nigrostriatal and mesocorticolimbic pathways might be impaired after traumatic brain injury (TBI). Alterations in DA levels can lead to oxidative stress and cellular dysfunction, and DA plays an important role in central nervous system inflammation. Therapeutic targeting of DA pathways may offer benefits for both neuronal survival and functional outcome after TBI. The purpose of this review is to discuss the role of DA pathology in acute TBI and the potential impact of therapies that target these systems for the treatment of TBI.
Collapse
Affiliation(s)
- Yuan-Hao Chen
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Yuan-Hao Chen, Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, 4F, No. 325, 2nd Sec., Cheng-Kung Road, Nei-Hu District, Taipei City, 114 Taiwan, Republic of China.
| | - Eagle Yi-Kung Huang
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Tung-Tai Kuo
- Graduate Institute of Computer and Communication Engineering, National Taipei University of Technology, Taipei, Taiwan, Republic of China
| | - Jonathan Miller
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yung-Hsiao Chiang
- Section of Neurosurgery, Department of Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Barry J. Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
33
|
Antoniou N, Vlachakis D, Memou A, Leandrou E, Valkimadi PE, Melachroinou K, Re DB, Przedborski S, Dauer WT, Stefanis L, Rideout HJ. A motif within the armadillo repeat of Parkinson's-linked LRRK2 interacts with FADD to hijack the extrinsic death pathway. Sci Rep 2018; 8:3455. [PMID: 29472595 PMCID: PMC5823876 DOI: 10.1038/s41598-018-21931-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/07/2018] [Indexed: 01/15/2023] Open
Abstract
In experimental models, both in vivo and cellular, over-expression of Parkinson’s linked mutant leucine-rich repeat kinase 2 (LRRK2) is sufficient to induce neuronal death. While several cell death associated proteins have been linked to LRRK2, either as protein interactors or as putative substrates, characterization of the neuronal death cascade remains elusive. In this study, we have mapped for the first time the domain within LRRK2 that mediates the interaction with FADD, thereby activating the molecular machinery of the extrinsic death pathway. Using homology modeling and molecular docking approaches, we have identified a critical motif within the N-terminal armadillo repeat region of LRRK2. Moreover, we show that co-expression of fragments of LRRK2 that contain the FADD binding motif, or deletion of this motif itself, blocks the interaction with FADD, and is neuroprotective. We further demonstrate that downstream of FADD, the mitochondrial proteins Bid and Bax are recruited to the death cascade and are necessary for neuronal death. Our work identifies multiple novel points within neuronal death signaling pathways that could potentially be targeted by candidate therapeutic strategies and highlight how the extrinsic pathway can be activated intracellularly in a pathogenic context.
Collapse
Affiliation(s)
- Nasia Antoniou
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Dimitrios Vlachakis
- Computational Biology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Anna Memou
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Emmanouela Leandrou
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Polytimi-Eleni Valkimadi
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Katerina Melachroinou
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Diane B Re
- Department of Environmental Health Sciences, Columbia University, New York, NY, USA
| | - Serge Przedborski
- Department of Neurology/Motor Neuron Center, Columbia University, New York, NY, USA
| | - William T Dauer
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Leonidas Stefanis
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Second Department of Neurology, University of Athens Medical School, Athens, Greece
| | - Hardy J Rideout
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
34
|
Understanding Miro GTPases: Implications in the Treatment of Neurodegenerative Disorders. Mol Neurobiol 2018; 55:7352-7365. [PMID: 29411264 PMCID: PMC6096957 DOI: 10.1007/s12035-018-0927-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/24/2018] [Indexed: 12/19/2022]
Abstract
The Miro GTPases represent an unusual subgroup of the Ras superfamily and have recently emerged as important mediators of mitochondrial dynamics and for maintaining neuronal health. It is now well-established that these enzymes act as essential components of a Ca2+-sensitive motor complex, facilitating the transport of mitochondria along microtubules in several cell types, including dopaminergic neurons. The Miros appear to be critical for both anterograde and retrograde mitochondrial transport in axons and dendrites, both of which are considered essential for neuronal health. Furthermore, the Miros may be significantly involved in the development of several serious pathological processes, including the development of neurodegenerative and psychiatric disorders. In this review, we discuss the molecular structure and known mitochondrial functions of the Miro GTPases in humans and other organisms, in the context of neurodegenerative disease. Finally, we consider the potential human Miros hold as novel therapeutic targets for the treatment of such disease.
