1
|
Rallis E, Grech VS, Lotsaris K, Tertipi N, Sfyri E, Kefala V. Skin and Induced Pluripotent Stem Cells as Biomarkers for Neurodegenerative Diseases. Genes (Basel) 2024; 15:1507. [PMID: 39766775 PMCID: PMC11675334 DOI: 10.3390/genes15121507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
As the global population ages, the rising prevalence of neurodegenerative diseases, characterized by abnormal protein aggregates, presents significant challenges for early diagnosis and disease monitoring. Identifying accessible tissue biomarkers is crucial for advancing our ability to detect and track the progression of these diseases. Among the most promising biomarkers is the skin, which shares a common embryological origin with the brain and central nervous system (CNS). This biological connection positions the skin as a potential reflection of CNS pathology. Over the past decades, gene expression studies have demonstrated that key genes involved in neurodegenerative diseases are also expressed in skin tissues. Genes such as APP, PSEN1, PPA2, PINK1, LRRK2, PLCB4, MAPT, SPAST, and SPG7 are prominent in this regard. Beyond gene expression, proteins related to neurodegenerative diseases-such as α-synuclein, TAU, PARKIN, and prion protein (PrP)-have been isolated from the skin of affected individuals, underscoring the skin's capacity to mirror neural degeneration. This non-invasive window into neurodegenerative processes is further enhanced by advances in stem cell technology, which have allowed for the generation of human-induced pluripotent stem cells (iPSCs) from patient-derived fibroblasts. These iPSCs offer a valuable model for studying disease mechanisms and developing therapeutic approaches. This review conducts a comprehensive analysis of the literature from databases such as PubMed, Google Scholar, and ResearchGate, emphasizing the unique potential of the skin as a non-invasive biomarker for neurodegenerative diseases. It explores how the skin serves as a bridge between gene expression and disease pathology in both the skin and the CNS. By leveraging this biological connection, the skin emerges as a promising model for enhancing our understanding of neurodegenerative disorders and developing innovative strategies for early detection and treatment. However, significant limitations remain, requiring further validation to establish the specificity and sensitivity of these biomarkers.
Collapse
Affiliation(s)
- Efstathios Rallis
- Department of Biomedical Sciences, School of Health and Care Sciences, University of West Attica, GR-12243 Athens, Greece; (E.R.); (N.T.); (E.S.); (V.K.)
| | - Vasiliki-Sofia Grech
- Department of Biomedical Sciences, School of Health and Care Sciences, University of West Attica, GR-12243 Athens, Greece; (E.R.); (N.T.); (E.S.); (V.K.)
| | - Kleomenis Lotsaris
- Psychiatrist in Department of Psychiatry, Athens General Hospital ‘Evaggelismos’, GR-10676 Athens, Greece;
| | - Niki Tertipi
- Department of Biomedical Sciences, School of Health and Care Sciences, University of West Attica, GR-12243 Athens, Greece; (E.R.); (N.T.); (E.S.); (V.K.)
| | - Eleni Sfyri
- Department of Biomedical Sciences, School of Health and Care Sciences, University of West Attica, GR-12243 Athens, Greece; (E.R.); (N.T.); (E.S.); (V.K.)
| | - Vassiliki Kefala
- Department of Biomedical Sciences, School of Health and Care Sciences, University of West Attica, GR-12243 Athens, Greece; (E.R.); (N.T.); (E.S.); (V.K.)
| |
Collapse
|
2
|
Kulkarni PG, Mohire VM, Waghmare PP, Banerjee T. Interplay of mitochondria-associated membrane proteins and autophagy: Implications in neurodegeneration. Mitochondrion 2024; 76:101874. [PMID: 38514017 DOI: 10.1016/j.mito.2024.101874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024]
Abstract
Since the discovery of membrane contact sites between ER and mitochondria called mitochondria-associated membranes (MAMs), several pieces of evidence identified their role in the regulation of different cellular processes such as Ca2+ signalling, mitochondrial transport, and dynamics, ER stress, inflammation, glucose homeostasis, and autophagy. The integrity of these membranes was found to be essential for the maintenance of these cellular functions. Accumulating pieces of evidence suggest that MAMs serve as a platform for autophagosome formation. However, the alteration within MAMs structure is associated with the progression of neurodegenerative diseases. Dysregulated autophagy is a hallmark of neurodegeneration. Here, in this review, we highlight the present knowledge on MAMs, their structural composition, and their roles in different cellular functions. We also discuss the association of MAMs proteins with impaired autophagy and their involvement in the progression of neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Prakash G Kulkarni
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007 India
| | - Vaibhavi M Mohire
- Molecular Neuroscience Research Centre, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y Patil Vidyapeeth, Survey No 87/88, Mumbai Bangalore Express Highway, Tathawade, Pune 411 033 India
| | - Pranjal P Waghmare
- Molecular Neuroscience Research Centre, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y Patil Vidyapeeth, Survey No 87/88, Mumbai Bangalore Express Highway, Tathawade, Pune 411 033 India
| | - Tanushree Banerjee
- Molecular Neuroscience Research Centre, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y Patil Vidyapeeth, Survey No 87/88, Mumbai Bangalore Express Highway, Tathawade, Pune 411 033 India; Infosys Ltd., SEZ unit VI, Plot No. 1, Rajiv Gandhi Infotech Park, Hinjawadi Phase I, Pune, Maharashtra 411057, India.
| |
Collapse
|
3
|
Payne T, Burgess T, Bradley S, Roscoe S, Sassani M, Dunning MJ, Hernandez D, Scholz S, McNeill A, Taylor R, Su L, Wilkinson I, Jenkins T, Mortiboys H, Bandmann O. Multimodal assessment of mitochondrial function in Parkinson's disease. Brain 2024; 147:267-280. [PMID: 38059801 PMCID: PMC10766247 DOI: 10.1093/brain/awad364] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 09/02/2023] [Accepted: 09/27/2023] [Indexed: 12/08/2023] Open
Abstract
The heterogenous aetiology of Parkinson's disease is increasingly recognized; both mitochondrial and lysosomal dysfunction have been implicated. Powerful, clinically applicable tools are required to enable mechanistic stratification for future precision medicine approaches. The aim of this study was to characterize bioenergetic dysfunction in Parkinson's disease by applying a multimodal approach, combining standardized clinical assessment with midbrain and putaminal 31-phosphorus magnetic resonance spectroscopy (31P-MRS) and deep phenotyping of mitochondrial and lysosomal function in peripheral tissue in patients with recent-onset Parkinson's disease and control subjects. Sixty participants (35 patients with Parkinson's disease and 25 healthy controls) underwent 31P-MRS for quantification of energy-rich metabolites [ATP, inorganic phosphate (Pi) and phosphocreatine] in putamen and midbrain. In parallel, skin biopsies were obtained from all research participants to establish fibroblast cell lines for subsequent quantification of total intracellular ATP and mitochondrial membrane potential (MMP) as well as mitochondrial and lysosomal morphology, using high content live cell imaging. Lower MMP correlated with higher intracellular ATP (r = -0.55, P = 0.0016), higher mitochondrial counts (r = -0.72, P < 0.0001) and higher lysosomal counts (r = -0.62, P = 0.0002) in Parkinson's disease patient-derived fibroblasts only, consistent with impaired mitophagy and mitochondrial uncoupling. 31P-MRS-derived posterior putaminal Pi/ATP ratio variance was considerably greater in Parkinson's disease than in healthy controls (F-tests, P = 0.0036). Furthermore, elevated 31P-MRS-derived putaminal, but not midbrain Pi/ATP ratios (indicative of impaired oxidative phosphorylation) correlated with both greater mitochondrial (r = 0.37, P = 0.0319) and lysosomal counts (r = 0.48, P = 0.0044) as well as lower MMP in both short (r = -0.52, P = 0.0016) and long (r = -0.47, P = 0.0052) mitochondria in Parkinson's disease. Higher 31P-MRS midbrain phosphocreatine correlated with greater risk of rapid disease progression (r = 0.47, P = 0.0384). Our data suggest that impaired oxidative phosphorylation in the striatal dopaminergic nerve terminals exceeds mitochondrial dysfunction in the midbrain of patients with early Parkinson's disease. Our data further support the hypothesis of a prominent link between impaired mitophagy and impaired striatal energy homeostasis as a key event in early Parkinson's disease.
Collapse
Affiliation(s)
- Thomas Payne
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Toby Burgess
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Stephen Bradley
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Sarah Roscoe
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Matilde Sassani
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, The University of Birmingham, Birmingham B15 2TT, UK
| | - Mark J Dunning
- The Bioinformatics Core, Sheffield Institute of Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Dena Hernandez
- Molecular Genetics Section, Laboratory of Neurogenetics, NIA, NIH, Bethesda, MD 20814, USA
| | - Sonja Scholz
- Neurodegenerative Diseases Research Unit, Laboratory of Neurogenetics, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20814, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD 21287, USA
| | - Alisdair McNeill
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Rosie Taylor
- Statistical Services Unit, The University of Sheffield, Shefield S3 7RH, UK
| | - Li Su
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
- Department of Psychiatry, University of Cambridge, Cambridge CB2 0SP, UK
| | - Iain Wilkinson
- Academic Unit of Radiology, University of Sheffield, Sheffield S10 2JF, UK
| | - Thomas Jenkins
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
- Department of Neurology, Royal Perth Hospital, Perth WA6000, Australia
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Oliver Bandmann
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield S10 2HQ, UK
| |
Collapse
|
4
|
Cronin SJF, Yu W, Hale A, Licht-Mayer S, Crabtree MJ, Korecka JA, Tretiakov EO, Sealey-Cardona M, Somlyay M, Onji M, An M, Fox JD, Turnes BL, Gomez-Diaz C, da Luz Scheffer D, Cikes D, Nagy V, Weidinger A, Wolf A, Reither H, Chabloz A, Kavirayani A, Rao S, Andrews N, Latremoliere A, Costigan M, Douglas G, Freitas FC, Pifl C, Walz R, Konrat R, Mahad DJ, Koslov AV, Latini A, Isacson O, Harkany T, Hallett PJ, Bagby S, Woolf CJ, Channon KM, Je HS, Penninger JM. Crucial neuroprotective roles of the metabolite BH4 in dopaminergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.08.539795. [PMID: 37214873 PMCID: PMC10197517 DOI: 10.1101/2023.05.08.539795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Dopa-responsive dystonia (DRD) and Parkinson's disease (PD) are movement disorders caused by the dysfunction of nigrostriatal dopaminergic neurons. Identifying druggable pathways and biomarkers for guiding therapies is crucial due to the debilitating nature of these disorders. Recent genetic studies have identified variants of GTP cyclohydrolase-1 (GCH1), the rate-limiting enzyme in tetrahydrobiopterin (BH4) synthesis, as causative for these movement disorders. Here, we show that genetic and pharmacological inhibition of BH4 synthesis in mice and human midbrain-like organoids accurately recapitulates motor, behavioral and biochemical characteristics of these human diseases, with severity of the phenotype correlating with extent of BH4 deficiency. We also show that BH4 deficiency increases sensitivities to several PD-related stressors in mice and PD human cells, resulting in worse behavioral and physiological outcomes. Conversely, genetic and pharmacological augmentation of BH4 protects mice from genetically- and chemically induced PD-related stressors. Importantly, increasing BH4 levels also protects primary cells from PD-affected individuals and human midbrain-like organoids (hMLOs) from these stressors. Mechanistically, BH4 not only serves as an essential cofactor for dopamine synthesis, but also independently regulates tyrosine hydroxylase levels, protects against ferroptosis, scavenges mitochondrial ROS, maintains neuronal excitability and promotes mitochondrial ATP production, thereby enhancing mitochondrial fitness and cellular respiration in multiple preclinical PD animal models, human dopaminergic midbrain-like organoids and primary cells from PD-affected individuals. Our findings pinpoint the BH4 pathway as a key metabolic program at the intersection of multiple protective mechanisms for the health and function of midbrain dopaminergic neurons, identifying it as a potential therapeutic target for PD.
Collapse
Affiliation(s)
- Shane J F Cronin
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Weonjin Yu
- Signature Program in Neuroscience and Behavioural Disorders, Duke-National University of Singapore (NUS) Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Ashley Hale
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Simon Licht-Mayer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Mark J Crabtree
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Joanna A Korecka
- Neurodegeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA
| | - Evgenii O Tretiakov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Marco Sealey-Cardona
- Department of Structural and Computational Biology, Max Perutz Labs, Vienna Biocenter Campus 5, 1030, Vienna, Austria
| | - Mate Somlyay
- Department of Structural and Computational Biology, Max Perutz Labs, Vienna Biocenter Campus 5, 1030, Vienna, Austria
| | - Masahiro Onji
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Meilin An
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Jesse D Fox
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Bruna Lenfers Turnes
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Carlos Gomez-Diaz
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Débora da Luz Scheffer
- LABOX, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC 88037-100, Brazil
| | - Domagoj Cikes
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Vanja Nagy
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD); Department of Neurology, Medical University of Vienna (MUW), 1090 Vienna, Austria
| | - Adelheid Weidinger
- Ludwig Boltzmann Institute for Traumatology. The Research Center in Cooperation with AUVA, Donaueschingen Str. 13, 1200 Vienna, Austria
| | - Alexandra Wolf
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Harald Reither
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Antoine Chabloz
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Anoop Kavirayani
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Shuan Rao
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Nick Andrews
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Alban Latremoliere
- Neurosurgery Department, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Michael Costigan
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Gillian Douglas
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | | | - Christian Pifl
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Roger Walz
- Center for Applied Neurocience, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil; Neurology Division, Internal Medicine Department, University Hospital of UFSC, Florianópolis, Brazil
| | - Robert Konrat
- Department of Structural and Computational Biology, Max Perutz Labs, Vienna Biocenter Campus 5, 1030, Vienna, Austria
| | - Don J Mahad
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Andrey V Koslov
- Ludwig Boltzmann Institute for Traumatology. The Research Center in Cooperation with AUVA, Donaueschingen Str. 13, 1200 Vienna, Austria
| | - Alexandra Latini
- LABOX, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC 88037-100, Brazil
| | - Ole Isacson
- Neurodegeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Neuroscience, Biomedicum 7D, Karolinska Institute, Solna, Sweden
| | - Penelope J Hallett
- Neurodegeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA
| | - Stefan Bagby
- Department of Biology and Biochemistry and the Milner Centre for Evolution, University of Bath, Bath, UK
| | - Clifford J Woolf
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Keith M Channon
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Hyunsoo Shawn Je
- Signature Program in Neuroscience and Behavioural Disorders, Duke-National University of Singapore (NUS) Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| |
Collapse
|
5
|
García-Beltrán O, Urrutia PJ, Núñez MT. On the Chemical and Biological Characteristics of Multifunctional Compounds for the Treatment of Parkinson's Disease. Antioxidants (Basel) 2023; 12:214. [PMID: 36829773 PMCID: PMC9952574 DOI: 10.3390/antiox12020214] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Protein aggregation, mitochondrial dysfunction, iron dyshomeostasis, increased oxidative damage and inflammation are pathognomonic features of Parkinson's disease (PD) and other neurodegenerative disorders characterized by abnormal iron accumulation. Moreover, the existence of positive feed-back loops between these pathological components, which accelerate, and sometimes make irreversible, the neurodegenerative process, is apparent. At present, the available treatments for PD aim to relieve the symptoms, thus improving quality of life, but no treatments to stop the progression of the disease are available. Recently, the use of multifunctional compounds with the capacity to attack several of the key components of neurodegenerative processes has been proposed as a strategy to slow down the progression of neurodegenerative processes. For the treatment of PD specifically, the necessary properties of new-generation drugs should include mitochondrial destination, the center of iron-reactive oxygen species interaction, iron chelation capacity to decrease iron-mediated oxidative damage, the capacity to quench free radicals to decrease the risk of ferroptotic neuronal death, the capacity to disrupt α-synuclein aggregates and the capacity to decrease inflammatory conditions. Desirable additional characteristics are dopaminergic neurons to lessen unwanted secondary effects during long-term treatment, and the inhibition of the MAO-B and COMPT activities to increase intraneuronal dopamine content. On the basis of the published evidence, in this work, we review the molecular basis underlying the pathological events associated with PD and the clinical trials that have used single-target drugs to stop the progress of the disease. We also review the current information on multifunctional compounds that may be used for the treatment of PD and discuss the chemical characteristics that underlie their functionality. As a projection, some of these compounds or modifications could be used to treat diseases that share common pathology features with PD, such as Friedreich's ataxia, Multiple sclerosis, Huntington disease and Alzheimer's disease.
