1
|
Wu X, Yang Z, Zou J, Gao H, Shao Z, Li C, Lei P. Protein kinases in neurodegenerative diseases: current understandings and implications for drug discovery. Signal Transduct Target Ther 2025; 10:146. [PMID: 40328798 PMCID: PMC12056177 DOI: 10.1038/s41392-025-02179-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/03/2025] [Accepted: 02/12/2025] [Indexed: 05/08/2025] Open
Abstract
Neurodegenerative diseases (e.g., Alzheimer's, Parkinson's, Huntington's disease, and Amyotrophic Lateral Sclerosis) are major health threats for the aging population and their prevalences continue to rise with the increasing of life expectancy. Although progress has been made, there is still a lack of effective cures to date, and an in-depth understanding of the molecular and cellular mechanisms of these neurodegenerative diseases is imperative for drug development. Protein phosphorylation, regulated by protein kinases and protein phosphatases, participates in most cellular events, whereas aberrant phosphorylation manifests as a main cause of diseases. As evidenced by pharmacological and pathological studies, protein kinases are proven to be promising therapeutic targets for various diseases, such as cancers, central nervous system disorders, and cardiovascular diseases. The mechanisms of protein phosphatases in pathophysiology have been extensively reviewed, but a systematic summary of the role of protein kinases in the nervous system is lacking. Here, we focus on the involvement of protein kinases in neurodegenerative diseases, by summarizing the current knowledge on the major kinases and related regulatory signal transduction pathways implicated in diseases. We further discuss the role and complexity of kinase-kinase networks in the pathogenesis of neurodegenerative diseases, illustrate the advances of clinical applications of protein kinase inhibitors or novel kinase-targeted therapeutic strategies (such as antisense oligonucleotides and gene therapy) for effective prevention and early intervention.
Collapse
Affiliation(s)
- Xiaolei Wu
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhangzhong Yang
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinjun Zou
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chuanzhou Li
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Bichler Z, Vanan S, Zhang Z, Dong Q(S, Lee JWL, Zhang C, Hang L, Jiang M, Padmanabhan P, Saw WT, Zhou Z, Gulyás B, Lim KL, Zeng L, Tan EK. Environmental Factors Exacerbate Parkinsonian Phenotypes in an Asian-Specific Knock-In LRRK2 Risk Variant in Mice. Int J Mol Sci 2025; 26:3556. [PMID: 40332013 PMCID: PMC12027425 DOI: 10.3390/ijms26083556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/30/2025] [Accepted: 04/06/2025] [Indexed: 05/08/2025] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder affecting nearly 10 million people worldwide, and for which no cure is currently known. Mutations in the Leucine-Rich Repeat Kinase 2 (LRRK2) gene, age, as well as environmental factors such as neurotoxin exposure and stress, are known to increase the risk of developing the disease in humans. To investigate the role of a specific Asian variant of the LRRK2 gene to induce susceptibility to stress and trigger PD phenotypes with time, knock-in (KI) mice bearing the human LRRK2 R1628P risk variant have been generated and studied from 2 to 16 months of age in the presence (or absence) of stress insults, including neurotoxin injections and chronic mild stress applied at 3 months of age. Pathophysiological and behavioural phenotypes have been measured at different ages and primary neurons and fibroblast cells were cultured from the KI mouse line and treated with H2O2 to study susceptibility towards oxidative stress in vitro. KI mice displayed specific PD features and these phenotypes were aggravated by environmental stresses. In particular, KI mice developed locomotion impairment and increased constipation. In addition, dopamine-related proteins were dysregulated in KI mice brains: Dopamine transporter (DAT) was decreased in the midbrain and striatum and dopamine levels were increased. Primary fibroblast cells and cortical neurons from KI mice also displayed increased susceptibility to oxidative stress. Therefore, the LRRK2 R1628P KI mice are an excellent model to study the progressive development of PD.
Collapse
Affiliation(s)
- Zoë Bichler
- Behavioural Neuroscience Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore; (Z.B.); (Q.D.)
- Center for Biometric Analysis, The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Sarivin Vanan
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore; (S.V.); (Z.Z.); (J.W.L.L.); (M.J.)
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (P.P.); (B.G.); (K.L.L.)
- Neurodegeneration Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore; (C.Z.); (L.H.)
| | - Zhiwei Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore; (S.V.); (Z.Z.); (J.W.L.L.); (M.J.)
| | - Qianying (Sally) Dong
- Behavioural Neuroscience Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore; (Z.B.); (Q.D.)
| | - Jolene Wei Ling Lee
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore; (S.V.); (Z.Z.); (J.W.L.L.); (M.J.)
- Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore; (W.T.S.); (Z.Z.)
| | - Chengwu Zhang
- Neurodegeneration Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore; (C.Z.); (L.H.)
| | - Liting Hang
- Neurodegeneration Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore; (C.Z.); (L.H.)
| | - Mei Jiang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore; (S.V.); (Z.Z.); (J.W.L.L.); (M.J.)
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan 523808, China
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (P.P.); (B.G.); (K.L.L.)
| | - Wuan Ting Saw
- Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore; (W.T.S.); (Z.Z.)
- Translational Therapeutics Lab, Research Department, National Neuroscience Institute, Singapore 169856, Singapore
| | - Zhidong Zhou
- Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore; (W.T.S.); (Z.Z.)
- Translational Therapeutics Lab, Research Department, National Neuroscience Institute, Singapore 169856, Singapore
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (P.P.); (B.G.); (K.L.L.)
| | - Kah Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (P.P.); (B.G.); (K.L.L.)
- Neurodegeneration Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore; (C.Z.); (L.H.)
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore; (S.V.); (Z.Z.); (J.W.L.L.); (M.J.)
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (P.P.); (B.G.); (K.L.L.)
- Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore; (W.T.S.); (Z.Z.)
| | - Eng King Tan
- Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore; (W.T.S.); (Z.Z.)
- Research Department, National Neuroscience Institute, Singapore General Hospital (SGH) Campus, Singapore 168581, Singapore
- Department of Neurology, National Neuroscience Institute, Singapore 308433, Singapore
| |
Collapse
|
3
|
Zhu C, Herbst S, Lewis PA. Leucine-rich repeat kinase 2 at a glance. J Cell Sci 2023; 136:jcs259724. [PMID: 37698513 PMCID: PMC10508695 DOI: 10.1242/jcs.259724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a multidomain scaffolding protein with dual guanosine triphosphatase (GTPase) and kinase enzymatic activities, providing this protein with the capacity to regulate a multitude of signalling pathways and act as a key mediator of diverse cellular processes. Much of the interest in LRRK2 derives from mutations in the LRRK2 gene being the most common genetic cause of Parkinson's disease, and from the association of the LRRK2 locus with a number of other human diseases, including inflammatory bowel disease. Therefore, the LRRK2 research field has focused on the link between LRRK2 and pathology, with the aim of uncovering the underlying mechanisms and, ultimately, finding novel therapies and treatments to combat them. From the biochemical and cellular functions of LRRK2, to its relevance to distinct disease mechanisms, this Cell Science at a Glance article and the accompanying poster deliver a snapshot of our current understanding of LRRK2 function, dysfunction and links to disease.
Collapse
Affiliation(s)
- Christiane Zhu
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London NW1 0TU, UK
- Department of Neurodegenerative diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Susanne Herbst
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London NW1 0TU, UK
- Department of Neurodegenerative diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Patrick A. Lewis
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London NW1 0TU, UK
- Department of Neurodegenerative diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
4
|
Mansour HM, Mohamed AF, El-Khatib AS, Khattab MM. Kinases control of regulated cell death revealing druggable targets for Parkinson's disease. Ageing Res Rev 2023; 85:101841. [PMID: 36608709 DOI: 10.1016/j.arr.2022.101841] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/31/2022] [Accepted: 12/31/2022] [Indexed: 01/05/2023]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder in the world. Motor impairment seen in PD is associated with dopaminergic neurotoxicity in the striatum, and dopaminergic neuronal death in the substantia nigra pars compacta. Cell death has a significant effect on the development and progression of PD. Extensive research over the last few decades has unveiled new regulated cell death (RCD) mechanisms that are not dependent on apoptosis such as necroptosis, ferroptosis, and others. In this review, we will overview the mechanistic pathways of different types of RCD. Unlike accidental cell death, RCD subroutines can be regulated and the RCD-associated kinases are potential druggable targets. Hence, we will address an overview and analysis of different kinases regulating apoptosis such as receptor-interacting protein kinase 1 (RIPK-1), RIPK3, mixed lineage kinase (MLK), Ataxia telangiectasia muted (ATM), cyclin-dependent kinase (CDK), death-associated protein kinase 1 (DAPK1), Apoptosis-signaling kinase-1 (ASK-1), and Leucine-rich repeat kinase-2 (LRRK2). In addition to the role of RIPK1, RIPK3, and Mixed Lineage Kinase Domain like Pseudokinase (MLKL) in necroptosis. We also overview functions of AMP-kinase (AMPK), protein kinase C (PKC), RIPK3, and ATM in ferroptosis. We will recap the anti-apoptotic, anti-necroptotic, and anti-ferroptotic effects of different kinase inhibitors in different models of PD. Finally, we will discuss future challenges in the repositioning of kinase inhibitors in PD. In conclusion, this review kicks-start targeting RCD from a kinases perspective, opening novel therapeutic disease-modifying therapeutic avenues for PD.
Collapse
Affiliation(s)
| | - Ahmed F Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Aiman S El-Khatib
- Egyptian Drug Authority, EDA, Giza, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
5
|
Garrido A, Pérez‐Sisqués L, Simonet C, Campoy‐Campos G, Solana‐Balaguer J, Martín‐Flores N, Fernández M, Soto M, Obiang D, Cámara A, Valldeoriola F, Muñoz E, Compta Y, Pérez‐Navarro E, Alberch J, Tolosa E, Martí M, Ezquerra M, Malagelada C, Fernández‐Santiago R. Increased Phospho-AKT in Blood Cells from LRRK2 G2019S Mutation Carriers. Ann Neurol 2022; 92:888-894. [PMID: 35929078 PMCID: PMC9827833 DOI: 10.1002/ana.26469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/15/2022] [Accepted: 07/29/2022] [Indexed: 01/12/2023]
Abstract
The purpose of this study was to investigate whether differential phosphorylation states of blood markers can identify patients with LRRK2 Parkinson's disease (PD). We assessed phospho(P)-Ser-935-LRRK2 and P-Ser-473-AKT levels in peripheral blood cells from patients with G2019S LRRK2-associated PD (L2PD, n = 31), G2019S LRRK2 non-manifesting carriers (L2NMC, n = 26), idiopathic PD (iPD, n = 25), and controls (n = 40, total n = 122). We found no differences at P-Ser-935-LRRK2 between groups but detected a specific increase of P-Ser-473-AKT levels in all G2019S carriers, either L2PD or L2NMC, absent in iPD. Although insensitive to LRRK2 inhibition, our study identifies P-Ser-473-AKT as an endogenous candidate biomarker for peripheral inflammation in G2019S carriers using accessible blood cells. ANN NEUROL 2022;92:888-894.