Collapse
|
35
|
Chen Z, Cao Z, Zhang W, Gu M, Zhou ZD, Li B, Li J, Tan EK, Zeng L. LRRK2 interacts with ATM and regulates Mdm2–p53 cell proliferation axis in response to genotoxic stress. Hum Mol Genet 2017; 26:4494-4505. [DOI: 10.1093/hmg/ddx337] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/06/2017] [Indexed: 01/29/2023] Open
|
36
|
Neuronal death signaling pathways triggered by mutant LRRK2. Biochem Soc Trans 2017; 45:123-129. [PMID: 28202665 DOI: 10.1042/bst20160256] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/11/2016] [Accepted: 10/14/2016] [Indexed: 11/17/2022]
Abstract
Autosomal dominantly inherited mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinson's disease. While considerable progress has been made in understanding its function and the many different cellular activities in which it participates, a clear understanding of the mechanism(s) of the induction of neuronal death by mutant forms of LRRK2 remains elusive. Although several in vivo models have documented the progressive loss of dopaminergic neurons of the substantia nigra, more complete interrogations of the modality of neuronal death have been gained from cellular models. Overexpression of mutant LRRK2 in neuronal-like cell lines or in primary neurons induces an apoptotic type of cell death involving components of the extrinsic as well as intrinsic death pathways. While informative, these studies are limited by their reliance upon isolated neuronal cells; and the pathways triggered by mutant LRRK2 in neurons may be further refined or modulated by extracellular signals. Nevertheless, the identification of specific cell death-associated signaling events set in motion by the dominant action of mutant LRRK2, the loss of an inhibitory function of wild-type LRRK2, or a combination of the two, expands the landscape of potential therapeutic targets for future intervention in the clinic.
Collapse
|
37
|
Rassu M, Del Giudice MG, Sanna S, Taymans JM, Morari M, Brugnoli A, Frassineti M, Masala A, Esposito S, Galioto M, Valle C, Carri MT, Biosa A, Greggio E, Crosio C, Iaccarino C. Role of LRRK2 in the regulation of dopamine receptor trafficking. PLoS One 2017; 12:e0179082. [PMID: 28582422 PMCID: PMC5459500 DOI: 10.1371/journal.pone.0179082] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 05/23/2017] [Indexed: 11/18/2022] Open
Abstract
Mutations in LRRK2 play a critical role in both familial and sporadic Parkinson’s disease (PD). Up to date, the role of LRRK2 in PD onset and progression remains largely unknown. However, experimental evidence highlights a critical role of LRRK2 in the control of vesicle trafficking that in turn may regulate different aspects of neuronal physiology. We have analyzed the role of LRRK2 in regulating dopamine receptor D1 (DRD1) and D2 (DRD2) trafficking. DRD1 and DRD2 are the most abundant dopamine receptors in the brain. They differ in structural, pharmacological and biochemical properties, as well as in localization and internalization mechanisms. Our results indicate that disease-associated mutant G2019S LRRK2 impairs DRD1 internalization, leading to an alteration in signal transduction. Moreover, the mutant forms of LRRK2 affect receptor turnover by decreasing the rate of DRD2 trafficking from the Golgi complex to the cell membrane. Collectively, our findings are consistent with the conclusion that LRRK2 influences the motility of neuronal vesicles and the neuronal receptor trafficking. These findings have important implications for the complex role that LRRK2 plays in neuronal physiology and the possible pathological mechanisms that may lead to neuronal death in PD.