Collapse
Affiliation(s)
- Olimpo García-Beltrán
- Facultad de Ciencias Naturales y Matemáticas, Universidad de Ibagué, Carrera 22 Calle 67, Ibagué 730002, Colombia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, General Gana 1702, Santiago 8370854, Chile
| | - Pamela J. Urrutia
- Faculty of Medicine and Science, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile
| | - Marco T. Núñez
- Faculty of Sciences, Universidad de Chile, Las Palmeras 3425, Santiago 7800024, Chile
| |
Collapse
|
6
|
Ruz C, Alcantud JL, Vives F, Arrebola F, Hardy J, Lewis PA, Manzoni C, Duran R. Seventy-Two-Hour LRRK2 Kinase Activity Inhibition Increases Lysosomal GBA Expression in H4, a Human Neuroglioma Cell Line. Int J Mol Sci 2022; 23:ijms23136935. [PMID: 35805938 PMCID: PMC9266636 DOI: 10.3390/ijms23136935] [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: 05/23/2022] [Revised: 06/19/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
Mutations in LRRK2 and GBA1 are key contributors to genetic risk of developing Parkinson's disease (PD). To investigate how LRRK2 kinase activity interacts with GBA and contributes to lysosomal dysfunctions associated with the pathology of PD. The activity of the lysosomal enzyme β-Glucocerebrosidase (GCase) was assessed in a human neuroglioma cell model treated with two selective inhibitors of LRKK2 kinase activity (LRRK2-in-1 and MLi-2) and a GCase irreversible inhibitor, condutirol-beta-epoxide (CBE), under 24 and 72 h experimental conditions. We observed levels of GCase activity comparable to controls in response to 24 and 72 h treatments with LRRK2-in-1 and MLi-2. However, GBA protein levels increased upon 72 h treatment with LRRK2-in-1. Moreover, LC3-II protein levels were increased after both 24 and 72 h treatments with LRRK2-in-1, suggesting an activation of the autophagic pathway. These results highlight a possible regulation of lysosomal function through the LRRK2 kinase domain and suggest an interplay between LRRK2 kinase activity and GBA. Although further investigations are needed, the enhancement of GCase activity might restore the defective protein metabolism seen in PD.
Collapse
Affiliation(s)
- Clara Ruz
- Department of Physiology, Faculty of Medicine, Universidad de Granada, 18016 Granada, Spain; (C.R.); (F.V.)
- Institute of Neurosciences “Federico Olóriz”, Centro de Investigación Biomédica (CIBM), Universidad de Granada, 18016 Granada, Spain; (J.L.A.); (F.A.)
| | - José Luis Alcantud
- Institute of Neurosciences “Federico Olóriz”, Centro de Investigación Biomédica (CIBM), Universidad de Granada, 18016 Granada, Spain; (J.L.A.); (F.A.)
| | - Francisco Vives
- Department of Physiology, Faculty of Medicine, Universidad de Granada, 18016 Granada, Spain; (C.R.); (F.V.)
- Institute of Neurosciences “Federico Olóriz”, Centro de Investigación Biomédica (CIBM), Universidad de Granada, 18016 Granada, Spain; (J.L.A.); (F.A.)
| | - Francisco Arrebola
- Institute of Neurosciences “Federico Olóriz”, Centro de Investigación Biomédica (CIBM), Universidad de Granada, 18016 Granada, Spain; (J.L.A.); (F.A.)
- Department of Histology, Faculty of Medicine, Universidad de Granada, 18016 Granada, Spain
| | - John Hardy
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK; (J.H.); (P.A.L.)
| | - Patrick A. Lewis
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK; (J.H.); (P.A.L.)
- Department of Comparative Biomedical Science, Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - Claudia Manzoni
- Department of Pharmacology, UCL School of Pharmacy, London WC1N 1AX, UK;
| | - Raquel Duran
- Department of Physiology, Faculty of Medicine, Universidad de Granada, 18016 Granada, Spain; (C.R.); (F.V.)
- Institute of Neurosciences “Federico Olóriz”, Centro de Investigación Biomédica (CIBM), Universidad de Granada, 18016 Granada, Spain; (J.L.A.); (F.A.)
- Correspondence:
| |
Collapse
|
7
|
Trinh J, Schymanski EL, Smajic S, Kasten M, Sammler E, Grünewald A. Molecular mechanisms defining penetrance of LRRK2-associated Parkinson's disease. MED GENET-BERLIN 2022; 34:103-116. [PMID: 38835904 PMCID: PMC11006382 DOI: 10.1515/medgen-2022-2127] [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] [Indexed: 06/06/2024]
Abstract
Mutations in Leucine-rich repeat kinase 2 (LRRK2) are the most frequent cause of dominantly inherited Parkinson's disease (PD). LRRK2 mutations, among which p.G2019S is the most frequent, are inherited with reduced penetrance. Interestingly, the disease risk associated with LRRK2 G2019S can vary dramatically depending on the ethnic background of the carrier. While this would suggest a genetic component in the definition of LRRK2-PD penetrance, only few variants have been shown to modify the age at onset of patients harbouring LRRK2 mutations, and the exact cellular pathways controlling the transition from a healthy to a diseased state currently remain elusive. In light of this knowledge gap, recent studies also explored environmental and lifestyle factors as potential modifiers of LRRK2-PD. In this article, we (i) describe the clinical characteristics of LRRK2 mutation carriers, (ii) review known genes linked to LRRK2-PD onset and (iii) summarize the cellular functions of LRRK2 with particular emphasis on potential penetrance-related molecular mechanisms. This section covers LRRK2's involvement in Rab GTPase and immune signalling as well as in the regulation of mitochondrial homeostasis and dynamics. Additionally, we explored the literature with regard to (iv) lifestyle and (v) environmental factors that may influence the penetrance of LRRK2 mutations, with a view towards further exposomics studies. Finally, based on this comprehensive overview, we propose potential future in vivo, in vitro and in silico studies that could provide a better understanding of the processes triggering PD in individuals with LRRK2 mutations.
Collapse
Affiliation(s)
- Joanne Trinh
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Emma L. Schymanski
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Semra Smajic
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Meike Kasten
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | - Esther Sammler
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Department of Neurology, School of Medicine, Dundee, Ninewells Hospital, Dundee, UK
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
8
|
Kumar S, Behl T, Sehgal A, Chigurupati S, Singh S, Mani V, Aldubayan M, Alhowail A, Kaur S, Bhatia S, Al-Harrasi A, Subramaniyan V, Fuloria S, Fuloria NK, Sekar M, Abdel Daim MM. Exploring the focal role of LRRK2 kinase in Parkinson's disease. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:32368-32382. [PMID: 35147886 DOI: 10.1007/s11356-022-19082-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/02/2022] [Indexed: 06/14/2023]
Abstract
The major breakthroughs in our knowledge of how biology plays a role in Parkinson's disease (PD) have opened up fresh avenues designed to know the pathogenesis of disease and identify possible therapeutic targets. Mitochondrial abnormal functioning is a key cellular feature in the pathogenesis of PD. An enzyme, leucine-rich repeat kinase 2 (LRRK2), involved in both the idiopathic and familial PD risk, is a therapeutic target. LRRK2 has a link to the endolysosomal activity. Enhanced activity of the LRRK2 kinase, endolysosomal abnormalities and aggregation of autophagic vesicles with imperfectly depleted substrates, such as α-synuclein, are all seen in the substantia nigra dopaminergic neurons in PD. Despite the fact that LRRK2 is involved in endolysosomal and autophagic activity, it is undefined if inhibiting LRRK2 kinase activity will prevent endolysosomal dysfunction or minimise the degeneration of dopaminergic neurons. The inhibitor's capability of LRRK2 kinase to inhibit endolysosomal and neuropathological alterations in human PD indicates that LRRK2 inhibitors could have significant therapeutic usefulness in PD. G2019S is perhaps the maximum common mutation in PD subjects. Even though LRRK2's well-defined structure has still not been established, numerous LRRK2 inhibitors have been discovered. This review summarises the role of LRRK2 kinase in Parkinson's disease.
Collapse
Affiliation(s)
- Sachin Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Vasudevan Mani
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Maha Aldubayan
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Ahmed Alhowail
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Satvinder Kaur
- GHG Khalsa College of Pharmacy, Gurusar Sadhar, Ludhiana, Punjab, India
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
- School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | | | - Shivkanya Fuloria
- Faculty of Pharmacy and Centre of Excellence for Biomaterials Engineering, AIMST University, Bedon, Kedah, Malaysia
| | - Neeraj Kumar Fuloria
- Faculty of Pharmacy and Centre of Excellence for Biomaterials Engineering, AIMST University, Bedon, Kedah, Malaysia
| | - Mahendran Sekar
- Department of Pharmaceutical Chemistrty, Faculty of Pharmacy and Health Science, Universiti Kuala Lumpur, Royal College of Medicine Perak, Ipoh, Perak, Malaysia
| | - Mohamed M Abdel Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
9
|
Investigation of USP30 inhibition to enhance Parkin-mediated mitophagy: tools and approaches. Biochem J 2021; 478:4099-4118. [PMID: 34704599 PMCID: PMC8718267 DOI: 10.1042/bcj20210508] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 12/20/2022]
Abstract
Mitochondrial dysfunction is implicated in Parkinson disease (PD). Mutations in Parkin, an E3 ubiquitin ligase, can cause juvenile-onset Parkinsonism, probably through impairment of mitophagy. Inhibition of the de-ubiquitinating enzyme USP30 may counter this effect to enhance mitophagy. Using different tools and cellular approaches, we wanted to independently confirm this claimed role for USP30. Pharmacological characterisation of additional tool compounds that selectively inhibit USP30 are reported. The consequence of USP30 inhibition by these compounds, siRNA knockdown and overexpression of dominant-negative USP30 on the mitophagy pathway in different disease-relevant cellular models was explored. Knockdown and inhibition of USP30 showed increased p-Ser65-ubiquitin levels and mitophagy in neuronal cell models. Furthermore, patient-derived fibroblasts carrying pathogenic mutations in Parkin showed reduced p-Ser65-ubiquitin levels compared with wild-type cells, levels that could be restored using either USP30 inhibitor or dominant-negative USP30 expression. Our data provide additional support for USP30 inhibition as a regulator of the mitophagy pathway.
Collapse
|
10
|
Sunanda T, Ray B, Mahalakshmi AM, Bhat A, Rashan L, Rungratanawanich W, Song BJ, Essa MM, Sakharkar MK, Chidambaram SB. Mitochondria-Endoplasmic Reticulum Crosstalk in Parkinson's Disease: The Role of Brain Renin Angiotensin System Components. Biomolecules 2021; 11:1669. [PMID: 34827667 PMCID: PMC8615717 DOI: 10.3390/biom11111669] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 12/12/2022] Open
Abstract
The past few decades have seen an increased emphasis on the involvement of the mitochondrial-associated membrane (MAM) in various neurodegenerative diseases, particularly in Parkinson's disease (PD) and Alzheimer's disease (AD). In PD, alterations in mitochondria, endoplasmic reticulum (ER), and MAM functions affect the secretion and metabolism of proteins, causing an imbalance in calcium homeostasis and oxidative stress. These changes lead to alterations in the translocation of the MAM components, such as IP3R, VDAC, and MFN1 and 2, and consequently disrupt calcium homeostasis and cause misfolded proteins with impaired autophagy, distorted mitochondrial dynamics, and cell death. Various reports indicate the detrimental involvement of the brain renin-angiotensin system (RAS) in oxidative stress, neuroinflammation, and apoptosis in various neurodegenerative diseases. In this review, we attempted to update the reports (using various search engines, such as PubMed, SCOPUS, Elsevier, and Springer Nature) demonstrating the pathogenic interactions between the various proteins present in mitochondria, ER, and MAM with respect to Parkinson's disease. We also made an attempt to speculate the possible involvement of RAS and its components, i.e., AT1 and AT2 receptors, angiotensinogen, in this crosstalk and PD pathology. The review also collates and provides updated information on the role of MAM in calcium signaling, oxidative stress, neuroinflammation, and apoptosis in PD.
Collapse
Affiliation(s)
- Tuladhar Sunanda
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India; (T.S.); (B.R.); (A.M.M.); (A.B.)
- Centre for Experimental Pharmacology and Toxicology, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Bipul Ray
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India; (T.S.); (B.R.); (A.M.M.); (A.B.)
- Centre for Experimental Pharmacology and Toxicology, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Arehally M. Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India; (T.S.); (B.R.); (A.M.M.); (A.B.)
| | - Abid Bhat
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India; (T.S.); (B.R.); (A.M.M.); (A.B.)
- Centre for Experimental Pharmacology and Toxicology, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Luay Rashan
- Biodiversity Research Centre, Dohfar University, Salalah 2059, Oman;
| | - Wiramon Rungratanawanich
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, 9000 Rockville Pike, Bethesda, MD 20892, USA; (W.R.); (B.-J.S.)
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, 9000 Rockville Pike, Bethesda, MD 20892, USA; (W.R.); (B.-J.S.)