Collapse
Affiliation(s)
- Alicia Garrido
- Parkinson Disease and Movement Disorders Unit, Neurology ServiceInstitut Clínic de Neurociències, Hospital Clínic de BarcelonaBarcelonaSpain,Laboratory of Parkinson Disease and Other Neurodegenerative Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain
| | - Leticia Pérez‐Sisqués
- Department of Biomedicine, Faculty of MedicineUniversitat de BarcelonaBarcelonaSpain,Institut de NeurociènciesUniversitat de BarcelonaBarcelonaSpain
| | - Cristina Simonet
- Parkinson Disease and Movement Disorders Unit, Neurology ServiceInstitut Clínic de Neurociències, Hospital Clínic de BarcelonaBarcelonaSpain,Laboratory of Parkinson Disease and Other Neurodegenerative Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain
| | - Genís Campoy‐Campos
- Department of Biomedicine, Faculty of MedicineUniversitat de BarcelonaBarcelonaSpain,Institut de NeurociènciesUniversitat de BarcelonaBarcelonaSpain
| | - Júlia Solana‐Balaguer
- Department of Biomedicine, Faculty of MedicineUniversitat de BarcelonaBarcelonaSpain,Institut de NeurociènciesUniversitat de BarcelonaBarcelonaSpain
| | - Núria Martín‐Flores
- Department of Biomedicine, Faculty of MedicineUniversitat de BarcelonaBarcelonaSpain,Institut de NeurociènciesUniversitat de BarcelonaBarcelonaSpain
| | - Manel Fernández
- Parkinson Disease and Movement Disorders Unit, Neurology ServiceInstitut Clínic de Neurociències, Hospital Clínic de BarcelonaBarcelonaSpain,Laboratory of Parkinson Disease and Other Neurodegenerative Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain,Institut de NeurociènciesUniversitat de BarcelonaBarcelonaSpain
| | - Marta Soto
- Parkinson Disease and Movement Disorders Unit, Neurology ServiceInstitut Clínic de Neurociències, Hospital Clínic de BarcelonaBarcelonaSpain,Laboratory of Parkinson Disease and Other Neurodegenerative Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain
| | - Donina Obiang
- Parkinson Disease and Movement Disorders Unit, Neurology ServiceInstitut Clínic de Neurociències, Hospital Clínic de BarcelonaBarcelonaSpain,Laboratory of Parkinson Disease and Other Neurodegenerative Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain
| | - Ana Cámara
- Parkinson Disease and Movement Disorders Unit, Neurology ServiceInstitut Clínic de Neurociències, Hospital Clínic de BarcelonaBarcelonaSpain,Laboratory of Parkinson Disease and Other Neurodegenerative Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain
| | - Francesc Valldeoriola
- Parkinson Disease and Movement Disorders Unit, Neurology ServiceInstitut Clínic de Neurociències, Hospital Clínic de BarcelonaBarcelonaSpain,Laboratory of Parkinson Disease and Other Neurodegenerative Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain
| | - Esteban Muñoz
- Parkinson Disease and Movement Disorders Unit, Neurology ServiceInstitut Clínic de Neurociències, Hospital Clínic de BarcelonaBarcelonaSpain,Laboratory of Parkinson Disease and Other Neurodegenerative Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain
| | - Yaroslau Compta
- Parkinson Disease and Movement Disorders Unit, Neurology ServiceInstitut Clínic de Neurociències, Hospital Clínic de BarcelonaBarcelonaSpain,Laboratory of Parkinson Disease and Other Neurodegenerative Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain
| | - Esther Pérez‐Navarro
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain,Department of Biomedicine, Faculty of MedicineUniversitat de BarcelonaBarcelonaSpain,Institut de NeurociènciesUniversitat de BarcelonaBarcelonaSpain
| | - Jordi Alberch
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain,Department of Biomedicine, Faculty of MedicineUniversitat de BarcelonaBarcelonaSpain,Institut de NeurociènciesUniversitat de BarcelonaBarcelonaSpain
| | - Eduardo Tolosa
- Parkinson Disease and Movement Disorders Unit, Neurology ServiceInstitut Clínic de Neurociències, Hospital Clínic de BarcelonaBarcelonaSpain,Laboratory of Parkinson Disease and Other Neurodegenerative Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain
| | - María‐José Martí
- Parkinson Disease and Movement Disorders Unit, Neurology ServiceInstitut Clínic de Neurociències, Hospital Clínic de BarcelonaBarcelonaSpain,Laboratory of Parkinson Disease and Other Neurodegenerative Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain
| | - Mario Ezquerra
- Parkinson Disease and Movement Disorders Unit, Neurology ServiceInstitut Clínic de Neurociències, Hospital Clínic de BarcelonaBarcelonaSpain,Laboratory of Parkinson Disease and Other Neurodegenerative Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain
| | - Cristina Malagelada
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain,Department of Biomedicine, Faculty of MedicineUniversitat de BarcelonaBarcelonaSpain,Institut de NeurociènciesUniversitat de BarcelonaBarcelonaSpain
| | - Rubén Fernández‐Santiago
- Parkinson Disease and Movement Disorders Unit, Neurology ServiceInstitut Clínic de Neurociències, Hospital Clínic de BarcelonaBarcelonaSpain,Laboratory of Parkinson Disease and Other Neurodegenerative Movement DisordersInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018‐ISCIII)BarcelonaSpain,Department of Biomedicine, Faculty of MedicineUniversitat de BarcelonaBarcelonaSpain
| |
Collapse
|
6
|
Ali MZ, Dholaniya PS. Oxidative phosphorylation mediated pathogenesis of Parkinson's disease and its implication via Akt signaling. Neurochem Int 2022; 157:105344. [PMID: 35483538 DOI: 10.1016/j.neuint.2022.105344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 12/21/2022]
Abstract
Substantia Nigra Pars-compacta (SNpc), in the basal ganglion region, is a primary source of dopamine release. These dopaminergic neurons require more energy than other neurons, as they are highly arborized and redundant. Neurons meet most of their energy demand (∼90%) from mitochondria. Oxidative phosphorylation (OxPhos) is the primary pathway for energy production. Many genes involved in Parkinson's disease (PD) have been associated with OxPhos, especially complex I. Abrogation in complex I leads to reduced ATP formation in these neurons, succumbing to death by inducing apoptosis. This review discusses the interconnection between complex I-associated PD genes and specific mitochondrial metabolic factors (MMFs) of OxPhos. Interestingly, all the complex I-associated PD genes discussed here have been linked to the Akt signaling pathway; thus, neuron survival is promoted and smooth mitochondrial function is ensured. Any changes in these genes disrupt the Akt pathway, which hampers the opening of the permeability transition pore (PTP) via GSK3β dephosphorylation; promotes destabilization of OxPhos; and triggers the release of pro-apoptotic factors.