Collapse
Affiliation(s)
- Mauro Rassu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | | | - Simona Sanna
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Jean Marc Taymans
- UMR-S1172, Jean-Pierre Aubert Research Center (Inserm – Université de Lille – CHRU de Lille), Lille, France
| | - Michele Morari
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy and National Institute for Neuroscience, Ferrara, Italy
| | - Alberto Brugnoli
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy and National Institute for Neuroscience, Ferrara, Italy
| | - Martina Frassineti
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy and National Institute for Neuroscience, Ferrara, Italy
| | - Alessandra Masala
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Sonia Esposito
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Manuela Galioto
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Cristiana Valle
- Fondazione Santa Lucia, IRCCS, Rome, Italy
- Institute of Cell Biology and Neurobiology, IBCN, CNR, Rome, Italy
| | - Maria Teresa Carri
- Fondazione Santa Lucia, IRCCS, Rome, Italy
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Alice Biosa
- Department of Biology, University of Padova, Padova, Italy
| | - Elisa Greggio
- Department of Biology, University of Padova, Padova, Italy
| | - Claudia Crosio
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Ciro Iaccarino
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- * E-mail:
| |
Collapse
|
38
|
Adeosun SO, Hou X, Zheng B, Melrose HL, Mosley T, Wang JM. Human LRRK2 G2019S mutation represses post-synaptic protein PSD95 and causes cognitive impairment in transgenic mice. Neurobiol Learn Mem 2017; 142:182-189. [PMID: 28487191 DOI: 10.1016/j.nlm.2017.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 05/03/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND LRRK2 G2019S mutation is associated with increased kinase activity and is the most common mutation associated with late-onset PD. However, the transgenic mouse model has not recapitulated cardinal PD-related motor phenotypes. Non-motor symptoms of PD including cognitive impairments are very common and may appear earlier than the motor symptoms. The objective of this study was to determine whether human LRRK2 with G2019S mutation causes hippocampus-dependent cognitive deficits in mice. RESULTS Male (LRRK2-G2019S) LRRK2-Tg mice showed impairments in the early portion of the Two-day radial arm water maze acquisition trial as well as in the reversal learning on the third day. However, their performance was similar to Non-Tg controls in the probe trial. LRRK2-Tg mice also displayed impairments in the novel arm discrimination test but not in the spontaneous alternation test in Y-maze. Interestingly, there was no statistically significant locomotor impairment during any of these cognitive test, nor in the locomotor tests including open field, accelerating rotarod and pole tests. Expression of the postsynaptic protein PSD-95 but not the presynaptic protein synaptophysin was lower in hippocampal homogenates of LRRK2-Tg mice. CONCLUSION Consistent with previous reports in human LRRK2 G2019S carriers, the current data suggests that cognitive dysfunctions are present in LRRK2-Tg mice even in the absence of locomotor impairment. LRRK2 G2019S mutation represses the postsynaptic protein PSD-95 but not the presynaptic protein synaptophysin. This study also suggests that mild cognitive impairment may appear earlier than motor dysfunctions in LRRK2-G2019S mutation carriers.
Collapse
Affiliation(s)
- Samuel O Adeosun
- Department of Pathology, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, United States.
| | - Xu Hou
- Department of Pathology, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, United States.
| | - Baoying Zheng
- Department of Pathology, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, United States.
| | - Heather L Melrose
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, United States.
| | - Thomas Mosley
- Department of Medicine, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, United States.
| | - Jun Ming Wang
- Department of Pathology, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, United States.