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat 123, Oman;
- Ageing and Dementia Research Group, Sultan Qaboos University, Muscat 123, Oman
| | - Meena Kishore Sakharkar
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India; (T.S.); (B.R.); (A.M.M.); (A.B.)
- Centre for Experimental Pharmacology and Toxicology, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| |
Collapse
|
11
|
Bharat V, Hsieh CH, Wang X. Mitochondrial Defects in Fibroblasts of Pathogenic MAPT Patients. Front Cell Dev Biol 2021; 9:765408. [PMID: 34805172 PMCID: PMC8595217 DOI: 10.3389/fcell.2021.765408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
Mutations in MAPT gene cause multiple neurological disorders, including frontal temporal lobar degeneration and parkinsonism. Increasing evidence indicates impaired mitochondrial homeostasis and mitophagy in patients and disease models of pathogenic MAPT. Here, using MAPT patients' fibroblasts as a model, we report that disease-causing MAPT mutations compromise early events of mitophagy. By employing biochemical and mitochondrial assays we discover that upon mitochondrial depolarization, the recruitment of LRRK2 and Parkin to mitochondria and degradation of the outer mitochondrial membrane protein Miro1 are disrupted. Using high resolution electron microscopy, we reveal that the contact of mitochondrial membranes with ER and cytoskeleton tracks is dissociated following mitochondrial damage. This membrane dissociation is blocked by a pathogenic MAPT mutation. Furthermore, we provide evidence showing that tau protein, which is encoded by MAPT gene, interacts with Miro1 protein, and this interaction is abolished by pathogenic MAPT mutations. Lastly, treating fibroblasts of a MAPT patient with a small molecule promotes Miro1 degradation following depolarization. Altogether, our results show molecular defects in a peripheral tissue of patients and suggest that targeting mitochondrial quality control may have a broad application for future therapeutic intervention.
Collapse
|
12
|
Rosenbusch KE, Oun A, Sanislav O, Lay ST, Keizer-Gunnink I, Annesley SJ, Fisher PR, Dolga AM, Kortholt A. A Conserved Role for LRRK2 and Roco Proteins in the Regulation of Mitochondrial Activity. Front Cell Dev Biol 2021; 9:734554. [PMID: 34568343 PMCID: PMC8455996 DOI: 10.3389/fcell.2021.734554] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/16/2021] [Indexed: 01/02/2023] Open
Abstract
Parkinson's Disease (PD) is the second most common neurodegenerative disease world-wide. Mutations in the multidomain protein Leucine Rich Repeat Kinase 2 (LRRK2) are the most frequent cause of hereditary PD. Furthermore, recent data suggest that independent of mutations, increased kinase activity of LRRK2 plays an essential role in PD pathogenesis. Isolated mitochondria of tissue samples from PD patients carrying LRRK2 mutations display a significant impairment of mitochondrial function. However, due to the complexity of the mitochondrial signaling network, the role of LRRK2 in mitochondrial metabolism is still not well understood. Previously we have shown that D. discoideum Roco4 is a suitable model to study the activation mechanism of LRRK2 in vivo. To get more insight in the LRRK2 pathways regulating mitochondrial activity we used this Roco4 model system in combination with murine RAW macrophages. Here we show that both Dictyostelium roco4 knockout and cells expressing PD-mutants show behavioral and developmental phenotypes that are characteristic for mitochondrial impairment. Mitochondrial activity measured by Seahorse technology revealed that the basal respiration of D. discoideum roco4- cells is significantly increased compared to the WT strain, while the basal and maximal respiration values of cells overexpressing Roco4 are reduced compared to the WT strain. Consistently, LRRK2 KO RAW 264.7 cells exhibit higher maximal mitochondrial respiration activity compared to the LRRK2 parental RAW264.7 cells. Measurement on isolated mitochondria from LRRK2 KO and parental RAW 264.7 cells revealed no difference in activity compared to the parental cells. Furthermore, neither D. discoideum roco4- nor LRRK2 KO RAW 264.7 showed a difference in either the number or the morphology of mitochondria compared to their respective parental strains. This suggests that the observed effects on the mitochondrial respiratory in cells are indirect and that LRRK2/Roco proteins most likely require other cytosolic cofactors to elicit mitochondrial effects.
Collapse
Affiliation(s)
| | - Asmaa Oun
- Department of Cell Biochemistry, University of Groningen, Groningen, Netherlands.,Groningen Research Institute of Pharmacy (GRIP), Molecular Pharmacology XB10, Groningen, Netherlands.,Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Oana Sanislav
- Department of Physiology Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Sui T Lay
- Department of Physiology Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Ineke Keizer-Gunnink
- Department of Physiology Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Sarah J Annesley
- Department of Physiology Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Paul R Fisher
- Department of Physiology Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Amalia M Dolga
- Groningen Research Institute of Pharmacy (GRIP), Molecular Pharmacology XB10, Groningen, Netherlands
| | - Arjan Kortholt
- Department of Cell Biochemistry, University of Groningen, Groningen, Netherlands.,Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
13
|
Paß T, Wiesner RJ, Pla-Martín D. Selective Neuron Vulnerability in Common and Rare Diseases-Mitochondria in the Focus. Front Mol Biosci 2021; 8:676187. [PMID: 34295920 PMCID: PMC8290884 DOI: 10.3389/fmolb.2021.676187] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/08/2021] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial dysfunction is a central feature of neurodegeneration within the central and peripheral nervous system, highlighting a strong dependence on proper mitochondrial function of neurons with especially high energy consumptions. The fitness of mitochondria critically depends on preservation of distinct processes, including the maintenance of their own genome, mitochondrial dynamics, quality control, and Ca2+ handling. These processes appear to be differently affected in common neurodegenerative diseases, such as Alzheimer’s and Parkinson’s disease, as well as in rare neurological disorders, including Huntington’s disease, Amyotrophic Lateral Sclerosis and peripheral neuropathies. Strikingly, particular neuron populations of different morphology and function perish in these diseases, suggesting that cell-type specific factors contribute to the vulnerability to distinct mitochondrial defects. Here we review the disruption of mitochondrial processes in common as well as in rare neurological disorders and its impact on selective neurodegeneration. Understanding discrepancies and commonalities regarding mitochondrial dysfunction as well as individual neuronal demands will help to design new targets and to make use of already established treatments in order to improve treatment of these diseases.
Collapse
Affiliation(s)
- Thomas Paß
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Rudolf J Wiesner
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - David Pla-Martín
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| |
Collapse
|
14
|
Abrahams S, Miller HC, Lombard C, van der Westhuizen FH, Bardien S. Curcumin pre-treatment may protect against mitochondrial damage in LRRK2-mutant Parkinson's disease and healthy control fibroblasts. Biochem Biophys Rep 2021; 27:101035. [PMID: 34189277 PMCID: PMC8219994 DOI: 10.1016/j.bbrep.2021.101035] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 05/19/2021] [Accepted: 05/23/2021] [Indexed: 12/05/2022] Open
Abstract
Mitochondrial dysfunction has been proposed as one of the pathobiological underpinnings in Parkinson's disease. Environmental stressors, such as paraquat, induce mitochondrial dysfunction and promote reactive oxygen species production. Targeting oxidative stress pathways could prevent mitochondrial dysfunction and thereby halt the neurodegeneration in Parkinson's disease. Since curcumin is touted as an antioxidant and neuroprotective agent, the aim of this study was to investigate if curcumin is a suitable therapy to target mitochondrial dysfunction in Parkinson's disease using a paraquat-toxicity induced model in fibroblasts from LRRK2-mutation positive Parkinson's disease individuals and healthy controls. The fibroblasts were exposed to five treatment groups, (i) untreated, (ii) curcumin only, (iii) paraquat only, (iv) pre-curcumin group: with curcumin for 2hr followed by paraquat for 24hr and (v) post-curcumin group: with paraquat for 24hr followed by curcumin for 2hr. Mitochondrial function was determined by measuring three parameters of mitochondrial respiration (maximal respiration, ATP-associated respiration, and spare respiratory capacity) using the Seahorse XFe96 Extracellular Flux Analyzer. As expected, paraquat effectively disrupted mitochondrial function for all parameters. Pre-curcumin treatment improved maximal and ATP-associated respiration whereas, post-curcumin treatment had no effect. These findings indicate that curcumin may be most beneficial as a pre-treatment before toxin exposure, which has implications for its therapeutic use. These promising findings warrant future studies testing different curcumin dosages, exposure times and curcumin formulations in larger sample sizes of Parkinson's disease and control participants.
Paraquat reduced respiration in Parkinson's disease and control fibroblasts. Curcumin, an antioxidant, improved mitochondrial respiration, as a pre-treatment. Post-treatment with curcumin did not improve mitochondrial respiration.
Collapse
Key Words
- ATP, Adenosine Triphosphate
- DMEM, Dulbecco's Modified Eagle Medium
- DMSO, Dimethyl Sulfoxide
- FCCP, Carbonyl cyanide-4-(trifluoromethoxy) phenylhydrazone
- LRRK2, Leucine Rich Repeat Kinase 2
- MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- Mitochondrial function
- OCR, oxygen consumption rate
- Oxidative stress
- PD, Parkinson's disease
- Paraquat
- Turmeric
Collapse
Affiliation(s)
- Shameemah Abrahams
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Hayley C Miller
- Human Metabolomics, Faculty of Natural Sciences, North West University, Potchefstroom, South Africa
| | - Carl Lombard
- Division of Epidemiology and Biostatistics, Department of Global Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.,Biostatistics Unit, South African Medical Research Council, Cape Town, South Africa
| | | | - Soraya Bardien
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
15
|
Calamini B, Geyer N, Huss-Braun N, Bernhardt A, Harsany V, Rival P, Cindhuchao M, Hoffmann D, Gratzer S. Development of a physiologically relevant and easily scalable LUHMES cell-based model of G2019S LRRK2-driven Parkinson's disease. Dis Model Mech 2021; 14:dmm048017. [PMID: 34114604 PMCID: PMC8214734 DOI: 10.1242/dmm.048017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 04/27/2021] [Indexed: 11/20/2022] Open
Abstract
Parkinson's disease (PD) is a fatal neurodegenerative disorder that is primarily caused by the degeneration and loss of dopaminergic neurons of the substantia nigra in the ventral midbrain. Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of late-onset PD identified to date, with G2019S being the most frequent LRRK2 mutation, which is responsible for up to 1-2% of sporadic PD and up to 6% of familial PD cases. As no treatment is available for this devastating disease, developing new therapeutic strategies is of foremost importance. Cellular models are commonly used for testing novel potential neuroprotective compounds. However, current cellular PD models either lack physiological relevance to dopaminergic neurons or are too complex and costly for scaling up the production process and for screening purposes. In order to combine biological relevance and throughput, we have developed a PD model in Lund human mesencephalic (LUHMES) cell-derived dopaminergic neurons by overexpressing wild-type (WT) and G2019S LRRK2 proteins. We show that these cells can differentiate into dopaminergic-like neurons and that expression of mutant LRRK2 causes a range of different phenotypes, including reduced nuclear eccentricity, altered mitochondrial and lysosomal morphologies, and increased dopaminergic cell death. This model could be used to elucidate G2019S LRRK2-mediated dopaminergic neural dysfunction and to identify novel molecular targets for disease intervention. In addition, our model could be applied to high-throughput and phenotypic screenings for the identification of novel PD therapeutics.
Collapse
Affiliation(s)
- Barbara Calamini
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Nathalie Geyer
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Nathalie Huss-Braun
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Annie Bernhardt
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Véronique Harsany
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Pierrick Rival
- BioTherapeutics/e-Biology - Bioinformatics, Sanofi Biologics Research, 13 quai Jules Guesde, 94400 Vitry-sur-Seine, France
| | - May Cindhuchao
- Molecular Screening Technology, Sanofi Biologics Research, 270 Albany Street, Cambridge, MA 02139, USA
| | - Dietmar Hoffmann
- Molecular Screening Technology, Sanofi Biologics Research, 270 Albany Street, Cambridge, MA 02139, USA
| | - Sabine Gratzer
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| |
Collapse
|
16
|
Ramos-Gonzalez P, Mato S, Chara JC, Verkhratsky A, Matute C, Cavaliere F. Astrocytic atrophy as a pathological feature of Parkinson's disease with LRRK2 mutation. NPJ Parkinsons Dis 2021; 7:31. [PMID: 33785762 PMCID: PMC8009947 DOI: 10.1038/s41531-021-00175-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/03/2021] [Indexed: 02/01/2023] Open
Abstract
The principal hallmark of Parkinson's disease (PD) is the selective neurodegeneration of dopaminergic neurones. Mounting evidence suggests that astrocytes may contribute to dopaminergic neurodegeneration through decreased homoeostatic support and deficient neuroprotection. In this study, we generated induced pluripotent stem cells (iPSC)-derived astrocytes from PD patients with LRRK2(G2019S) mutation and healthy donors of the similar age. In cell lines derived from PD patients, astrocytes were characterised by a significant decrease in S100B and GFAP-positive astrocytic profiles associated with marked decrease in astrocyte complexity. In addition, PD-derived astrocytes demonstrated aberrant mitochondrial morphology, decreased mitochondrial activity and ATP production along with an increase of glycolysis and increased production of reactive oxygen species. Taken together, our data indicate that astrocytic asthenia observed in patient-derived cultures with LRRK2(G2019S) mutation may contribute to neuronal death through decreased homoeostatic support, elevated oxidative stress and failed neuroprotection.
Collapse
Affiliation(s)
- Paula Ramos-Gonzalez
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Susana Mato
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Biocruces, Bizkaia, Barakaldo, Spain
| | - Juan Carlos Chara
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Alexei Verkhratsky
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Carlos Matute
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Fabio Cavaliere
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain.
- Achucarro Basque Center for Neuroscience, Leioa, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
17
|
LRRK2 at the Crossroad of Aging and Parkinson's Disease. Genes (Basel) 2021; 12:genes12040505. [PMID: 33805527 PMCID: PMC8066012 DOI: 10.3390/genes12040505] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 01/01/2023] Open
Abstract
Parkinson's disease (PD) is a heterogeneous neurodegenerative disease characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta and the widespread occurrence of proteinaceous inclusions known as Lewy bodies and Lewy neurites. The etiology of PD is still far from clear, but aging has been considered as the highest risk factor influencing the clinical presentations and the progression of PD. Accumulating evidence suggests that aging and PD induce common changes in multiple cellular functions, including redox imbalance, mitochondria dysfunction, and impaired proteostasis. Age-dependent deteriorations in cellular dysfunction may predispose individuals to PD, and cellular damages caused by genetic and/or environmental risk factors of PD may be exaggerated by aging. Mutations in the LRRK2 gene cause late-onset, autosomal dominant PD and comprise the most common genetic causes of both familial and sporadic PD. LRRK2-linked PD patients show clinical and pathological features indistinguishable from idiopathic PD patients. Here, we review cellular dysfunctions shared by aging and PD-associated LRRK2 mutations and discuss how the interplay between the two might play a role in PD pathologies.