Collapse
Affiliation(s)
- Md Zainul Ali
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India
| | - Pankaj Singh Dholaniya
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India.
| |
Collapse
|
7
|
Ecovoiu AA, Ratiu AC, Micheu MM, Chifiriuc MC. Inter-Species Rescue of Mutant Phenotype-The Standard for Genetic Analysis of Human Genetic Disorders in Drosophila melanogaster Model. Int J Mol Sci 2022; 23:2613. [PMID: 35269756 PMCID: PMC8909942 DOI: 10.3390/ijms23052613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 11/16/2022] Open
Abstract
Drosophila melanogaster (the fruit fly) is arguably a superstar of genetics, an astonishing versatile experimental model which fueled no less than six Nobel prizes in medicine. Nowadays, an evolving research endeavor is to simulate and investigate human genetic diseases in the powerful D. melanogaster platform. Such a translational experimental strategy is expected to allow scientists not only to understand the molecular mechanisms of the respective disorders but also to alleviate or even cure them. In this regard, functional gene orthology should be initially confirmed in vivo by transferring human or vertebrate orthologous transgenes in specific mutant backgrounds of D. melanogaster. If such a transgene rescues, at least partially, the mutant phenotype, then it qualifies as a strong candidate for modeling the respective genetic disorder in the fruit fly. Herein, we review various examples of inter-species rescue of relevant mutant phenotypes of the fruit fly and discuss how these results recommend several human genes as candidates to study and validate genetic variants associated with human diseases. We also consider that a wider implementation of this evolutionist exploratory approach as a standard for the medicine of genetic disorders would allow this particular field of human health to advance at a faster pace.
Collapse
Affiliation(s)
- Alexandru Al. Ecovoiu
- Department of Genetics, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania;
| | - Attila Cristian Ratiu
- Department of Genetics, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania;
| | - Miruna Mihaela Micheu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania;
| | - Mariana Carmen Chifiriuc
- The Research Institute of the University of Bucharest and Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania;
| |
Collapse
|
8
|
Yu J, Cho E, Kwon H, Jeon J, Seong Sin J, Kwon Park J, Kim JS, Woong Choi J, Jin Park S, Jun M, Choon Lee Y, Hoon Ryu J, Lee J, Moon M, Lee S, Hyun Cho J, Hyun Kim D. Akt and calcium-permeable AMPA receptor are involved in the effect of pinoresinol on amyloid β-induced synaptic plasticity and memory deficits. Biochem Pharmacol 2021; 184:114366. [PMID: 33310049 DOI: 10.1016/j.bcp.2020.114366] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/08/2020] [Accepted: 12/08/2020] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative disorders characterized by memory deficits. Although no drug has given promising results, synaptic dysfunction-modulating agents might be considered potential candidates for alleviating this disorder. Pinoresinol, a lignan found in Forsythia suspensa, is a memory-enhancing agent with excitatory synaptic activation. In the present study, we tested whether pinoresinol reduces learning and memory and excitatory synaptic deficits in an amyloid β (Aβ)-induced AD-like mouse model. Pinoresinol enhanced hippocampal long-term potentiation (LTP) through calcium-permeable AMPA receptor, which was mediated by Akt activation. Moreover, pinoresinol ameliorated LTP deficits in amyloid β (Aβ)-treated hippocampal slices via Akt signaling. Oral administration of pinoresinol ameliorated Aβ-induced memory deficits without sensory dysfunction. Moreover, AD-like pathology, including neuroinflammation and synaptic deficit, were ameliorated by pinoresinol administration. Collectively, pinoresinol may be a good candidate for AD therapy by modulating synaptic functions.
Collapse
Affiliation(s)
- Jimin Yu
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Eunbi Cho
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Huiyoung Kwon
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Jieun Jeon
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Jae Seong Sin
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Jun Kwon Park
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Ji-Su Kim
- Primate Resources Center (PRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongup-si, Jeollabuk-do 56216, Republic of Korea
| | - Ji Woong Choi
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, Republic of Korea
| | - Se Jin Park
- School of Natural Resources and Environmental Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Mira Jun
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Young Choon Lee
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea; Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | - Jong Hoon Ryu
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jeongwon Lee
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | - Seungheon Lee
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea.
| | - Jong Hyun Cho
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea; Laboratory of Anti-viral Drug Discovery, Dong-A University, Busan, Republic of Korea.
| | - Dong Hyun Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea; Laboratory of Anti-viral Drug Discovery, Dong-A University, Busan, Republic of Korea; Institute of Convergence Bio-Health, Dong-A University, Busan, Republic of Korea.