| |
Collapse
|
39
|
Giannoccaro MP, La Morgia C, Rizzo G, Carelli V. Mitochondrial DNA and primary mitochondrial dysfunction in Parkinson's disease. Mov Disord 2017; 32:346-363. [PMID: 28251677 DOI: 10.1002/mds.26966] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/27/2017] [Accepted: 01/30/2017] [Indexed: 12/15/2022] Open
Abstract
In 1979, it was observed that parkinsonism could be induced by a toxin inhibiting mitochondrial respiratory complex I. This initiated the long-standing hypothesis that mitochondrial dysfunction may play a key role in the pathogenesis of Parkinson's disease (PD). This hypothesis evolved, with accumulating evidence pointing to complex I dysfunction, which could be caused by environmental or genetic factors. Attention was focused on the mitochondrial DNA, considering the occurrence of mutations, polymorphic haplogroup-specific variants, and defective mitochondrial DNA maintenance with the accumulation of multiple deletions and a reduction of copy number. Genetically determined diseases of mitochondrial DNA maintenance frequently manifest with parkinsonism, but the age-related accumulation of somatic mitochondrial DNA errors also represents a major driving mechanism for PD. Recently, the discovery of the genetic cause of rare inherited forms of PD highlighted an extremely complex homeostatic control over mitochondria, involving their dynamic fission/fusion cycle, the balancing of mitobiogenesis and mitophagy, and consequently the quality control surveillance that corrects faulty mitochondrial DNA maintenance. Many genes came into play, including the PINK1/parkin axis, but also OPA1, as pieces of the same puzzle, together with mitochondrial DNA damage, complex I deficiency and increased oxidative stress. The search for answers will drive future research to reach the understanding necessary to provide therapeutic options directed not only at limiting the clinical evolution of symptoms but also finally addressing the pathogenic mechanisms of neurodegeneration in PD. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Maria Pia Giannoccaro
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Chiara La Morgia
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giovanni Rizzo
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Valerio Carelli
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
40
|
Modulation of ARTS and XIAP by Parkin Is Associated with Carnosic Acid Protects SH-SY5Y Cells against 6-Hydroxydopamine-Induced Apoptosis. Mol Neurobiol 2017; 55:1786-1794. [DOI: 10.1007/s12035-017-0443-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/03/2017] [Indexed: 01/27/2023]
|
41
|
Rideout HJ, Re DB. LRRK2 and the "LRRKtosome" at the Crossroads of Programmed Cell Death: Clues from RIP Kinase Relatives. ADVANCES IN NEUROBIOLOGY 2017; 14:193-208. [PMID: 28353285 DOI: 10.1007/978-3-319-49969-7_10] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since its cloning and identification in 2004, considerable gains have been made in the understanding of the basic functionality of leucine-rich repeat kinase 2 (LRRK2), including its kinase and GTPase activities, its protein interactors and subcellular localization, and its expression in the CNS and peripheral tissues. However, the mechanism(s) by which expression of mutant forms of LRRK2 lead to the death of dopaminergic neurons of the ventral midbrain remains largely uncharacterized. Because of its complex domain structure, LRRK2 exhibits similarities with multiple protein families including ROCO proteins, as well as the RIP kinases. Cellular models in which mutant LRRK2 is overexpressed in neuronal-like cell lines or in primary neurons have found evidence of apoptotic cell death involving components of the extrinsic as well as intrinsic death pathways. However, since the expression of LRRK2 is comparatively quite low in ventral midbrain dopaminergic neurons, the possibility exists that non-cell autonomous signaling also contributes to the loss of these neurons. In this chapter, we will discuss the different neuronal death pathways that may be activated by mutant forms of LRRK2, guided in part by the behavior of other members of the RIP kinase protein family.
Collapse
Affiliation(s)
- Hardy J Rideout
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Soranou Efessiou 4, Athens, 115 27, Greece.
| | - Diane B Re
- EHS Department and Motor Neuron Center, Columbia University, 722 W 168th Street Suite 1107-b, New York, NY, 10032, USA
| |
Collapse
|
42
|
Kang UB, Marto JA. Leucine-rich repeat kinase 2 and Parkinson's disease. Proteomics 2016; 17. [PMID: 27723254 DOI: 10.1002/pmic.201600092] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/13/2016] [Accepted: 10/06/2016] [Indexed: 12/21/2022]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a large multidomain protein that is expressed in many tissues and participates in numerous biological pathways. Mutations in LRRK2 are recognized as genetic risk factors for familial Parkinson's disease (PD) and may also represent causal factors in the more common sporadic form of PD. The structure of LRRK2 comprises a combination of GTPase, kinase, and scaffolding domains. This functional diversity, combined with a potentially central role in genetic and idiopathic PD motivates significant effort to further credential LRRK2 as a therapeutic target. Here, we review the current understanding for LRRK2 function in normal physiology and PD, with emphasis on insight gained from proteomic approaches.