Collapse
|
18
|
He X, Yuan W, Liu F, Feng J, Guo Y. Acylated Ghrelin is Protective Against 6-OHDA-induced Neurotoxicity by Regulating Autophagic Flux. Front Pharmacol 2021; 11:586302. [PMID: 33584263 PMCID: PMC7872958 DOI: 10.3389/fphar.2020.586302] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/21/2020] [Indexed: 11/13/2022] Open
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative disorders, and our previous study revealed that autophagic flux dysfunction contributes to the neuron death in 6-OHDA-induced PD models. Acylated ghrelin is a neuropeptide that has a variety of actions in the central nervous system. In the current study, we aimed to investigate whether ghrelin is neuroprotective in 6-OHDA-induced rat model and SH-SY5Y cell model and whether it is related to autophagic flux regulation. We observed that ghrelin could effectively reduce apomorphine-induced contralateral rotation in 6-OHDA-induced PD rats, preserve the expression of tyrosine hydroxylase (TH) and increase the cell viability. It could upregulate the expression of autophagy related proteins like Atg7 and LC3-II and downregulate p62, and downregulate apoptosis related proteins like bax and cleaved caspase 3. SH-SY5Y cells transfected with adenovirus Ad-mCherry-GFP-LC3B further revealed that ghrelin could relieve the autophagic flux dysfunction induced by 6-OHDA. Lysotracker staining showed that ghrelin could reverse the decrease in lysosomes induced by 6-OHDA and immunofluorescence staining revealed a reverse of TFEB level in SH-SY5Y cells. Blocking autophagy activation with 3-methyladenine (3-MA) in rats treated with ghrelin and 6-OHDA showed no notable change in apoptosis-related markers, while blocking autophagosome fusion with lysosomes with chloroquine could notably reverse the downregulation of bax/bcl-2 ratio and cleaved caspase three expression by ghrelin. Additionally, knockdown ATG7, the upstream regulator of autophagy, with siRNA could further decrease the number of apoptotic cells in SH-SY5Y cells exposed to 6-OHDA and treated with ghrelin, while knockdown TFEB, a key transcription factor for lysosome biosynthesis and function, with siRNA could completely abolish the anti-apoptosis effect of ghrelin. These data suggest that ghrelin is neuroprotective in 6-OHDA-induced PD models via improving autophagic flux dysfunction and restoration of TFEB level.
Collapse
Affiliation(s)
- Xin He
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wei Yuan
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| | - Fei Liu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yanxia Guo
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
19
|
Thomas R, Moloney EB, Macbain ZK, Hallett PJ, Isacson O. Fibroblasts from idiopathic Parkinson's disease exhibit deficiency of lysosomal glucocerebrosidase activity associated with reduced levels of the trafficking receptor LIMP2. Mol Brain 2021; 14:16. [PMID: 33468204 PMCID: PMC7816505 DOI: 10.1186/s13041-020-00712-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/07/2020] [Indexed: 12/25/2022] Open
Abstract
Lysosomal dysfunction is a central pathway associated with Parkinson's disease (PD) pathogenesis. Haploinsufficiency of the lysosomal hydrolase GBA (encoding glucocerebrosidase (GCase)) is one of the largest genetic risk factors for developing PD. Deficiencies in the activity of the GCase enzyme have been observed in human tissues from both genetic (harboring mutations in the GBA gene) and idiopathic forms of the disease. To understand the mechanisms behind the deficits of lysosomal GCase enzyme activity in idiopathic PD, this study utilized a large cohort of fibroblast cells from control subjects and PD patients with and without mutations in the GBA gene (N370S mutation) (control, n = 15; idiopathic PD, n = 31; PD with GBA N370S mutation, n = 6). The current data demonstrates that idiopathic PD fibroblasts devoid of any mutations in the GBA gene also exhibit reduction in lysosomal GCase activity, similar to those with the GBA N370S mutation. This reduced GCase enzyme activity in idiopathic PD cells was accompanied by decreased expression of the GBA trafficking receptor, LIMP2, and increased ER retention of the GBA protein in these cells. Importantly, in idiopathic PD fibroblasts LIMP2 protein levels correlated significantly with GCase activity, which was not the case in control subjects or in genetic PD GBA N370S cells. In conclusion, idiopathic PD fibroblasts have decreased GCase activity primarily driven by altered LIMP2-mediated transport of GBA to lysosome and the reduced GCase activity exhibited by the genetic GBA N370S derived PD fibroblasts occurs through a different mechanism.
Collapse
Affiliation(s)
- Ria Thomas
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA
| | - Elizabeth B Moloney
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA
| | - Zachary K Macbain
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA
| | - Penelope J Hallett
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA.
| | - Ole Isacson
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA.
| |
Collapse
|
20
|
Liu H, Ho PWL, Leung CT, Pang SYY, Chang EES, Choi ZYK, Kung MHW, Ramsden DB, Ho SL. Aberrant mitochondrial morphology and function associated with impaired mitophagy and DNM1L-MAPK/ERK signaling are found in aged mutant Parkinsonian LRRK2 R1441G mice. Autophagy 2020; 17:3196-3220. [PMID: 33300446 PMCID: PMC8526027 DOI: 10.1080/15548627.2020.1850008] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mitochondrial dysfunction causes energy deficiency and nigrostriatal neurodegeneration which is integral to the pathogenesis of Parkinson disease (PD). Clearance of defective mitochondria involves fission and ubiquitin-dependent degradation via mitophagy to maintain energy homeostasis. We hypothesize that LRRK2 (leucine-rich repeat kinase 2) mutation disrupts mitochondrial turnover causing accumulation of defective mitochondria in aging brain. We found more ubiquitinated mitochondria with aberrant morphology associated with impaired function in aged (but not young) LRRK2R1441G knockin mutant mouse striatum compared to wild-type (WT) controls. LRRK2R1441G mutant mouse embryonic fibroblasts (MEFs) exhibited reduced MAP1LC3/LC3 activation indicating impaired macroautophagy/autophagy. Mutant MEFs under FCCP-induced (mitochondrial uncoupler) stress showed increased LC3-aggregates demonstrating impaired mitophagy. Using a novel flow cytometry assay to quantify mitophagic rates in MEFs expressing photoactivatable mito-PAmCherry, we found significantly slower mitochondria clearance in mutant cells. Specific LRRK2 kinase inhibition using GNE-7915 did not alleviate impaired mitochondrial clearance suggesting a lack of direct relationship to increased kinase activity alone. DNM1L/Drp1 knockdown in MEFs slowed mitochondrial clearance indicating that DNM1L is a prerequisite for mitophagy. DNM1L knockdown in slowing mitochondrial clearance was less pronounced in mutant MEFs, indicating preexisting impaired DNM1L activation. DNM1L knockdown disrupted mitochondrial network which was more evident in mutant MEFs. DNM1L-Ser616 and MAPK/ERK phosphorylation which mediate mitochondrial fission and downstream mitophagic processes was apparent in WT using FCCP-induced stress but not mutant MEFs, despite similar total MAPK/ERK and DNM1L levels. In conclusion, aberrant mitochondria morphology and dysfunction associated with impaired mitophagy and DNM1L-MAPK/ERK signaling are found in mutant LRRK2 MEFs and mouse brain. Abbreviations: ATP: adenosine triphosphate; BAX: BCL2-associated X protein; CDK1: cyclin-dependent kinase 1; CDK5: cyclin-dependent kinase 5; CQ: chloroquine; CSF: cerebrospinal fluid; DNM1L/DRP1: dynamin 1-like; ELISA: enzyme-linked immunosorbent assay; FACS: fluorescence-activated cell sorting; FCCP: carbonyl cyanide-4-(trifluoromethoxy)phenylhydrazone; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; LAMP2A: lysosomal-associated membrane protein 2A; LRRK2: leucine-rich repeat kinase 2; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MAPK1/ERK2: mitogen-activated protein kinase 1; MEF: mouse embryonic fibroblast; MFN1: mitofusin 1; MMP: mitochondrial membrane potential; PAmCherry: photoactivatable-mCherry; PD: Parkinson disease; PINK1: PTEN induced putative kinase 1; PRKN/PARKIN: parkin RBR E3 ubiquitin protein ligase; RAB10: RAB10, member RAS oncogene family; RAF: v-raf-leukemia oncogene; SNCA: synuclein, alpha; TEM: transmission electron microscopy; VDAC: voltage-dependent anion channel; WT: wild type; SQSTM1/p62: sequestosome 1.
Collapse
Affiliation(s)
- Huifang Liu
- Division of Neurology, Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Philip Wing-Lok Ho
- Division of Neurology, Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Chi-Ting Leung
- Division of Neurology, Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Shirley Yin-Yu Pang
- Division of Neurology, Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Eunice Eun Seo Chang
- Division of Neurology, Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Zoe Yuen-Kiu Choi
- Division of Neurology, Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Michelle Hiu-Wai Kung
- Division of Neurology, Department of Medicine, University of Hong Kong, Hong Kong, China
| | - David Boyer Ramsden
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Shu-Leong Ho
- Division of Neurology, Department of Medicine, University of Hong Kong, Hong Kong, China
| |
Collapse
|
21
|
Disrupted Mitochondrial and Metabolic Plasticity Underlie Comorbidity between Age-Related and Degenerative Disorders as Parkinson Disease and Type 2 Diabetes Mellitus. Antioxidants (Basel) 2020; 9:antiox9111063. [PMID: 33143119 PMCID: PMC7693963 DOI: 10.3390/antiox9111063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/17/2022] Open
Abstract
Idiopathic Parkinson’s disease (iPD) and type 2 diabetes mellitus (T2DM) are chronic, multisystemic, and degenerative diseases associated with aging, with eventual epidemiological co-morbidity and overlap in molecular basis. This study aims to explore if metabolic and mitochondrial alterations underlie the previously reported epidemiologic and clinical co-morbidity from a molecular level. To evaluate the adaptation of iPD to a simulated pre-diabetogenic state, we exposed primary cultured fibroblasts from iPD patients and controls to standard (5 mM) and high (25 mM) glucose concentrations to further characterize metabolic and mitochondrial resilience. iPD fibroblasts showed increased organic and amino acid levels related to mitochondrial metabolism with respect to controls, and these differences were enhanced in high glucose conditions (citric, suberic, and sebacic acids levels increased, as well as alanine, glutamate, aspartate, arginine, and ornithine amino acids; p-values between 0.001 and 0.05). The accumulation of metabolites in iPD fibroblasts was associated with (and probably due to) the concomitant mitochondrial dysfunction observed at enzymatic, oxidative, respiratory, and morphologic level. Metabolic and mitochondrial plasticity of controls was not observed in iPD fibroblasts, which were unable to adapt to different glucose conditions. Impaired metabolism and mitochondrial activity in iPD may limit energy supply for cell survival. Moreover, reduced capacity to adapt to disrupted glucose balance characteristic of T2DM may underlay the co-morbidity between both diseases. Conclusions: Fibroblasts from iPD patients showed mitochondrial impairment, resulting in the accumulation of organic and amino acids related to mitochondrial metabolism, especially when exposed to high glucose. Mitochondrial and metabolic defects down warding cell plasticity to adapt to changing glucose bioavailability may explain the comorbidity between iPD and T2DM.
Collapse
|
22
|
Palese F, Pontis S, Realini N, Piomelli D. NAPE-specific phospholipase D regulates LRRK2 association with neuronal membranes. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 90:217-238. [PMID: 33706934 DOI: 10.1016/bs.apha.2020.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
N-acylphosphatidylethanolamines (NAPEs) are glycerophospholipid precursors for bioactive lipid amides and potential regulators of membrane function. They are hydrolyzed by NAPE-specific phospholipase D (NAPE-PLD) and have been implicated in neurodegenerative disorders such as Parkinson's disease. Here, we used siRNA-mediated silencing of NAPE-PLD in human SH-SY5Y cells and NAPE-PLD-/- mice to determine whether NAPEs influence the membrane association of LRRK2, a multifunctional protein kinase that is frequently mutated in persons with sporadic Parkinson's disease. NAPE-PLD deletion caused a significant accumulation of non-metabolized NAPEs, which was accompanied by a shift of LRRK2 from membrane to cytosol and a reduction in total LRRK2 content. Conversely, exposure of intact SH-SY5Y cells to bacterial PLD lowered NAPE levels and enhanced LRRK2 association with membranes. The results suggest that NAPE-PLD activity may contribute to the control of LRRK2 localization by regulating membrane NAPE levels.
Collapse
Affiliation(s)
- Francesca Palese
- Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy; Departments of Anatomy and Neurobiology, Pharmacology and Biological Chemistry, University of California, Irvine, CA, United States
| | - Silvia Pontis
- Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Natalia Realini
- Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Daniele Piomelli
- Departments of Anatomy and Neurobiology, Pharmacology and Biological Chemistry, University of California, Irvine, CA, United States.
| |
Collapse
|
23
|
Thomas R, Hallett PJ, Isacson O. Experimental studies of mitochondrial and lysosomal function in in vitro and in vivo models relevant to Parkinson's disease genetic risk. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:279-302. [PMID: 32739007 DOI: 10.1016/bs.irn.2020.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Several studies have identified the involvement of mitochondrial and lysosomal dysfunction in Parkinson's disease (PD) pathology. In this review we discuss recent work that has identified deficits in mitophagy, mitochondrial network formation, increased sensitivity to mitochondrial stressors and alterations in proteins regulating mitochondrial fission and fusion associated with patient-derived fibroblasts harboring mutations in LRRK2 gene and from sporadic PD patient cells. We further focus on alterations of lysosomal enzymes, in particular glucocerebrosidase activity, and resultant lipid dyshomeostasis in PD and aging, in human tissue and in vivo rodent models. Future studies aimed at understanding the convergence of mitochondrial and lysosomal pathways will be of essence for the identification of unique cellular defects in PD and for the development of new treatments.