| |
Collapse
|
9
|
Panagiotakopoulou V, Ivanyuk D, De Cicco S, Haq W, Arsić A, Yu C, Messelodi D, Oldrati M, Schöndorf DC, Perez MJ, Cassatella RP, Jakobi M, Schneiderhan-Marra N, Gasser T, Nikić-Spiegel I, Deleidi M. Interferon-γ signaling synergizes with LRRK2 in neurons and microglia derived from human induced pluripotent stem cells. Nat Commun 2020; 11:5163. [PMID: 33057020 PMCID: PMC7560616 DOI: 10.1038/s41467-020-18755-4] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease-associated kinase LRRK2 has been linked to IFN type II (IFN-γ) response in infections and to dopaminergic neuronal loss. However, whether and how LRRK2 synergizes with IFN-γ remains unclear. In this study, we employed dopaminergic neurons and microglia differentiated from patient-derived induced pluripotent stem cells carrying LRRK2 G2019S, the most common Parkinson's disease-associated mutation. We show that IFN-γ enhances the LRRK2 G2019S-dependent negative regulation of AKT phosphorylation and NFAT activation, thereby increasing neuronal vulnerability to immune challenge. Mechanistically, LRRK2 G2019S suppresses NFAT translocation via calcium signaling and possibly through microtubule reorganization. In microglia, LRRK2 modulates cytokine production and the glycolytic switch in response to IFN-γ in an NFAT-independent manner. Activated LRRK2 G2019S microglia cause neurite shortening, indicating that LRRK2-driven immunological changes can be neurotoxic. We propose that synergistic LRRK2/IFN-γ activation serves as a potential link between inflammation and neurodegeneration in Parkinson's disease.
Collapse
Affiliation(s)
- Vasiliki Panagiotakopoulou
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, 72076, Germany
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, 72076, Germany
| | - Dina Ivanyuk
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, 72076, Germany
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, 72076, Germany
| | - Silvia De Cicco
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, 72076, Germany
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, 72076, Germany
| | - Wadood Haq
- Centre for Ophthalmology, Institute for Ophthalmic Research University of Tübingen, University of Tübingen, Tübingen, 72076, Germany
| | - Aleksandra Arsić
- Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, 72076, Germany
| | - Cong Yu
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, 72076, Germany
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, 72076, Germany
| | - Daria Messelodi
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, 72076, Germany
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, 72076, Germany
| | - Marvin Oldrati
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, 72076, Germany
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, 72076, Germany
| | - David C Schöndorf
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, 72076, Germany
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, 72076, Germany
| | - Maria-Jose Perez
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, 72076, Germany
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, 72076, Germany
| | - Ruggiero Pio Cassatella
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, 72076, Germany
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, 72076, Germany
| | - Meike Jakobi
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Nicole Schneiderhan-Marra
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Thomas Gasser
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, 72076, Germany
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, 72076, Germany
| | - Ivana Nikić-Spiegel
- Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, 72076, Germany
| | - Michela Deleidi
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, 72076, Germany.
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, 72076, Germany.
| |
Collapse
|
10
|
Furlong RM, Lindsay A, Anderson KE, Hawkins PT, Sullivan AM, O'Neill C. The Parkinson's disease gene PINK1 activates Akt via PINK1 kinase-dependent regulation of the phospholipid PI(3,4,5)P 3. J Cell Sci 2019; 132:jcs.233221. [PMID: 31540955 DOI: 10.1242/jcs.233221] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/12/2019] [Indexed: 12/12/2022] Open
Abstract
Akt signalling is central to cell survival, metabolism, protein and lipid homeostasis, and is impaired in Parkinson's disease (PD). Akt activation is reduced in the brain in PD, and by many PD-causing genes, including PINK1 This study investigated the mechanisms by which PINK1 regulates Akt signalling. Our results reveal for the first time that PINK1 constitutively activates Akt in a PINK1-kinase dependent manner in the absence of growth factors, and enhances Akt activation in normal growth medium. In PINK1-modified MEFs, agonist-induced Akt signalling failed in the absence of PINK1, due to PINK1 kinase-dependent increases in PI(3,4,5)P3 at both plasma membrane and Golgi being significantly impaired. In the absence of PINK1, PI(3,4,5)P3 levels did not increase in the Golgi, and there was significant Golgi fragmentation, a recognised characteristic of PD neuropathology. PINK1 kinase activity protected the Golgi from fragmentation in an Akt-dependent fashion. This study demonstrates a new role for PINK1 as a primary upstream activator of Akt via PINK1 kinase-dependent regulation of its primary activator PI(3,4,5)P3, providing novel mechanistic information on how loss of PINK1 impairs Akt signalling in PD.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Rachel M Furlong
- School of Biochemistry and Cell Biology, Biosciences Institute, University College Cork, Cork City T12 YT20, Ireland.,Department of Anatomy and Neuroscience, Western Gateway Building, University College Cork, Cork City T12 XF62, Ireland.,Cork NeuroScience Centre, University College Cork, Cork City T12 YT20, Ireland
| | - Andrew Lindsay
- School of Biochemistry and Cell Biology, Biosciences Institute, University College Cork, Cork City T12 YT20, Ireland
| | - Karen E Anderson
- Signalling Programme, Babraham Institute, Cambridge CB22 3AT, UK
| | | | - Aideen M Sullivan
- Department of Anatomy and Neuroscience, Western Gateway Building, University College Cork, Cork City T12 XF62, Ireland.,Cork NeuroScience Centre, University College Cork, Cork City T12 YT20, Ireland
| | - Cora O'Neill
- School of Biochemistry and Cell Biology, Biosciences Institute, University College Cork, Cork City T12 YT20, Ireland .,Cork NeuroScience Centre, University College Cork, Cork City T12 YT20, Ireland
| |
Collapse
|
11
|
Jęśko H, Stępień A, Lukiw WJ, Strosznajder RP. The Cross-Talk Between Sphingolipids and Insulin-Like Growth Factor Signaling: Significance for Aging and Neurodegeneration. Mol Neurobiol 2019; 56:3501-3521. [PMID: 30140974 PMCID: PMC6476865 DOI: 10.1007/s12035-018-1286-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/25/2018] [Indexed: 12/20/2022]
Abstract
Bioactive sphingolipids: sphingosine, sphingosine-1-phosphate (S1P), ceramide, and ceramide-1-phosphate (C1P) are increasingly implicated in cell survival, proliferation, differentiation, and in multiple aspects of stress response in the nervous system. The opposite roles of closely related sphingolipid species in cell survival/death signaling is reflected in the concept of tightly controlled sphingolipid rheostat. Aging has a complex influence on sphingolipid metabolism, disturbing signaling pathways and the properties of lipid membranes. A metabolic signature of stress resistance-associated sphingolipids correlates with longevity in humans. Moreover, accumulating evidence suggests extensive links between sphingolipid signaling and the insulin-like growth factor I (IGF-I)-Akt-mTOR pathway (IIS), which is involved in the modulation of aging process and longevity. IIS integrates a wide array of metabolic signals, cross-talks with p53, nuclear factor κB (NF-κB), or reactive oxygen species (ROS) and influences gene expression to shape the cellular metabolic profile and stress resistance. The multiple connections between sphingolipids and IIS signaling suggest possible engagement of these compounds in the aging process itself, which creates a vulnerable background for the majority of neurodegenerative disorders.