Collapse
Affiliation(s)
- Un-Beom Kang
- Department of Cancer Biology and Blais Proteomics Center, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Jarrod A Marto
- Department of Cancer Biology and Blais Proteomics Center, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
43
|
Requejo-Aguilar R, Bolaños JP. Mitochondrial control of cell bioenergetics in Parkinson's disease. Free Radic Biol Med 2016; 100:123-137. [PMID: 27091692 PMCID: PMC5065935 DOI: 10.1016/j.freeradbiomed.2016.04.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 12/15/2022]
Abstract
Parkinson disease (PD) is a neurodegenerative disorder characterized by a selective loss of dopaminergic neurons in the substantia nigra. The earliest biochemical signs of the disease involve failure in mitochondrial-endoplasmic reticulum cross talk and lysosomal function, mitochondrial electron chain impairment, mitochondrial dynamics alterations, and calcium and iron homeostasis abnormalities. These changes are associated with increased mitochondrial reactive oxygen species (mROS) and energy deficiency. Recently, it has been reported that, as an attempt to compensate for the mitochondrial dysfunction, neurons invoke glycolysis as a low-efficient mode of energy production in models of PD. Here, we review how mitochondria orchestrate the maintenance of cellular energetic status in PD, with special focus on the switch from oxidative phosphorylation to glycolysis, as well as the implication of endoplasmic reticulum and lysosomes in the control of bioenergetics.
Collapse
Affiliation(s)
- Raquel Requejo-Aguilar
- Department of Biochemistry and Molecular Biology, University of Cordoba, Institute Maimonides of Biomedical Investigation of Cordoba (IMIBIC), Cordoba, Spain
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), University of Salamanca-CSIC, Zacarias Gonzalez, 2, 37007 Salamanca, Spain.
| |
Collapse
|
44
|
Murdoch JD, Rostosky CM, Gowrisankaran S, Arora AS, Soukup SF, Vidal R, Capece V, Freytag S, Fischer A, Verstreken P, Bonn S, Raimundo N, Milosevic I. Endophilin-A Deficiency Induces the Foxo3a-Fbxo32 Network in the Brain and Causes Dysregulation of Autophagy and the Ubiquitin-Proteasome System. Cell Rep 2016; 17:1071-1086. [PMID: 27720640 PMCID: PMC5080600 DOI: 10.1016/j.celrep.2016.09.058] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 06/24/2016] [Accepted: 09/19/2016] [Indexed: 12/21/2022] Open
Abstract
Endophilin-A, a well-characterized endocytic adaptor essential for synaptic vesicle recycling, has recently been linked to neurodegeneration. We report here that endophilin-A deficiency results in impaired movement, age-dependent ataxia, and neurodegeneration in mice. Transcriptional analysis of endophilin-A mutant mice, complemented by proteomics, highlighted ataxia- and protein-homeostasis-related genes and revealed upregulation of the E3-ubiquitin ligase FBXO32/atrogin-1 and its transcription factor FOXO3A. FBXO32 overexpression triggers apoptosis in cultured cells and neurons but, remarkably, coexpression of endophilin-A rescues it. FBXO32 interacts with all three endophilin-A proteins. Similarly to endophilin-A, FBXO32 tubulates membranes and localizes on clathrin-coated structures. Additionally, FBXO32 and endophilin-A are necessary for autophagosome formation, and both colocalize transiently with autophagosomes. Our results point to a role for endophilin-A proteins in autophagy and protein degradation, processes that are impaired in their absence, potentially contributing to neurodegeneration and ataxia. Endophilin-A is needed for autophagosome formation in mammalian neurons and brain Absence of endophilin-A upregulates the E3-ubiquitin ligase FBXO32 FBXO32-endophilin-A interaction maintains neuronal health and protein homeostasis Endophilin-A KO mice show age-dependent ataxia, motor impairments, and neurodegeneration
Collapse
Affiliation(s)
- John D Murdoch
- European Neuroscience Institute (ENI), 37077 Göttingen, Germany; Institute of Cellular Biochemistry, University Medical Center Göttingen (UMG), 37073 Göttingen, Germany
| | | | | | | | - Sandra-Fausia Soukup
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven Department of Human Genetics, Leuven Institute for Neurodegenerative Disease (LIND), 3000 Leuven, Belgium
| | - Ramon Vidal
- Computational Systems Biology, German Center for Neurodegenerative Diseases (DZNE), 37077 Göttingen, Germany
| | - Vincenzo Capece
- Computational Systems Biology, German Center for Neurodegenerative Diseases (DZNE), 37077 Göttingen, Germany
| | - Siona Freytag
- European Neuroscience Institute (ENI), 37077 Göttingen, Germany
| | - Andre Fischer
- Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), 37077 Göttingen, Germany; Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37077 Göttingen, Germany
| | - Patrik Verstreken
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven Department of Human Genetics, Leuven Institute for Neurodegenerative Disease (LIND), 3000 Leuven, Belgium
| | - Stefan Bonn
- Computational Systems Biology, German Center for Neurodegenerative Diseases (DZNE), 37077 Göttingen, Germany
| | - Nuno Raimundo
- Institute of Cellular Biochemistry, University Medical Center Göttingen (UMG), 37073 Göttingen, Germany.