Collapse
Affiliation(s)
- Ria Thomas
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, United States.
| | - Penelope J Hallett
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, United States.
| | - Ole Isacson
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, United States.
| |
Collapse
|
24
|
Korecka JA, Thomas R, Hinrich AJ, Moskites AM, Macbain ZK, Hallett PJ, Isacson O, Hastings ML. Splice-Switching Antisense Oligonucleotides Reduce LRRK2 Kinase Activity in Human LRRK2 Transgenic Mice. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 21:623-635. [PMID: 32736291 PMCID: PMC7393423 DOI: 10.1016/j.omtn.2020.06.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 04/15/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022]
Abstract
Parkinson’s disease (PD) is a progressive neurological disorder estimated to affect 7–10 million people worldwide. There is no treatment available that cures or slows the progression of PD. Elevated leucine-rich repeat kinase 2 (LRRK2) activity has been associated with genetic and sporadic forms of PD and, thus, reducing LRRK2 function is a promising therapeutic strategy. We have previously reported that an antisense oligonucleotide (ASO) that blocks splicing of LRRK2 exon 41, which encodes part of the kinase domain, reverses aberrant endoplasmic reticulum (ER) calcium levels and mitophagy defects in PD patient-derived cell lines harboring the LRRK2 G2019S mutation. In this study, we show that treating transgenic mice expressing human wild-type or G2019S LRRK2 with a single intracerebroventricular injection of ASO induces exon 41 skipping and results in a decrease in phosphorylation of the LRRK2 kinase substrate RAB10. Exon 41 skipping also reverses LRRK2 kinase-dependent changes in LC3B II/I ratios, a marker for the autophagic process. These results demonstrate the potential of LRRK2 exon 41 skipping as a possible therapeutic strategy to modulate pathogenic LRRK2 kinase activity associated with PD development.
Collapse
Affiliation(s)
- Joanna A Korecka
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA.
| | - Ria Thomas
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Anthony J Hinrich
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Alyssa M Moskites
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Zach K Macbain
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Penelope J Hallett
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Ole Isacson
- Neuroregeneration Research Institute, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Michelle L Hastings
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.
| |
Collapse
|
25
|
Toffoli M, Vieira SRL, Schapira AHV. Genetic causes of PD: A pathway to disease modification. Neuropharmacology 2020; 170:108022. [PMID: 32119885 DOI: 10.1016/j.neuropharm.2020.108022] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/17/2020] [Accepted: 02/26/2020] [Indexed: 01/08/2023]
Abstract
The underline neuropathology of Parkinson disease is pleiomorphic and its genetic background diverse. Possibly because of this heterogeneity, no effective disease modifying therapy is available. In this paper we give an overview of the genetics of Parkinson disease and explain how this is relevant for the development of new therapies. This article is part of the special issue entitled 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- M Toffoli
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, United Kingdom
| | - S R L Vieira
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, United Kingdom
| | - A H V Schapira
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, United Kingdom.
| |
Collapse
|
26
|
Martín-Jiménez R, Lurette O, Hebert-Chatelain E. Damage in Mitochondrial DNA Associated with Parkinson's Disease. DNA Cell Biol 2020; 39:1421-1430. [PMID: 32397749 DOI: 10.1089/dna.2020.5398] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mitochondria are the only organelles that contain their own genetic material (mtDNA). Mitochondria are involved in several key physiological functions, including ATP production, Ca2+ homeostasis, and metabolism of neurotransmitters. Since these organelles perform crucial processes to maintain neuronal homeostasis, mitochondrial dysfunctions can lead to various neurodegenerative diseases. Several mitochondrial proteins involved in ATP production are encoded by mtDNA. Thus, any mtDNA alteration can ultimately lead to mitochondrial dysfunction and cell death. Accumulation of mutations, deletions, and rearrangements in mtDNA has been observed in animal models and patients suffering from Parkinson's disease (PD). Also, specific inherited variations associated with mtDNA genetic groups (known as mtDNA haplogroups) are associated with lower or higher risk of developing PD. Consequently, mtDNA alterations should now be considered important hallmarks of this neurodegenerative disease. This review provides an update about the role of mtDNA alterations in the physiopathology of PD.
Collapse
Affiliation(s)
- Rebeca Martín-Jiménez
- Department of Biology and Université de Moncton, Moncton, Canada
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Université de Moncton, Moncton, Canada
| | - Olivier Lurette
- Department of Biology and Université de Moncton, Moncton, Canada
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Université de Moncton, Moncton, Canada
| | - Etienne Hebert-Chatelain
- Department of Biology and Université de Moncton, Moncton, Canada
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Université de Moncton, Moncton, Canada
| |
Collapse
|
27
|
Korecka JA, Thomas R, Christensen DP, Hinrich AJ, Ferrari EJ, Levy SA, Hastings ML, Hallett PJ, Isacson O. Mitochondrial clearance and maturation of autophagosomes are compromised in LRRK2 G2019S familial Parkinson's disease patient fibroblasts. Hum Mol Genet 2020; 28:3232-3243. [PMID: 31261377 DOI: 10.1093/hmg/ddz126] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 04/16/2019] [Accepted: 06/07/2019] [Indexed: 12/13/2022] Open
Abstract
This study utilized human fibroblasts as a preclinical discovery and diagnostic platform for identification of cell biological signatures specific for the LRRK2 G2019S mutation producing Parkinson's disease (PD). Using live cell imaging with a pH-sensitive Rosella biosensor probe reflecting lysosomal breakdown of mitochondria, mitophagy rates were found to be decreased in fibroblasts carrying the LRRK2 G2019S mutation compared to cells isolated from healthy subject (HS) controls. The mutant LRRK2 increased kinase activity was reduced by pharmacological inhibition and targeted antisense oligonucleotide treatment, which normalized mitophagy rates in the G2019S cells and also increased mitophagy levels in HS cells. Detailed mechanistic analysis showed a reduction of mature autophagosomes in LRRK2 G2019S fibroblasts, which was rescued by LRRK2 specific kinase inhibition. These findings demonstrate an important role for LRRK2 protein in regulation of mitochondrial clearance by the lysosomes, which is hampered in PD with the G2019S mutation. The current results are relevant for cell phenotypic diagnostic approaches and potentially for stratification of PD patients for targeted therapy.
Collapse
Affiliation(s)
- Joanna A Korecka
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Ria Thomas
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Dan P Christensen
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Anthony J Hinrich
- Center for Genetic Diseases, Department of Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Eliza J Ferrari
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Simon A Levy
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Michelle L Hastings
- Center for Genetic Diseases, Department of Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Penelope J Hallett
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Ole Isacson
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| |
Collapse
|
28
|
Weindel CG, Bell SL, Vail KJ, West KO, Patrick KL, Watson RO. LRRK2 maintains mitochondrial homeostasis and regulates innate immune responses to Mycobacterium tuberculosis. eLife 2020; 9:51071. [PMID: 32057291 PMCID: PMC7159881 DOI: 10.7554/elife.51071] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 02/14/2020] [Indexed: 12/11/2022] Open
Abstract
The Parkinson’s disease (PD)-associated gene leucine-rich repeat kinase 2 (LRRK2) has been studied extensively in the brain. However, several studies have established that mutations in LRRK2 confer susceptibility to mycobacterial infection, suggesting LRRK2 also controls immunity. We demonstrate that loss of LRRK2 in macrophages induces elevated basal levels of type I interferon (IFN) and interferon stimulated genes (ISGs) and causes blunted interferon responses to mycobacterial pathogens and cytosolic nucleic acid agonists. Altered innate immune gene expression in Lrrk2 knockout (KO) macrophages is driven by a combination of mitochondrial stresses, including oxidative stress from low levels of purine metabolites and DRP1-dependent mitochondrial fragmentation. Together, these defects promote mtDNA leakage into the cytosol and chronic cGAS engagement. While Lrrk2 KO mice can control Mycobacterium tuberculosis (Mtb) replication, they have exacerbated inflammation and lower ISG expression in the lungs. These results demonstrate previously unappreciated consequences of LRRK2-dependent mitochondrial defects in controlling innate immune outcomes. Parkinson’s disease is a progressive nervous system disorder that causes tremors, slow movements, and stiff and inflexible muscles. The symptoms are caused by the loss of cells known as neurons in a specific part of the brain that helps to regulate how the body moves. Researchers have identified mutations in several genes that are associated with an increased risk of developing Parkinson’s. The most common of these mutations occur in a gene called LRRK2. This gene produces a protein that has been shown to be important for maintaining cellular compartments known as mitochondria, which play a crucial role in generating energy. It remains unclear how these mutations lead to the death of neurons. Mutations in LRRK2 have also been shown to make individuals more susceptible to bacterial infections, suggesting that the protein that LRRK2 codes for may help our immune system. Weindel, Bell et al. set out to understand how this protein works in immune cells called macrophages, which ‘eat’ invading bacteria and produce type I interferons, molecules that promote immune responses. Mouse cells were used to measure the ability of normal macrophages and macrophages that lack the mouse equivalent to LRRK2 (referred to as Lrrk2 knockout macrophages) to make type I interferons. The experiments showed that the Lrrk2 knockout macrophages made type I interferons even when they were not infected with bacteria, suggesting they are subject to stress that triggers immune responses. It was possible to correct the behavior of the Lrrk2 knockout macrophages by repairing their mitochondria. When mice missing the gene equivalent to LRRK2 were infected with the bacterium that causes tuberculosis, they experienced more severe disease. The protein encoded by the LRRK2 gene is considered a potential target for therapies to treat Parkinson’s disease, and several drugs that inhibit this protein are being tested in clinical trials. The findings of Weindel, Bell et al. suggest that these drugs may have unintended negative effects on a patient’s ability to fight infection. This work also indicates that LRRK2 mutations may disrupt immune responses in the brain, where macrophage-like cells called microglia play a crucial role in maintaining healthy neurons. Future studies that examine how mutations in LRRK2 affect microglia may help us understand how Parkinson’s disease develops.
Collapse
Affiliation(s)
- Chi G Weindel
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, United States
| | - Samantha L Bell
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, United States
| | - Krystal J Vail
- Department of Veterinary Pathobiology, Texas A&M University College of Veterinary Medicine and Biomedical Sciences, College Station, United States
| | - Kelsi O West
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, United States
| | - Kristin L Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, United States
| | - Robert O Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, United States
| |
Collapse
|
29
|
Bonello F, Hassoun SM, Mouton-Liger F, Shin YS, Muscat A, Tesson C, Lesage S, Beart PM, Brice A, Krupp J, Corvol JC, Corti O. LRRK2 impairs PINK1/Parkin-dependent mitophagy via its kinase activity: pathologic insights into Parkinson's disease. Hum Mol Genet 2020; 28:1645-1660. [PMID: 30629163 DOI: 10.1093/hmg/ddz004] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/05/2018] [Accepted: 01/01/2019] [Indexed: 11/12/2022] Open
Abstract
Mutations of LRRK2, encoding leucine-rich repeat kinase 2 (LRRK2), are the leading cause of autosomal dominant Parkinson's disease (PD). The most frequent of these mutations, G2019S substitution, increases kinase activity, but it remains unclear how it causes PD. Recent studies suggest that LRRK2 modulates mitochondrial homeostasis. Mitochondrial dysfunction plays a key role in the pathogenesis of autosomal recessive PD forms linked to PARK2 and PINK1, encoding the cytosolic E3 ubiquitin-protein ligase Parkin and the mitochondrial kinase PINK1, which jointly regulate mitophagy. We explored the role of LRRK2 and its kinase activity in PINK1/Parkin-dependent mitophagy. LRRK2 increased mitochondrial aggregation and attenuated mitochondrial clearance in cells coexpressing Parkin and exposed to the protonophore carbonylcyanide m-chlorophenylhydrazone. Förster resonance energy transfer imaging microscopy showed that LRRK2 impaired the interactions between Parkin and Drp1 and their mitochondrial targets early in mitophagy. The inhibition of LRRK2 kinase activity by a 'kinase-dead' LRRK2 mutation or with a pharmacological inhibitor (LRRK2-IN-1) restored these interactions. The monitoring of mitophagy in human primary fibroblasts with the novel dual-fluorescence mtRosella reporter and a new hypothermic shock paradigm revealed similar defects in PD patients with the G2019S LRRK2 substitution or PARK2 mutations relative to healthy subjects. This defect was restored by LRRK2-IN-1 treatment in LRRK2 patients only. Our results suggest that PD forms due to LRRK2 and PARK2 mutations involve pathogenic mechanisms converging on PINK1/Parkin-dependent mitophagy.
Collapse
Affiliation(s)
- Fiona Bonello
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France
| | - Sidi-Mohamed Hassoun
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France
| | - François Mouton-Liger
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France
| | - Yea Seul Shin
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Adeline Muscat
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France
| | - Christelle Tesson
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France
| | - Suzanne Lesage
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France
| | - Philip M Beart
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Alexis Brice
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France.,AP-HP, Hôpital de la Pitié-Salpêtrière, Department of Genetics, Paris, France
| | | | - Jean-Christophe Corvol
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France.,AP-HP, Hôpital de la Pitié-Salpêtrière, Clinical Investigation Center for Neurology (CIC), Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Olga Corti
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Inserm, Paris, France.,CNRS, UMR, Paris, France.,Sorbonne Universités, Paris, France
| |
Collapse
|
30
|
Antony PMA, Kondratyeva O, Mommaerts K, Ostaszewski M, Sokolowska K, Baumuratov AS, Longhino L, Poulain JF, Grossmann D, Balling R, Krüger R, Diederich NJ. Fibroblast mitochondria in idiopathic Parkinson's disease display morphological changes and enhanced resistance to depolarization. Sci Rep 2020; 10:1569. [PMID: 32005875 PMCID: PMC6994699 DOI: 10.1038/s41598-020-58505-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/10/2020] [Indexed: 12/18/2022] Open
Abstract
Mitochondrial dysfunction is a hallmark in idiopathic Parkinson's disease (IPD). Here, we established screenable phenotypes of mitochondrial morphology and function in primary fibroblasts derived from patients with IPD. Upper arm punch skin biopsy was performed in 41 patients with mid-stage IPD and 21 age-matched healthy controls. At the single-cell level, the basal mitochondrial membrane potential (Ψm) was higher in patients with IPD than in controls. Similarly, under carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP) stress, the remaining Ψm was increased in patients with IPD. Analysis of mitochondrial morphometric parameters revealed significantly decreased mitochondrial connectivity in patients with IPD, with 9 of 14 morphometric mitochondrial parameters differing from those in controls. Significant morphometric mitochondrial changes included the node degree, mean volume, skeleton size, perimeter, form factor, node count, erosion body count, endpoints, and mitochondria count (all P-values < 0.05). These functional data reveal that resistance to depolarization was increased by treatment with the protonophore FCCP in patients with IPD, whereas morphometric data revealed decreased mitochondrial connectivity and increased mitochondrial fragmentation.