Collapse
Affiliation(s)
- Henryk Jęśko
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Pawińskiego, 5, 02-106, Poland
| | - Adam Stępień
- Central Clinical Hospital of the Ministry of National Defense, Department of Neurology, Military Institute of Medicine, Warsaw, Szaserów, 128, 04-141, Poland
| | - Walter J Lukiw
- LSU Neuroscience Center and Departments of Neurology and Ophthalmology, Louisiana State University School of Medicine, New Orleans, USA
| | - Robert P Strosznajder
- Laboratory of Preclinical Research and Environmental Agents, Department of Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Pawińskiego, 5, 02-106, Poland.
| |
Collapse
|
12
|
Zhou ZD, Selvaratnam T, Lee JCT, Chao YX, Tan EK. Molecular targets for modulating the protein translation vital to proteostasis and neuron degeneration in Parkinson's disease. Transl Neurodegener 2019; 8:6. [PMID: 30740222 PMCID: PMC6360798 DOI: 10.1186/s40035-019-0145-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/14/2019] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder, which is characterized by the progressive loss of dopaminergic neurons in the Substantia Nigra pars compacta concomitant with Lewy body formation in affected brain areas. The detailed pathogenic mechanisms underlying the selective loss of dopaminergic neurons in PD are unclear, and no drugs or treatments have been developed to alleviate progressive dopaminergic neuron degeneration in PD. However, the formation of α-synuclein-positive protein aggregates in Lewy body has been identified as a common pathological feature of PD, possibly stemming from the consequence of protein misfolding and dysfunctional proteostasis. Proteostasis is the mechanism for maintaining protein homeostasis via modulation of protein translation, enhancement of chaperone capacity and the prompt clearance of misfolded protein by the ubiquitin proteasome system and autophagy. Deregulated protein translation and impaired capacities of chaperone or protein degradation can disturb proteostasis processes, leading to pathological protein aggregation and neurodegeneration in PD. In recent years, multiple molecular targets in the modulation of protein translation vital to proteostasis and dopaminergic neuron degeneration have been identified. The potential pathophysiological and therapeutic significance of these molecular targets to neurodegeneration in PD is highlighted.
Collapse
Affiliation(s)
- Zhi Dong Zhou
- Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
- Signature Research Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School Singapore, 8 College Road, Singapore, Singapore
| | - Thevapriya Selvaratnam
- Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Ji Chao Tristan Lee
- Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Yin Xia Chao
- Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Eng-King Tan
- Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
- Department of Neurology, Singapore General Hospital, Outram Road, Singapore, 169608 Singapore
- Signature Research Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School Singapore, 8 College Road, Singapore, Singapore
| |
Collapse
|
13
|
Roles of Autophagy-Related Genes in the Pathogenesis of Inflammatory Bowel Disease. Cells 2019; 8:cells8010077. [PMID: 30669622 PMCID: PMC6356351 DOI: 10.3390/cells8010077] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/09/2019] [Accepted: 01/11/2019] [Indexed: 12/13/2022] Open
Abstract
Autophagy is an intracellular catabolic process that is essential for a variety of cellular responses. Due to its role in the maintenance of biological homeostasis in conditions of stress, dysregulation or disruption of autophagy may be linked to human diseases such as inflammatory bowel disease (IBD). IBD is a complicated inflammatory colitis disorder; Crohn’s disease and ulcerative colitis are the principal types. Genetic studies have shown the clinical relevance of several autophagy-related genes (ATGs) in the pathogenesis of IBD. Additionally, recent studies using conditional knockout mice have led to a comprehensive understanding of ATGs that affect intestinal inflammation, Paneth cell abnormality and enteric pathogenic infection during colitis. In this review, we discuss the various ATGs involved in macroautophagy and selective autophagy, including ATG16L1, IRGM, LRRK2, ATG7, p62, optineurin and TFEB in the maintenance of intestinal homeostasis. Although advances have been made regarding the involvement of ATGs in maintaining intestinal homeostasis, determining the precise contribution of autophagy has remained elusive. Recent efforts based on direct targeting of ATGs and autophagy will further facilitate the development of new therapeutic opportunities for IBD.