| | - Ira Milosevic
- European Neuroscience Institute (ENI), 37077 Göttingen, Germany.
| |
Collapse
|
45
|
Bose A, Beal MF. Mitochondrial dysfunction in Parkinson's disease. J Neurochem 2016; 139 Suppl 1:216-231. [PMID: 27546335 DOI: 10.1111/jnc.13731] [Citation(s) in RCA: 599] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 07/04/2016] [Accepted: 07/06/2016] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. About 2% of the population above the age of 60 is affected by the disease. The pathological hallmarks of the disease include the loss of dopaminergic neurons in the substantia nigra and the presence of Lewy bodies that are made of α-synuclein. Several theories have been suggested for the pathogenesis of PD, of which mitochondrial dysfunction plays a pivotal role in both sporadic and familial forms of the disease. Dysfunction of the mitochondria that is caused by bioenergetic defects, mutations in mitochondrial DNA, nuclear DNA gene mutations linked to mitochondria, and changes in dynamics of the mitochondria such fusion or fission, changes in size and morphology, alterations in trafficking or transport, altered movement of mitochondria, impairment of transcription, and the presence of mutated proteins associated with mitochondria are implicated in PD. In this review, we provide a detailed overview of the mechanisms that can cause mitochondrial dysfunction in PD. We bring to the forefront, new signaling pathways such as the retromer-trafficking pathway and its implication in the disease and also provide a brief overview of therapeutic strategies to improve mitochondrial defects in PD. Bioenergetic defects, mutations in mitochondrial DNA, nuclear DNA gene mutations, alterations in mitochondrial dynamics, alterations in trafficking/transport and mitochondrial movement, abnormal size and morphology, impairment of transcription and the presence of mutated proteins associated with mitochondria are implicated in PD. In this review, we focus on the mechanisms underlying mitochondrial dysfunction in PD and bring to the forefront new signaling pathways that may be involved in PD. We also provide an overview of therapeutic strategies to improve mitochondrial defects in PD. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Anindita Bose
- Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York City, New York, USA.
| | - M Flint Beal
- Brain and Mind Research Institute, Weill Cornell Medicine, New York City, New York, USA.
| |
Collapse
|
46
|
Activation Mechanism of LRRK2 and Its Cellular Functions in Parkinson's Disease. PARKINSONS DISEASE 2016; 2016:7351985. [PMID: 27293958 PMCID: PMC4880697 DOI: 10.1155/2016/7351985] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 04/19/2016] [Indexed: 01/09/2023]
Abstract
Human LRRK2 (Leucine-Rich Repeat Kinase 2) has been associated with both familial and idiopathic Parkinson's disease (PD). Although several LRRK2 mediated pathways and interaction partners have been identified, the cellular functions of LRRK2 and LRRK2 mediated progression of PD are still only partially understood. LRRK2 belongs to the group of Roco proteins which are characterized by the presence of a Ras-like G-domain (Roc), a C-terminal of Roc domain (COR), a kinase, and several protein-protein interaction domains. Roco proteins exhibit a complex activation mechanism involving intramolecular signaling, dimerization, and substrate/effector binding. Importantly, PD mutations in LRRK2 have been linked to a decreased GTPase and impaired kinase activity, thus providing putative therapeutic targets. To fully explore these potential targets it will be crucial to understand the function and identify the pathways responsible for LRRK2-linked PD. Here, we review the recent progress in elucidating the complex LRRK2 activation mechanism, describe the accumulating evidence that link LRRK2-mediated PD to mitochondrial dysfunction and aberrant autophagy, and discuss possible ways for therapeutically targeting LRRK2.