Collapse
Affiliation(s)
- P M A Antony
- LCSB, University of Luxembourg, Belvaux, Luxembourg.
| | | | | | | | | | | | | | | | - D Grossmann
- LCSB, University of Luxembourg, Belvaux, Luxembourg
| | - R Balling
- LCSB, University of Luxembourg, Belvaux, Luxembourg
| | - R Krüger
- LCSB, University of Luxembourg, Belvaux, Luxembourg
- CHL, Luxembourg, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | | |
Collapse
|
31
|
Rani L, Mondal AC. Emerging concepts of mitochondrial dysfunction in Parkinson’s disease progression: Pathogenic and therapeutic implications. Mitochondrion 2020; 50:25-34. [DOI: 10.1016/j.mito.2019.09.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/13/2019] [Accepted: 09/18/2019] [Indexed: 01/22/2023]
|
32
|
Berwick DC, Heaton GR, Azeggagh S, Harvey K. LRRK2 Biology from structure to dysfunction: research progresses, but the themes remain the same. Mol Neurodegener 2019; 14:49. [PMID: 31864390 PMCID: PMC6925518 DOI: 10.1186/s13024-019-0344-2] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
Since the discovery of leucine-rich repeat kinase 2 (LRRK2) as a protein that is likely central to the aetiology of Parkinson’s disease, a considerable amount of work has gone into uncovering its basic cellular function. This effort has led to the implication of LRRK2 in a bewildering range of cell biological processes and pathways, and probable roles in a number of seemingly unrelated medical conditions. In this review we summarise current knowledge of the basic biochemistry and cellular function of LRRK2. Topics covered include the identification of phosphorylation substrates of LRRK2 kinase activity, in particular Rab proteins, and advances in understanding the activation of LRRK2 kinase activity via dimerisation and association with membranes, especially via interaction with Rab29. We also discuss biochemical studies that shed light on the complex LRRK2 GTPase activity, evidence of roles for LRRK2 in a range of cell signalling pathways that are likely cell type specific, and studies linking LRRK2 to the cell biology of organelles. The latter includes the involvement of LRRK2 in autophagy, endocytosis, and processes at the trans-Golgi network, the endoplasmic reticulum and also key microtubule-based cellular structures. We further propose a mechanism linking LRRK2 dimerisation, GTPase function and membrane recruitment with LRRK2 kinase activation by Rab29. Together these data paint a picture of a research field that in many ways is moving forward with great momentum, but in other ways has not changed fundamentally. Many key advances have been made, but very often they seem to lead back to the same places.
Collapse
Affiliation(s)
- Daniel C Berwick
- School of Health, Life and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK.
| | - George R Heaton
- Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Sonia Azeggagh
- School of Health, Life and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Kirsten Harvey
- Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK.
| |
Collapse
|
33
|
Malik BR, Maddison DC, Smith GA, Peters OM. Autophagic and endo-lysosomal dysfunction in neurodegenerative disease. Mol Brain 2019; 12:100. [PMID: 31783880 PMCID: PMC6884906 DOI: 10.1186/s13041-019-0504-x] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022] Open
Abstract
Due to their post-mitotic state, metabolic demands and often large polarised morphology, the function and survival of neurons is dependent on an efficient cellular waste clearance system both for generation of materials for metabolic processes and removal of toxic components. It is not surprising therefore that deficits in protein clearance can tip the balance between neuronal health and death. Here we discuss how autophagy and lysosome-mediated degradation pathways are disrupted in several neurological disorders. Both genetic and cell biological evidence show the diversity and complexity of vesicular clearance dysregulation in cells, and together may ultimately suggest a unified mechanism for neuronal demise in degenerative conditions. Causative and risk-associated mutations in Alzheimer's disease, Frontotemporal Dementia, Amyotrophic Lateral Sclerosis, Parkinson's disease, Huntington's disease and others have given the field a unique mechanistic insight into protein clearance processes in neurons. Through their broad implication in neurodegenerative diseases, molecules involved in these genetic pathways, in particular those involved in autophagy, are emerging as appealing therapeutic targets for intervention in neurodegeneration.
Collapse
Affiliation(s)
- Bilal R Malik
- UK Dementia Research Institute at Cardiff University, Cardiff, Wales, UK
- School of Biosciences, Cardiff University, Cardiff, Wales, UK
| | - Daniel C Maddison
- UK Dementia Research Institute at Cardiff University, Cardiff, Wales, UK
- School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Gaynor A Smith
- UK Dementia Research Institute at Cardiff University, Cardiff, Wales, UK.
- School of Medicine, Cardiff University, Cardiff, Wales, UK.
| | - Owen M Peters
- UK Dementia Research Institute at Cardiff University, Cardiff, Wales, UK.
- School of Biosciences, Cardiff University, Cardiff, Wales, UK.
| |
Collapse
|
34
|
Deus CM, Pereira SP, Cunha-Oliveira T, Pereira FB, Raimundo N, Oliveira PJ. Mitochondrial remodeling in human skin fibroblasts from sporadic male Parkinson's disease patients uncovers metabolic and mitochondrial bioenergetic defects. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165615. [PMID: 31759069 DOI: 10.1016/j.bbadis.2019.165615] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/24/2019] [Accepted: 11/15/2019] [Indexed: 12/18/2022]
Abstract
Parkinson's Disease (PD) is characterized by dopaminergic neurodegeneration in the substantia nigra. The exact mechanism by which dopaminergic neurodegeneration occurs is still unknown; however, mitochondrial dysfunction has long been implicated in PD pathogenesis. To investigate the sub-cellular events that lead to disease progression and to develop personalized interventions, non-neuronal cells which are collected in a minimally invasive manner can be key to test interventions aimed at improving mitochondrial function. We used human skin fibroblasts from sporadic PD (sPD) patients as a cell proxy to detect metabolic and mitochondrial alterations which would also exist in a non-neuronal cell type. In this model, we used a glucose-free/galactose- glutamine- and pyruvate-containing cell culture medium, which forces cells to be more dependent on oxidative phosphorylation (OXPHOS) for energy production, in order to reveal hidden metabolic and mitochondrial alterations present in fibroblasts from sPD patients. We demonstrated that fibroblasts from sPD patients show hyperpolarized and elongated mitochondrial networks and higher mitochondrial ROS concentration, as well as decreased ATP levels and glycolysis-related ECAR. Our results also showed that abnormalities of fibroblasts from sPD patients became more evident when stimulating OXPHOS. Under these culture conditions, fibroblasts from sPD cells presented decreased basal respiration, ATP-linked OCR and maximal respiration, and increased mitochondria-targeting phosphorylation of DRP1 when compared to control cells. Our work validates the relevance of using fibroblasts from sPD patients to study cellular and molecular changes that are characteristic of dopaminergic neurodegeneration of PD, and shows that forcing mitochondrial OXPHOS uncovers metabolic defects that were otherwise hidden.
Collapse
Affiliation(s)
- Cláudia M Deus
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech, Biocant Park, 3060-197 Cantanhede, Portugal; IIIUC - Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Susana P Pereira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech, Biocant Park, 3060-197 Cantanhede, Portugal; IIIUC - Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal; LaMetEx - Laboratory of Metabolism and Exercise, Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
| | - Teresa Cunha-Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech, Biocant Park, 3060-197 Cantanhede, Portugal
| | - Francisco B Pereira
- Center for Informatics and Systems, University of Coimbra, Polo II, Pinhal de Marrocos, 3030-290 Coimbra, Portugal; Coimbra Polytechnic - ISEC, 3030-193 Coimbra, Portugal.
| | - Nuno Raimundo
- Institute of Cellular Biochemistry, University Medical Center Goettingen, Humboldtallee 23, D-37073 Goettingen, Germany.
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech, Biocant Park, 3060-197 Cantanhede, Portugal; IIIUC - Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal.
| |
Collapse
|
35
|
Autophagic- and Lysosomal-Related Biomarkers for Parkinson's Disease: Lights and Shadows. Cells 2019; 8:cells8111317. [PMID: 31731485 PMCID: PMC6912814 DOI: 10.3390/cells8111317] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder that currently affects 1% of the population over the age of 60 years, for which no disease-modifying treatments exist. This lack of effective treatments is related to the advanced stage of neurodegeneration existing at the time of diagnosis. Thus, the identification of early stage biomarkers is crucial. Biomarker discovery is often guided by the underlying molecular mechanisms leading to the pathology. One of the central pathways deregulated during PD, supported both by genetic and functional studies, is the autophagy-lysosomal pathway. Hence, this review presents different studies on the expression and activity of autophagic and lysosomal proteins, and their functional consequences, performed in peripheral human biospecimens. Although most biomarkers are inconsistent between studies, some of them, namely HSC70 levels in sporadic PD patients, and cathepsin D levels and glucocerebrosidase activity in PD patients carrying GBA mutations, seem to be consistent. Hence, evidence exists that the impairment of the autophagy-lysosomal pathway underlying PD pathophysiology can be detected in peripheral biosamples and further tested as potential biomarkers. However, longitudinal, stratified, and standardized analyses are needed to confirm their clinical validity and utility.
Collapse
|
36
|
Abstract
Kinase activating missense mutations in leucine-rich repeat kinase 2 (LRRK2) are pathogenically linked to neurodegenerative Parkinson's disease (PD). Over the past decade, substantial effort has been devoted to the development of potent and selective small molecule inhibitors of LRRK2, as well as their preclinical testing across different Parkinson's disease models. This review outlines the genetic and biochemical evidence that pathogenic missense mutations increase LRRK2 kinase activity, which in turn provides the rationale for the development of small molecule inhibitors as potential PD therapeutics. An overview of progress in the development of LRRK2 inhibitors is provided, which in particular indicates that highly selective and potent compounds capable of clinical utility have been developed. We outline evidence from rodent- and human-induced pluripotent stem cell models that support a pathogenic role for LRRK2 kinase activity, and review the substantial experiments aimed at evaluating the safety of LRRK2 inhibitors. We address challenges still to overcome in the translational therapeutic pipeline, including biomarker development and clinical trial strategies, and finally outline the potential utility of LRRK2 inhibitors for other genetic forms of PD and ultimately sporadic PD. Collective evidence supports the ongoing clinical translation of LRRK2 inhibitors as a therapeutic intervention for PD is greatly needed.
Collapse
|
37
|
González-Casacuberta I, Juárez-Flores DL, Morén C, Garrabou G. Bioenergetics and Autophagic Imbalance in Patients-Derived Cell Models of Parkinson Disease Supports Systemic Dysfunction in Neurodegeneration. Front Neurosci 2019; 13:894. [PMID: 31551675 PMCID: PMC6748355 DOI: 10.3389/fnins.2019.00894] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder worldwide affecting 2-3% of the population over 65 years. This prevalence is expected to rise as life expectancy increases and diagnostic and therapeutic protocols improve. PD encompasses a multitude of clinical, genetic, and molecular forms of the disease. Even though the mechanistic of the events leading to neurodegeneration remain largely unknown, some molecular hallmarks have been repeatedly reported in most patients and models of the disease. Neuroinflammation, protein misfolding, disrupted endoplasmic reticulum-mitochondria crosstalk, mitochondrial dysfunction and consequent bioenergetic failure, oxidative stress and autophagy deregulation, are amongst the most commonly described. Supporting these findings, numerous familial forms of PD are caused by mutations in genes that are crucial for mitochondrial and autophagy proper functioning. For instance, late and early onset PD associated to mutations in Leucine-rich repeat kinase 2 (LRRK2) and Parkin (PRKN) genes, responsible for the most frequent dominant and recessive inherited forms of PD, respectively, have emerged as promising examples of disease due to their established role in commanding bioenergetic and autophagic balance. Concomitantly, the development of animal and cell models to investigate the etiology of the disease, potential biomarkers and therapeutic approaches are being explored. One of the emerging approaches in this context is the use of patient's derived cells models, such as skin-derived fibroblasts that preserve the genetic background and some environmental cues of the patients. An increasing number of reports in these PD cell models postulate that deficient mitochondrial function and impaired autophagic flux may be determinant in PD accelerated nigral cell death in terms of limitation of cell energy supply and accumulation of obsolete and/or unfolded proteins or dysfunctional organelles. The reliance of neurons on mitochondrial oxidative metabolism and their post-mitotic nature, may explain their increased vulnerability to undergo degeneration upon mitochondrial challenges or autophagic insults. In this scenario, proper mitochondrial function and turnover through mitophagy, are gaining in strength as protective targets to prevent neurodegeneration, together with the use of patient-derived fibroblasts to further explore these events. These findings point out the presence of molecular damage beyond the central nervous system (CNS) and proffer patient-derived cell platforms to the clinical and scientific community, which enable the study of disease etiopathogenesis and therapeutic approaches focused on modifying the natural history of PD through, among others, the enhancement of mitochondrial function and autophagy.
Collapse
Affiliation(s)
- Ingrid González-Casacuberta
- Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona, Barcelona, Spain.,CIBERER-U722, Madrid, Spain
| | - Diana Luz Juárez-Flores
- Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona, Barcelona, Spain.,CIBERER-U722, Madrid, Spain
| | - Constanza Morén
- Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona, Barcelona, Spain.,CIBERER-U722, Madrid, Spain
| | - Gloria Garrabou
- Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona, Barcelona, Spain.,CIBERER-U722, Madrid, Spain
| |
Collapse
|
38
|
Hallett PJ, Engelender S, Isacson O. Lipid and immune abnormalities causing age-dependent neurodegeneration and Parkinson's disease. J Neuroinflammation 2019; 16:153. [PMID: 31331333 PMCID: PMC6647317 DOI: 10.1186/s12974-019-1532-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 06/25/2019] [Indexed: 12/31/2022] Open
Abstract
This article describes pathogenic concepts and factors, in particular glycolipid abnormalities, that create cell dysfunction and synaptic loss in neurodegenerative diseases. By phenocopying lysosomal storage disorders, such as Gaucher disease and related disorders, age- and dose-dependent changes in glycolipid cell metabolism can lead to Parkinson's disease and related dementias. Recent results show that perturbation of sphingolipid metabolism can precede or is a part of abnormal protein handling in both genetic and idiopathic Parkinson's disease and Lewy body dementia. In aging and genetic predisposition with lipid disturbance, α-synuclein's normal vesicular and synaptic role may be detrimentally shifted toward accommodating and binding such lipids. Specific neuronal glycolipid, protein, and vesicular interactions create potential pathophysiology that is amplified by astroglial and microglial immune mechanisms resulting in neurodegeneration. This perspective provides a new logic for therapeutic interventions that do not focus on protein aggregation, but rather provides a guide to the complex biology and the common sequence of events that lead to age-dependent neurodegenerative disorders.