Collapse
|
14
|
Zhang Y, Xiang Y, Wang X, Zhu L, Li H, Wang S, Pan X, Zhao H. Cerebral dopamine neurotrophic factor protects microglia by combining with AKT and by regulating FoxO1/mTOR signaling during neuroinflammation. Biomed Pharmacother 2019; 109:2278-2284. [DOI: 10.1016/j.biopha.2018.11.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 11/06/2018] [Accepted: 11/06/2018] [Indexed: 01/13/2023] Open
|
15
|
The role of LRRK2 in cell signalling. Biochem Soc Trans 2018; 47:197-207. [PMID: 30578345 DOI: 10.1042/bst20180464] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/13/2018] [Accepted: 11/19/2018] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is a common late-onset neurodegenerative disorder known primarily for its motor features. Mutations and risk variants in LRRK2 cause familial and idiopathic forms of PD. Mutations segregating with disease are found in the LRRK2 GTPase and kinase domains, affecting catalytic activity and protein-protein interactions. This likely results in an overall gain of LRRK2 cell signalling function contributing to PD pathogenesis. This concept supports the development of LRRK2 kinase inhibitors as disease-modifying treatments, at least for a subset of patients. However, the function of LRRK2 as a cell signalling protein with two catalytic and several protein-protein interaction domains is highly complex. For example, LRRK2 plays important roles in several inflammatory diseases, raising the possibility that it may mediate immune responses in PD. Consistently, LRRK2-mediated cell signalling was not only shown to be important for neuronal function, including neuronal development and homeostasis, but also for peripheral and central immune responses. The catalytic activity of LRRK2 is regulated by autophosphorylation, protein monomer/dimer cycling, and upstream kinases and GTPases, affecting its subcellular localisation and downstream signalling. Part of LRRK2-mediated signalling is likely facilitated by Rab protein phosphorylation, affecting primarily membrane trafficking, including vesicle release at the trans-Golgi network. However, LRRK2 also displays intrinsic GTPase activity and functions as a signalling scaffold. As an example, LRRK2 was suggested to be part of the NRON complex and β-catenin destruction complex, inhibiting NFAT and canonical Wnt signalling, respectively. In summary, continuous research into LRRK2 signalling function contributes to novel diagnostic and therapeutic concepts in PD.
Collapse
|
16
|
Dong Z, Chu G, Sima Y, Chen G. Djhsp90s are crucial regulators during planarian regeneration and tissue homeostasis. Biochem Biophys Res Commun 2018; 498:723-728. [PMID: 29555472 DOI: 10.1016/j.bbrc.2018.03.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/07/2018] [Indexed: 12/11/2022]
Abstract
Heat shock protein 90 family members (HSP90s), as molecular chaperones, have conserved roles in the physiological processes of eukaryotes regulating cytoprotection, increasing host resistance and so on. However, whether HSP90s affect regeneration in animals is unclear. Planarians are emerging models for studying regeneration in vivo. Here, the roles of three hsp90 genes from planarian Dugesia japonica are investigated by WISH and RNAi. The results show that: (1) Djhsp90s expressions are induced by heat and cold shock, tissue damage and ionic liquid; (2) Djhsp90s mRNA are mainly distributed each side of the body in intact worms as well as blastemas in regenerative worms; (3) the worms show head regression, lysis, the body curling and the regeneration arrest or even failure after Djhsp90s RNAi; (4) Djhsp90s are involved in autophagy and locomotion of the body. The research results suggest that Djhsp90s are not only conserved in cytoprotection, but also involved in homeostasis maintenance and regeneration process by regulating different pathways in planarians.
Collapse
Affiliation(s)
- Zimei Dong
- College of Life Science, Henan Normal University, Xinxiang, 453007, Henan, China
| | - Gengbo Chu
- College of Life Science, Henan Normal University, Xinxiang, 453007, Henan, China
| | - Yingxu Sima
- College of Life Science, Henan Normal University, Xinxiang, 453007, Henan, China
| | - Guangwen Chen
- College of Life Science, Henan Normal University, Xinxiang, 453007, Henan, China.
| |
Collapse
|
17
|
MiR-503 modulates epithelial-mesenchymal transition in silica-induced pulmonary fibrosis by targeting PI3K p85 and is sponged by lncRNA MALAT1. Sci Rep 2017; 7:11313. [PMID: 28900284 PMCID: PMC5596016 DOI: 10.1038/s41598-017-11904-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/31/2017] [Indexed: 12/19/2022] Open
Abstract
Silicosis is a kind of chronic, progressive and incurable lung fibrotic diseases with largely unknown and complex pathogenesis and molecular mechanisms. Mounting evidence suggests that microRNAs (miRNAs, miRs) are involved in the pathogenesis of silicosis. Our previous study based on miRNA microarray had shown that the expression levels of miR-503 were down-regulated in mouse lung tissues of silica-induced pulmonary fibrosis. Here, we validated the decreased expression of miR-503 in the fibrotic mouse lung tissues, human bronchial epithelial cells (HBE) and human lung adenocarcinoma A549 cells which were exposed to silica. In addition, overexpressed miR-503 inhibited silica-induced pulmonary fibrosis by attenuating the severity and the distribution of lesions in vivo and limiting the process of epithelial-mesenchymal transition (EMT) in vitro. Our molecular study further demonstrated that PI3K p85 is one of the target genes of miR-503 and the downstream molecules (Akt, mTOR and Snail) are tightly associated with EMT. Furthermore, the up-regulated lncRNA Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), acted as a competing endogenous RNA (ceRNA), can directly bound to miR-503, which indicated that lncRNA MALAT1 may modulate the expression of miR-503 thus triggering the activation of downstream fibrotic signaling pathways. Taken together, our data suggested that MALAT1-miR-503-PI3K/Akt/mTOR/Snail pathway plays critical roles in silica-induced pulmonary fibrosis.