Collapse
|
47
|
De Rose F, Marotta R, Poddighe S, Talani G, Catelani T, Setzu MD, Solla P, Marrosu F, Sanna E, Kasture S, Acquas E, Liscia A. Functional and Morphological Correlates in the Drosophila LRRK2 loss-of-function Model of Parkinson's Disease: Drug Effects of Withania somnifera (Dunal) Administration. PLoS One 2016; 11:e0146140. [PMID: 26727265 PMCID: PMC4699764 DOI: 10.1371/journal.pone.0146140] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/14/2015] [Indexed: 12/14/2022] Open
Abstract
The common fruit fly Drosophila melanogaster (Dm) is a simple animal species that contributed significantly to the development of neurobiology whose leucine-rich repeat kinase 2 mutants (LRRK2) loss-of-function in the WD40 domain represent a very interesting tool to look into physiopathology of Parkinson's disease (PD). Accordingly, LRRK2 Dm have also the potential to contribute to reveal innovative therapeutic approaches to its treatment. Withania somnifera Dunal, a plant that grows spontaneously also in Mediterranean regions, is known in folk medicine for its anti-inflammatory and protective properties against neurodegeneration. The aim of this study was to evaluate the neuroprotective effects of its standardized root methanolic extract (Wse) on the LRRK2 loss-of-function Dm model of PD. To this end mutant and wild type (WT) flies were administered Wse, through diet, at different concentrations as larvae and adults (L+/A+) or as adults (L-/A+) only. LRRK2 mutants have a significantly reduced lifespan and compromised motor function and mitochondrial morphology compared to WT flies 1% Wse-enriched diet, administered to Dm LRRK2 as L-/A+and improved a) locomotor activity b) muscle electrophysiological response to stimuli and also c) protected against mitochondria degeneration. In contrast, the administration of Wse to Dm LRRK2 as L+/A+, no matter at which concentration, worsened lifespan and determined the appearance of increased endosomal activity in the thoracic ganglia. These results, while confirming that the LRRK2 loss-of-function in the WD40 domain represents a valid model of PD, reveal that under appropriate concentrations Wse can be usefully employed to counteract some deficits associated with the disease. However, a careful assessment of the risks, likely related to the impaired endosomal activity, is required.
Collapse
Affiliation(s)
| | - Roberto Marotta
- Nanochemistry Department, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Simone Poddighe
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Giuseppe Talani
- Institute of Neuroscience, National Research Council (CNR), Monserrato, Cagliari, Italy
| | - Tiziano Catelani
- Nanochemistry Department, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | | | - Paolo Solla
- Department of Public Health, Clinical and Molecular Medicine, University of Cagliari, Cagliari, Italy
| | - Francesco Marrosu
- Department of Public Health, Clinical and Molecular Medicine, University of Cagliari, Cagliari, Italy
| | - Enrico Sanna
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | | | - Elio Acquas
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Anna Liscia
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
48
|
Ho DH, Kim H, Kim J, Sim H, Ahn H, Kim J, Seo H, Chung KC, Park BJ, Son I, Seol W. Leucine-Rich Repeat Kinase 2 (LRRK2) phosphorylates p53 and induces p21(WAF1/CIP1) expression. Mol Brain 2015; 8:54. [PMID: 26384650 PMCID: PMC4575451 DOI: 10.1186/s13041-015-0145-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/04/2015] [Indexed: 11/25/2022] Open
Abstract
Background Leucine-rich repeat kinase 2 (LRRK2) is a gene in which a mutation causes Parkinson’s disease (PD), and p53 is a prototype tumor suppressor. In addition, activation of p53 in patient with PD has been reported by several studies. Because phosphorylation of p53 is critical for regulating its activity and LRRK2 is a kinase, we tested whether p53 is phosphorylated by LRRK2. Results LRRK2 phosphorylates threonine (Thr) at TXR sites in an in vitro kinase assay, and the T304 and T377 were identified as putative phosphorylated residues. An increase of phospho-Thr in the p53 TXR motif was confirmed in the cells overexpressing G2019S, and human induced pluripotent stem (iPS) cells of a G2019S carrier. Interactions between LRRK2 and p53 were confirmed by co-immunoprecipitation of lysates of differentiated SH-SY5Y cells. LRRK2 mediated p53 phosphorylation translocalizes p53 predominantly to nucleus and increases p21WAF1/CIP1 expression in SH-SY5Y cells based on