Collapse
Affiliation(s)
- Penelope J Hallett
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, Boston, USA
| | - Simone Engelender
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, Boston, USA.,Present Address: Department of Biochemistry, Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, 31096, Haifa, Israel
| | - Ole Isacson
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, Boston, USA.
| |
Collapse
|
39
|
Smith GA, Lin TH, Sheehan AE, Van der Goes van Naters W, Neukomm LJ, Graves HK, Bis-Brewer DM, Züchner S, Freeman MR. Glutathione S-Transferase Regulates Mitochondrial Populations in Axons through Increased Glutathione Oxidation. Neuron 2019; 103:52-65.e6. [PMID: 31101394 PMCID: PMC6616599 DOI: 10.1016/j.neuron.2019.04.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 12/13/2018] [Accepted: 04/10/2019] [Indexed: 01/14/2023]
Abstract
Mitochondria are essential in long axons to provide metabolic support and sustain neuron integrity. A healthy mitochondrial pool is maintained by biogenesis, transport, mitophagy, fission, and fusion, but how these events are regulated in axons is not well defined. Here, we show that the Drosophila glutathione S-transferase (GST) Gfzf prevents mitochondrial hyperfusion in axons. Gfzf loss altered redox balance between glutathione (GSH) and oxidized glutathione (GSSG) and initiated mitochondrial fusion through the coordinated action of Mfn and Opa1. Gfzf functioned epistatically with the thioredoxin peroxidase Jafrac1 and the thioredoxin reductase 1 TrxR-1 to regulate mitochondrial dynamics. Altering GSH:GSSG ratios in mouse primary neurons in vitro also induced hyperfusion. Mitochondrial changes caused deficits in trafficking, the metabolome, and neuronal physiology. Changes in GSH and oxidative state are associated with neurodegenerative diseases like Alzheimer's. Our demonstration that GSTs are key in vivo regulators of axonal mitochondrial length and number provides a potential mechanistic link.
Collapse
Affiliation(s)
- Gaynor A Smith
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA; UK Dementia Research Institute, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK.
| | - Tzu-Huai Lin
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Amy E Sheehan
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | | | - Lukas J Neukomm
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne VD, Switzerland
| | - Hillary K Graves
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dana M Bis-Brewer
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA; Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
| | - Stephan Züchner
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA; Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
| | - Marc R Freeman
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
40
|
Grünewald A, Kumar KR, Sue CM. New insights into the complex role of mitochondria in Parkinson’s disease. Prog Neurobiol 2019; 177:73-93. [DOI: 10.1016/j.pneurobio.2018.09.003] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 07/09/2018] [Accepted: 09/10/2018] [Indexed: 02/07/2023]
|
41
|
Lucero M, Suarez AE, Chambers JW. Phosphoregulation on mitochondria: Integration of cell and organelle responses. CNS Neurosci Ther 2019; 25:837-858. [PMID: 31025544 PMCID: PMC6566066 DOI: 10.1111/cns.13141] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/29/2019] [Accepted: 04/04/2019] [Indexed: 12/20/2022] Open
Abstract
Mitochondria are highly integrated organelles that are crucial to cell adaptation and mitigating adverse physiology. Recent studies demonstrate that fundamental signal transduction pathways incorporate mitochondrial substrates into their biological programs. Reversible phosphorylation is emerging as a useful mechanism to modulate mitochondrial function in accordance with cellular changes. Critical serine/threonine protein kinases, such as the c-Jun N-terminal kinase (JNK), protein kinase A (PKA), PTEN-induced kinase-1 (PINK1), and AMP-dependent protein kinase (AMPK), readily translocate to the outer mitochondrial membrane (OMM), the interface of mitochondria-cell communication. OMM protein kinases phosphorylate diverse mitochondrial substrates that have discrete effects on organelle dynamics, protein import, respiratory complex activity, antioxidant capacity, and apoptosis. OMM phosphorylation events can be tempered through the actions of local protein phosphatases, such as mitogen-activated protein kinase phosphatase-1 (MKP-1) and protein phosphatase 2A (PP2A), to regulate the extent and duration of signaling. The central mediators of OMM signal transduction are the scaffold proteins because the relative abundance of these accessory proteins determines the magnitude and duration of a signaling event on the mitochondrial surface, which dictates the biological outcome of a local signal transduction pathway. The concentrations of scaffold proteins, such as A-kinase anchoring proteins (AKAPs) and Sab (or SH3 binding protein 5-SH3BP5), have been shown to influence neuronal survival and vulnerability, respectively, in models of Parkinson's disease (PD), highlighting the importance of OMM signaling to health and disease. Despite recent progress, much remains to be discovered concerning the mechanisms of OMM signaling. Nonetheless, enhancing beneficial OMM signaling events and inhibiting detrimental protein-protein interactions on the mitochondrial surface may represent highly selective approaches to restore mitochondrial health and homeostasis and mitigate organelle dysfunction in conditions such as PD.
Collapse
Affiliation(s)
- Maribel Lucero
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, the Biomolecular Sciences Institute, Florida International University, Miami, Florida
| | - Ana E Suarez
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, the Biomolecular Sciences Institute, Florida International University, Miami, Florida
| | - Jeremy W Chambers
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, the Biomolecular Sciences Institute, Florida International University, Miami, Florida
| |
Collapse
|
42
|
Walter J, Bolognin S, Antony PMA, Nickels SL, Poovathingal SK, Salamanca L, Magni S, Perfeito R, Hoel F, Qing X, Jarazo J, Arias-Fuenzalida J, Ignac T, Monzel AS, Gonzalez-Cano L, Pereira de Almeida L, Skupin A, Tronstad KJ, Schwamborn JC. Neural Stem Cells of Parkinson's Disease Patients Exhibit Aberrant Mitochondrial Morphology and Functionality. Stem Cell Reports 2019; 12:878-889. [PMID: 30982740 PMCID: PMC6522948 DOI: 10.1016/j.stemcr.2019.03.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 03/14/2019] [Accepted: 03/14/2019] [Indexed: 12/22/2022] Open
Abstract
Emerging evidence suggests that Parkinson's disease (PD), besides being an age-associated disorder, might also have a neurodevelopment component. Disruption of mitochondrial homeostasis has been highlighted as a crucial cofactor in its etiology. Here, we show that PD patient-specific human neuroepithelial stem cells (NESCs), carrying the LRRK2-G2019S mutation, recapitulate key mitochondrial defects previously described only in differentiated dopaminergic neurons. By combining high-content imaging approaches, 3D image analysis, and functional mitochondrial readouts we show that LRRK2-G2019S mutation causes aberrations in mitochondrial morphology and functionality compared with isogenic controls. LRRK2-G2019S NESCs display an increased number of mitochondria compared with isogenic control lines. However, these mitochondria are more fragmented and exhibit decreased membrane potential. Functional alterations in LRRK2-G2019S cultures are also accompanied by a reduced mitophagic clearance via lysosomes. These findings support the hypothesis that preceding mitochondrial developmental defects contribute to the manifestation of the PD pathology later in life.
Mitochondrial gene expression is altered in NESCs carrying the LRRK2-G2019 mutation LRRK2-G2019S mutation induces alterations in mitochondrial morphology in NESCs Mitophagy is affected in PD-specific NESCs carrying the LRRK2-G2019S mutation Mitochondrial phenotypes in NESC are rescued by genetic correction of LRRK2-G2019S
Collapse
Affiliation(s)
- Jonas Walter
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Silvia Bolognin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Paul M A Antony
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Sarah L Nickels
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg; Life Science Research Unit (LSRU), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Suresh K Poovathingal
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Luis Salamanca
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Stefano Magni
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Rita Perfeito
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal
| | - Fredrik Hoel
- Department of Biomedicine, University of Bergen, 5020 Bergen, Norway
| | - Xiaobing Qing
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Javier Jarazo
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Jonathan Arias-Fuenzalida
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Tomasz Ignac
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Anna S Monzel
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Laura Gonzalez-Cano
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Luis Pereira de Almeida
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg; Center for Research of Biological Systems, University of California San Diego, La Jolla, CA 92093, USA
| | - Karl J Tronstad
- Department of Biomedicine, University of Bergen, 5020 Bergen, Norway
| | - Jens C Schwamborn
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg.
| |
Collapse
|
43
|
Singh A, Zhi L, Zhang H. LRRK2 and mitochondria: Recent advances and current views. Brain Res 2019; 1702:96-104. [PMID: 29894679 PMCID: PMC6281802 DOI: 10.1016/j.brainres.2018.06.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [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
|
44
|
Korecka JA, Talbot S, Osborn TM, de Leeuw SM, Levy SA, Ferrari EJ, Moskites A, Atkinson E, Jodelka FM, Hinrich AJ, Hastings ML, Woolf CJ, Hallett PJ, Isacson O. Neurite Collapse and Altered ER Ca 2+ Control in Human Parkinson Disease Patient iPSC-Derived Neurons with LRRK2 G2019S Mutation. Stem Cell Reports 2018; 12:29-41. [PMID: 30595548 PMCID: PMC6335600 DOI: 10.1016/j.stemcr.2018.11.021] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 12/26/2022] Open
Abstract
The Parkinson disease (PD) genetic LRRK2 gain-of-function mutations may relate to the ER pathological changes seen in PD patients at postmortem. Human induced pluripotent stem cell (iPSC)-derived neurons with the PD pathogenic LRRK2 G2019S mutation exhibited neurite collapse when challenged with the ER Ca2+ influx sarco/ER Ca2+-ATPase inhibitor thapsigargin (THP). Baseline ER Ca2+ levels measured with the ER Ca2+ indicator CEPIA-ER were lower in LRRK2 G2019S human neurons, including in differentiated midbrain dopamine neurons in vitro. After THP challenge, PD patient-derived neurons displayed increased Ca2+ influx and decreased intracellular Ca2+ buffering upon membrane depolarization. These effects were reversed following LRRK2 mutation correction by antisense oligonucleotides and gene editing. Gene expression analysis in LRRK2 G2019S neurons identified modified levels of key store-operated Ca2+ entry regulators, with no alterations in ER Ca2+ efflux. These results demonstrate PD gene mutation LRRK2 G2019S ER calcium-dependent pathogenic effects in human neurons.
Parkinson-linked LRRK2 G2019S induces neurite collapse upon ER Ca2+ influx block LRRK2 G2019S mutation alters Ca2+ uptake and buffering upon ER Ca2+ influx block The LRRK2 G2019S mutation decreases basal ER Ca2+ levels in human iPSC neurons The LRRK2 G2019S mutation modifies gene expression of key SOCE regulators
Collapse
Affiliation(s)
- Joanna A Korecka
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA.
| | - Sebastien Talbot
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Teresia M Osborn
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Sherida M de Leeuw
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Simon A Levy
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Eliza J Ferrari
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Alyssa Moskites
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Elise Atkinson
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Francine M Jodelka
- Center for Genetics Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Anthony J Hinrich
- Center for Genetics Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Michelle L Hastings
- Center for Genetics Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Penelope J Hallett
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA
| | - Ole Isacson
- Neuroregeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA 02478, USA.
| |
Collapse
|
45
|
Wu S, Lei L, Song Y, Liu M, Lu S, Lou D, Shi Y, Wang Z, He D. Mutation of hop-1 and pink-1 attenuates vulnerability of neurotoxicity in C. elegans: the role of mitochondria-associated membrane proteins in Parkinsonism. Exp Neurol 2018; 309:67-78. [PMID: 30076829 DOI: 10.1016/j.expneurol.2018.07.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 07/04/2018] [Accepted: 07/31/2018] [Indexed: 12/22/2022]
Abstract
Mitochondrial dysfunction is considered as a critical mechanism in the pathogenesis of Parkinson's disease (PD). Increasing evidence supports the notion of mitochondria-associated membranes (MAMs) in mitochondrial dysfunction; yet little is known about the role of MAMs-related proteins in the pathogenesis of PD. Herein we exposed the nematode Caenorhabditis elegans to 0.5-10.0 μM rotenone (RO) or 0.2-1.6 mM paraquat (PQ) for 3 days. Our results showed that both RO and PQ induced similar Parkinsonism including motor deficits and dopaminergic degeneration. RO/PQ caused mitochondrial damages characterized by the increase of vacuole areas and autophagy vesicles, but the decrease of mitochondrial cristae. RO/PQ-impacted mitochondrial function was also demonstrated by the decrease of ATP level and mitochondrial membrane potential. Additionally, the attachment or surrounding of endoplasmic reticulum to the damaged mitochondria indicates ultrastructural alterations in MAMs. Using fluorescently labeled transgenic nematodes, we further found that the expression of tomm-7 and genes of Complex I, II and III was reduced, whereas the expression of pink-1 was increased in the exposed animals. To determine MAMs in toxicity toward PD, we investigated the mutants of hop-1 and pink-1, encoding presenilin and PTEN-induced putative kinase 1 (PINK1) in mitochondria-associated membranes, respectively. Results demonstrated that the mutation of both hop-1 and pink-1 reduced the vulnerability of lethal, behavioral, and mitochondrial toxicity induced by RO/PQ. These findings suggest that presenilin and PINK1 play important roles in the RO/PQ-induced neurotoxicity through the mechanisms involved in mitochondria-associated membranes.
Collapse
Affiliation(s)
- Siyu Wu
- Laboratory of Toxicology, School of Ecological and Environmental Sciences, East China Normal University, 500# DongChuan RD, Shanghai 200241, China
| | - Lili Lei
- Laboratory of Toxicology, School of Ecological and Environmental Sciences, East China Normal University, 500# DongChuan RD, Shanghai 200241, China
| | - Yang Song
- Laboratory of Toxicology, School of Ecological and Environmental Sciences, East China Normal University, 500# DongChuan RD, Shanghai 200241, China
| | - Mengting Liu
- Laboratory of Toxicology, School of Ecological and Environmental Sciences, East China Normal University, 500# DongChuan RD, Shanghai 200241, China
| | - Shibo Lu
- Laboratory of Toxicology, School of Ecological and Environmental Sciences, East China Normal University, 500# DongChuan RD, Shanghai 200241, China
| | - Dan Lou
- Department of Environmental Health & Engineering, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St., Baltimore 21205, USA
| | - Yonghong Shi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518# Ziyue RD, Shanghai 200241, China
| | - Zhibin Wang
- Department of Environmental Health & Engineering, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St., Baltimore 21205, USA.
| | - Defu He
- Laboratory of Toxicology, School of Ecological and Environmental Sciences, East China Normal University, 500# DongChuan RD, Shanghai 200241, China; Shanghai Key Laboratory for Urban Ecological Processes and Eco-Restoration, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
46
|
Mitochondrial abnormalities in Parkinson's disease and Alzheimer's disease: can mitochondria be targeted therapeutically? Biochem Soc Trans 2018; 46:891-909. [PMID: 30026371 DOI: 10.1042/bst20170501] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 06/19/2018] [Accepted: 06/25/2018] [Indexed: 12/13/2022]
Abstract
Mitochondrial abnormalities have been identified as a central mechanism in multiple neurodegenerative diseases and, therefore, the mitochondria have been explored as a therapeutic target. This review will focus on the evidence for mitochondrial abnormalities in the two most common neurodegenerative diseases, Parkinson's disease and Alzheimer's disease. In addition, we discuss the main strategies which have been explored in these diseases to target the mitochondria for therapeutic purposes, focusing on mitochondrially targeted antioxidants, peptides, modulators of mitochondrial dynamics and phenotypic screening outcomes.
Collapse
|
47
|
Schulze M, Sommer A, Plötz S, Farrell M, Winner B, Grosch J, Winkler J, Riemenschneider MJ. Sporadic Parkinson's disease derived neuronal cells show disease-specific mRNA and small RNA signatures with abundant deregulation of piRNAs. Acta Neuropathol Commun 2018; 6:58. [PMID: 29986767 PMCID: PMC6038190 DOI: 10.1186/s40478-018-0561-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 06/27/2018] [Indexed: 01/04/2023] Open
Abstract
Differentiated neurons established via iPSCs from patients that suffer from familial Parkinson's disease (PD) have allowed insights into the mechanisms of neurodegeneration. In the larger cohort of patients with sporadic PD, iPSC based information on disease specific cellular phenotypes is rare. We asked whether differences may be present on genomic and epigenomic levels and performed a comprehensive transcriptomic and epigenomic analysis of fibroblasts, iPSCs and differentiated neuronal cells of sporadic PD-patients and controls. We found that on mRNA level, although fibroblasts and iPSCs are largely indistinguishable, differentiated neuronal cells of sporadic PD patients show significant alterations enriched in pathways known to be involved in disease aetiology, like the CREB-pathway and the pathway regulating PGC1α. Moreover, miRNAs and piRNAs/piRNA-like molecules are largely differentially regulated in cells and post-mortem tissue samples between control- and PD-patients. The most striking differences can be found in piRNAs/piRNA-like molecules, with SINE- and LINE-derived piRNAs highly downregulated in a disease specific manner. We conclude that neuronal cells derived from sporadic PD-patients help to elucidate novel disease mechanisms and provide relevant insight into the epigenetic landscape of sporadic Parkinson's disease as particularly regulated by small RNAs.
Collapse
Affiliation(s)
- Markus Schulze
- Department of Neuropathology, Regensburg University Hospital, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
- Present address: Division of Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Annika Sommer
- Department of Stem Cell Biology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Sonja Plötz
- Department of Stem Cell Biology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Michaela Farrell
- Department of Stem Cell Biology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Janina Grosch
- Department of Molecular Neurology, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Markus J Riemenschneider
- Department of Neuropathology, Regensburg University Hospital, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
48
|
Juárez-Flores DL, González-Casacuberta I, Ezquerra M, Bañó M, Carmona-Pontaque F, Catalán-García M, Guitart-Mampel M, Rivero JJ, Tobias E, Milisenda JC, Tolosa E, Marti MJ, Fernández-Santiago R, Cardellach F, Morén C, Garrabou G. Exhaustion of mitochondrial and autophagic reserve may contribute to the development of LRRK2 G2019S -Parkinson's disease. J Transl Med 2018; 16:160. [PMID: 29884186 PMCID: PMC5994110 DOI: 10.1186/s12967-018-1526-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/26/2018] [Indexed: 12/13/2022] Open
Abstract
Background Mutations in leucine rich repeat kinase 2 (LRRK2) are the most common cause of familial Parkinson’s disease (PD). Mitochondrial and autophagic dysfunction has been described as etiologic factors in different experimental models of PD. We aimed to study the role of mitochondria and autophagy in LRRK2G2019S-mutation, and its relationship with the presence of PD-symptoms. Methods Fibroblasts from six non-manifesting LRRK2G2019S-carriers (NM-LRRK2G2019S) and seven patients with LRRK2G2019S-associated PD (PD-LRRK2G2019S) were compared to eight healthy controls (C). An exhaustive assessment of mitochondrial performance and autophagy was performed after 24-h exposure to standard (glucose) or mitochondrial-challenging environment (galactose), where mitochondrial and autophagy impairment may be heightened. Results A similar mitochondrial phenotype of NM-LRRK2G2019S and controls, except for an early mitochondrial depolarization (54.14% increased, p = 0.04), was shown in glucose. In response to galactose, mitochondrial dynamics of NM-LRRK2G2019S improved (− 17.54% circularity, p = 0.002 and + 42.53% form factor, p = 0.051), probably to maintain ATP levels over controls. A compromised bioenergetic function was suggested in PD-LRRK2G2019S when compared to controls in glucose media. An inefficient response to galactose and worsened mitochondrial dynamics (− 37.7% mitochondrial elongation, p = 0.053) was shown, leading to increased oxidative stress. Autophagy initiation (SQTSM/P62) was upregulated in NM-LRRK2G2019S when compared to controls (glucose + 118.4%, p = 0.014; galactose + 114.44%, p = 0.009,) and autophagosome formation increased in glucose media. Despite of elevated SQSTM1/P62 levels of PD-NMG2019S when compared to controls (glucose + 226.14%, p = 0.04; galactose + 78.5%, p = 0.02), autophagosome formation was deficient in PD-LRRK2G2019S when compared to NM-LRRK2G2019S (− 71.26%, p = 0.022). Conclusions Enhanced mitochondrial performance of NM-LRRK2G2019S in mitochondrial-challenging conditions and upregulation of autophagy suggests that an exhaustion of mitochondrial bioenergetic and autophagic reserve, may contribute to the development of PD in LRRK2G2019S mutation carriers. Electronic supplementary material The online version of this article (10.1186/s12967-018-1526-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Diana Luz Juárez-Flores
- Laboratory of Muscle Research and Mitochondrial Function-CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Department of Internal Medicine-Hospital Clínic of Barcelona, Faculty of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Ingrid González-Casacuberta
- Laboratory of Muscle Research and Mitochondrial Function-CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Department of Internal Medicine-Hospital Clínic of Barcelona, Faculty of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Mario Ezquerra
- Laboratory of Parkinson disease and other Neurodegenerative Movement Disorders: Clinical and Experimental Research, Department of Neurology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Barcelona, Spain.,CIBER de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - María Bañó
- Laboratory of Muscle Research and Mitochondrial Function-CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Department of Internal Medicine-Hospital Clínic of Barcelona, Faculty of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | | | - Marc Catalán-García
- Laboratory of Muscle Research and Mitochondrial Function-CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Department of Internal Medicine-Hospital Clínic of Barcelona, Faculty of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Mariona Guitart-Mampel
- Laboratory of Muscle Research and Mitochondrial Function-CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Department of Internal Medicine-Hospital Clínic of Barcelona, Faculty of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Juan José Rivero
- Laboratory of Muscle Research and Mitochondrial Function-CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Department of Internal Medicine-Hospital Clínic of Barcelona, Faculty of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Ester Tobias
- Laboratory of Muscle Research and Mitochondrial Function-CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Department of Internal Medicine-Hospital Clínic of Barcelona, Faculty of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Jose Cesar Milisenda
- Laboratory of Muscle Research and Mitochondrial Function-CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Department of Internal Medicine-Hospital Clínic of Barcelona, Faculty of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Eduard Tolosa
- Laboratory of Parkinson disease and other Neurodegenerative Movement Disorders: Clinical and Experimental Research, Department of Neurology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Barcelona, Spain.,CIBER de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Maria Jose Marti
- Laboratory of Parkinson disease and other Neurodegenerative Movement Disorders: Clinical and Experimental Research, Department of Neurology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Barcelona, Spain.,CIBER de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ruben Fernández-Santiago
- Laboratory of Parkinson disease and other Neurodegenerative Movement Disorders: Clinical and Experimental Research, Department of Neurology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Barcelona, Spain.,CIBER de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Francesc Cardellach
- Laboratory of Muscle Research and Mitochondrial Function-CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Department of Internal Medicine-Hospital Clínic of Barcelona, Faculty of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Constanza Morén
- Laboratory of Muscle Research and Mitochondrial Function-CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Department of Internal Medicine-Hospital Clínic of Barcelona, Faculty of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
| | - Glòria Garrabou
- Laboratory of Muscle Research and Mitochondrial Function-CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Department of Internal Medicine-Hospital Clínic of Barcelona, Faculty of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
| |
Collapse
|
49
|
Howlett EH, Jensen N, Belmonte F, Zafar F, Hu X, Kluss J, Schüle B, Kaufman BA, Greenamyre JT, Sanders LH. LRRK2 G2019S-induced mitochondrial DNA damage is LRRK2 kinase dependent and inhibition restores mtDNA integrity in Parkinson's disease. Hum Mol Genet 2018; 26:4340-4351. [PMID: 28973664 DOI: 10.1093/hmg/ddx320] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 08/10/2017] [Indexed: 12/19/2022] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are associated with increased risk for developing Parkinson's disease (PD). Previously, we found that LRRK2 G2019S mutation carriers have increased mitochondrial DNA (mtDNA) damage and after zinc finger nuclease-mediated gene mutation correction, mtDNA damage was no longer detectable. While the mtDNA damage phenotype can be unambiguously attributed to the LRRK2 G2019S mutation, the underlying mechanism(s) is unknown. Here, we examine the role of LRRK2 kinase function in LRRK2 G2019S-mediated mtDNA damage, using both genetic and pharmacological approaches in cultured neurons and PD patient-derived cells. Expression of LRRK2 G2019S induced mtDNA damage in primary rat midbrain neurons, but not in cortical neuronal cultures. In contrast, the expression of LRRK2 wild type or LRRK2 D1994A mutant (kinase dead) had no effect on mtDNA damage in either midbrain or cortical neuronal cultures. In addition, human LRRK2 G2019S patient-derived lymphoblastoid cell lines (LCL) demonstrated increased mtDNA damage relative to age-matched controls. Importantly, treatment of LRRK2 G2019S expressing midbrain neurons or patient-derived LRRK2 G2019S LCLs with the LRRK2 kinase inhibitor GNE-7915, either prevented or restored mtDNA damage to control levels. These findings support the hypothesis that LRRK2 G2019S-induced mtDNA damage is LRRK2 kinase activity dependent, uncovering a novel pathological role for this kinase. Blocking or reversing mtDNA damage via LRRK2 kinase inhibition or other therapeutic approaches may be useful to slow PD-associated pathology.
Collapse
Affiliation(s)
- Evan H Howlett
- Department of Neurology, Pittsburgh Institute for Neurodegenerative Diseases
| | - Nicholas Jensen
- Department of Neurology, Pittsburgh Institute for Neurodegenerative Diseases
| | - Frances Belmonte
- Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Faria Zafar
- Parkinson's Institute and Clinical Center, Sunnyvale, CA 94085, USA
| | - Xiaoping Hu
- Department of Neurology, Pittsburgh Institute for Neurodegenerative Diseases
| | - Jillian Kluss
- Department of Neurology, Pittsburgh Institute for Neurodegenerative Diseases
| | - Birgitt Schüle
- Parkinson's Institute and Clinical Center, Sunnyvale, CA 94085, USA
| | - Brett A Kaufman
- Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - J T Greenamyre
- Department of Neurology, Pittsburgh Institute for Neurodegenerative Diseases
| | - Laurie H Sanders
- Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
50
|
Zhao Y, Perera G, Takahashi-Fujigasaki J, Mash DC, Vonsattel JPG, Uchino A, Hasegawa K, Jeremy Nichols R, Holton JL, Murayama S, Dzamko N, Halliday GM. Reduced LRRK2 in association with retromer dysfunction in post-mortem brain tissue from LRRK2 mutation carriers. Brain 2018; 141:486-495. [PMID: 29253086 PMCID: PMC5837795 DOI: 10.1093/brain/awx344] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/03/2017] [Accepted: 10/25/2017] [Indexed: 12/25/2022] Open
Abstract
Missense mutations in leucine-rich repeat kinase 2 (LRRK2) are pathogenic for familial Parkinson's disease. However, it is unknown whether levels of LRRK2 protein in the brain are altered in patients with LRRK2-associated Parkinson's disease. Because LRRK2 mutations are relatively rare, accounting for approximately 1% of all Parkinson's disease, we accessioned cases from five international brain banks to investigate levels of the LRRK2 protein, and other genetically associated Parkinson's disease proteins. Brain tissue was obtained from 17 LRRK2 mutation carriers (12 with the G2019S mutation and five with the I2020T mutation) and assayed by immunoblot. Compared to matched controls and idiopathic Parkinson's disease cases, we found levels of LRRK2 protein were reduced in the LRRK2 mutation cases. We also measured a decrease in two other proteins genetically implicated in Parkinson's disease, the core retromer component, vacuolar protein sorting associated protein 35 (VPS35), and the lysosomal hydrolase, glucocerebrosidase (GBA). Moreover, the classical retromer cargo protein, cation-independent mannose-6-phosphate receptor (MPR300, encoded by IGF2R), was also reduced in the LRRK2 mutation cohort and protein levels of the receptor were correlated to levels of LRRK2. These results provide new data on LRRK2 protein expression in brain tissue from LRRK2 mutation carriers and support a relationship between LRRK2 and retromer dysfunction in LRRK2-associated Parkinson's disease brain.
Collapse
Affiliation(s)
- Ye Zhao
- Brain and Mind Centre, Sydney Medical School, University of Sydney, Camperdown, 2050, Australia
- School of Medical Sciences, University of NSW, Kensington, 2033, Australia
- Neuroscience Research Australia, Randwick, 2031, Australia
| | - Gayathri Perera
- Brain and Mind Centre, Sydney Medical School, University of Sydney, Camperdown, 2050, Australia
- Neuroscience Research Australia, Randwick, 2031, Australia
| | - Junko Takahashi-Fujigasaki
- Department of Neuropathology, Brain Bank for Aging Research, Tokyo Metropolitan Geriatric0 Hospital and Institute of Gerontology, Tokyo, 173-0015, Japan
| | - Deborah C Mash
- University of Miami Brain Endowment Bank™, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
| | - Jean Paul G Vonsattel
- New York Brain Bank, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, 10032, USA
| | - Akiko Uchino
- Department of Neuropathology, Brain Bank for Aging Research, Tokyo Metropolitan Geriatric0 Hospital and Institute of Gerontology, Tokyo, 173-0015, Japan
| | - Kazuko Hasegawa
- Department of Neurology, Sagamihara National Hospital, Kangawa, 252-0315, Japan
| | - R Jeremy Nichols
- Parkinson’s Institute and Clinical Center, Sunnyvale, California, 94085, USA
| | - Janice L Holton
- Queen Square Brain Bank, UCL Institute of Neurology, University College London, London, WC1N 1PJ, UK
| | - Shigeo Murayama
- Department of Neuropathology, Brain Bank for Aging Research, Tokyo Metropolitan Geriatric0 Hospital and Institute of Gerontology, Tokyo, 173-0015, Japan
| | - Nicolas Dzamko
- Brain and Mind Centre, Sydney Medical School, University of Sydney, Camperdown, 2050, Australia
- School of Medical Sciences, University of NSW, Kensington, 2033, Australia
- Neuroscience Research Australia, Randwick, 2031, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, Sydney Medical School, University of Sydney, Camperdown, 2050, Australia
- School of Medical Sciences, University of NSW, Kensington, 2033, Australia
- Neuroscience Research Australia, Randwick, 2031, Australia
| |
Collapse
|