reverse transcription-polymerase chain reaction and Western blot assay results. The luciferase assay using the p21WAF1/CIP1 promoter-reporter also confirmed that LRRK2 kinase activity increases p21 expression. Exogenous expression of G2019S and the phosphomimetic p53 T304/377D mutants increased expression of p21WAF1/CIP1 and cleaved PARP, and cytotoxicity in the same cells. We also observed increase of p21 expression in rat primary neuron cells after transient expression of p53 T304/377D mutants and the mid-brain lysates of the G2019S transgenic mice. Conclusion p53 is a LRRK2 kinase substrate. Phosphorylation of p53 by LRRK2 induces p21WAF1/CIP1 expression and apoptosis in differentiated SH-SY5Y cells and rat primary neurons. Electronic supplementary material The online version of this article (doi:10.1186/s13041-015-0145-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dong Hwan Ho
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea.,Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, Republic of Korea
| | - Hyejung Kim
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea
| | - Jisun Kim
- Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, Republic of Korea
| | - Hyuna Sim
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea.,Korea University of Science & Technology (UST), 113 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Hyunjun Ahn
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea.,Korea University of Science & Technology (UST), 113 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Janghwan Kim
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea.,Korea University of Science & Technology (UST), 113 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Hyemyung Seo
- Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, Republic of Korea
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seodaemun-gu, Seoul, Republic of Korea
| | - Bum-Joon Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Pusan, Republic of Korea
| | - Ilhong Son
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea. .,Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea.
| | - Wongi Seol
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea.
| |
Collapse
|
49
|
Abstract
Parkinson’s disease is a common, adult-onset neurodegenerative disorder whose pathogenesis is still under intense investigation. Substantial evidence from postmortem human brain tissue, genetic- and toxin-induced animal and cellular models indicates that mitochondrial dysfunction plays a central role in the pathophysiology of the disease. This review discusses our current understanding of Parkinson’s disease–related mitochondrial dysfunction, including bioenergetic defects, mitochondrial DNA alterations, altered mitochondrial dynamics, activation of mitochondrial-dependent programmed cell death, and perturbations in mitochondrial tethering to the endoplasmic reticulum. Whether a primary or secondary event, mitochondrial dysfunction holds promise as a potential therapeutic target to halt the progression of neurodegeneration in Parkinson’s disease.
Collapse
|
50
|
Li JQ, Tan L, Yu JT. The role of the LRRK2 gene in Parkinsonism. Mol Neurodegener 2014; 9:47. [PMID: 25391693 PMCID: PMC4246469 DOI: 10.1186/1750-1326-9-47] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 10/21/2014] [Indexed: 12/13/2022] Open
Abstract
Parkinson’s disease (PD), like many common age-related conditions, has been recognized to have a substantial genetic component. Multiple lines of evidence suggest that Leucine-rich repeat kinase 2 (LRRK2) is a crucial factor to understanding the etiology of PD. LRRK2 is a large, widely expressed, multi-domain and multifunctional protein. LRRK2 mutations are the major cause to inherited and sporadic PD. In this review, we discuss the pathology and clinical features which show diversity and variability of LRRK2-associated PD. In addition, we do a thorough literature review and provide theoretical data for gene counseling. Further, we present the evidence linking LRRK2 to various possible pathogenic mechanism of PD such as α-synuclein, tau, inflammatory response, oxidative stress, mitochondrial dysfunction, synaptic dysfunction as well as autophagy-lysosomal system. Based on the above work, we investigate activities both within GTPase and outside enzymatic regions in order to obtain a potential therapeutic approach to solve the LRRK2 problem.
Collapse
Affiliation(s)
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No, 5 Donghai Middle Road, Qingdao 266071, PR China.
| | | |
Collapse
|