Collapse
|
18
|
Lu Q, Schnitzler GR, Vallaster CS, Ueda K, Erdkamp S, Briggs CE, Iyer LK, Jaffe IZ, Karas RH. Unliganded estrogen receptor alpha regulates vascular cell function and gene expression. Mol Cell Endocrinol 2017; 442:12-23. [PMID: 27888004 DOI: 10.1016/j.mce.2016.11.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/03/2016] [Accepted: 11/21/2016] [Indexed: 01/15/2023]
Abstract
The unliganded form of the estrogen receptor is generally thought to be inactive. Our prior studies, however, suggested that unliganded estrogen receptor alpha (ERα) exacerbates adverse vascular injury responses in mice. Here, we show that the presence of unliganded ERα decreases vascular endothelial cell (EC) migration and proliferation, increases smooth muscle cell (SMC) proliferation, and increases inflammatory responses in cultured ECs and SMCs. Unliganded ERα also regulates many genes in vascular ECs and mouse aorta. Activation of ERα by E2 reverses the cell physiological effects of unliganded ERα, and promotes gene regulatory effects that are predicted to counter the effects of unliganded ERα. These results reveal that the unliganded form of ERα is not inert, but significantly impacts gene expression and physiology of vascular cells. Furthermore, they indicate that the cardiovascular protective effects of estrogen may be connected to its ability to counteract these effects of unliganded ERα.
Collapse
Affiliation(s)
- Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Gavin R Schnitzler
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA.
| | - Caroline S Vallaster
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Kazutaka Ueda
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Stephanie Erdkamp
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Christine E Briggs
- Tufts Center for Neuroscience Research, Neuroscience Department, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Lakshmanan K Iyer
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Richard H Karas
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA.
| |
Collapse
|
19
|
Cross-talk between LRRK2 and PKA: implication for Parkinson's disease? Biochem Soc Trans 2017; 45:261-267. [DOI: 10.1042/bst20160396] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/12/2016] [Accepted: 12/19/2016] [Indexed: 01/16/2023]
Abstract
Evidence indicates that leucine-rich repeat kinase 2 (LRRK2) controls multiple processes in neurons and glia cells. Deregulated LRRK2 activity due to gene mutation represents the most common cause of autosomal dominant Parkinson's disease (PD). Protein kinase A (PKA)-mediated signaling is a key regulator of brain function. PKA-dependent pathways play an important role in brain homeostasis, neuronal development, synaptic plasticity, control of microglia activation and inflammation. On the other hand, a decline of PKA signaling was shown to contribute to the progression of several neurodegenerative diseases, including PD. In this review, we will discuss the accumulating evidence linking PKA and LRRK2 in neuron and microglia functions, and offer an overview of the enigmatic cross-talk between these two kinases with molecular and cellular implications.
Collapse
|
20
|
Hosseinzadeh Z, Singh Y, Shimshek DR, van der Putten H, Wagner CA, Lang F. Leucine-Rich Repeat Kinase 2 (Lrrk2)-Sensitive Na +/K + ATPase Activity in Dendritic Cells. Sci Rep 2017; 7:41117. [PMID: 28120865 PMCID: PMC5264149 DOI: 10.1038/srep41117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/14/2016] [Indexed: 12/18/2022] Open
Abstract
Leucine-rich repeat kinase 2 (Lrrk2) has been implicated in the pathophysiology of Parkinson's disease. Lrrk2 is expressed in diverse cells including neurons and dendritic cells (DCs). In DCs Lrrk2 was shown to up-regulate Na+/Ca2+-exchanger activity. The elimination of Ca2+ by Na+/Ca2+ -exchangers requires maintenance of the Na+ gradient by the Na+/K+ -ATPase. The present study thus explored whether Lrrk2 impacts on Na+/K+ -ATPase expression and function. To this end DCs were isolated from gene-targeted mice lacking Lrrk2 (Lrrk2-/-) and their wild-type littermates (Lrrk2+/+). Na+/K+ -ATPase activity was estimated from K+ induced, ouabain sensitive, current determined by whole cell patch clamp. Na+/K+ -ATPase α1 subunit transcript and protein levels were determined by RT-qPCR and flow cytometry. As a result, the K+ induced current was significantly smaller in Lrrk2-/- than in Lrrk2+/+ DCs and was completely abolished by ouabain (100 μM) in both genotypes. The K+ induced, ouabain sensitive, current in Lrrk2+/+ DCs was significantly blunted by Lrrk2 inhibitor GSK2578215A (1 μM, 24 hours). The Na+/K+ -ATPase α1 subunit transcript and protein levels were significantly lower in Lrrk2-/- than in Lrrk2+/+ DCs and significantly decreased by Lrrk2 inhibitor GSK2578215A (1 μM, 24 hours). In conclusion, Lrrk2 is a powerful regulator of Na+/K+ -ATPase expression and activity in dendritic cells.
Collapse
Affiliation(s)
- Zohreh Hosseinzadeh
- Department of Cardiology, Vascular Medicine and Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
- Experimental Retinal Prosthetics Group, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Yogesh Singh
- Department of Cardiology, Vascular Medicine and Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
| | - Derya R. Shimshek
- Department of Neuroscience, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Herman van der Putten
- Department of Neuroscience, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
- National Contest for Life (NCL) Foundation, 203555 Hamburg, Germany
| | - Carsten A. Wagner
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Florian Lang
- Department of Cardiology, Vascular Medicine and Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
| |
Collapse
|
21
|
Abstract
The power of Drosophila genetics has attracted attention in tackling important biomedical challenges such as the understanding and prevention of neurodegenerative diseases. Parkinson's disease (PD) is the most common neurodegenerative movement disorder which results from the relentless degeneration of midbrain dopaminergic neurons. Over the past two decades tremendous advances have been made in identifying genes responsible for inherited forms of PD. The ease of genetic manipulation in Drosophila has spurred the development of numerous models of PD, including expression of human genes carrying pathogenic mutations or the targeted mutation of conserved orthologs. The genetic and cellular analysis of these models is beginning to reveal fundamental insights into the pathogenic mechanisms. Numerous pathways and processes are disrupted in these models but some common themes are emerging. These often implicate aberrant synaptic function, protein aggregation, autophagy, oxidative stress, and mitochondrial dysfunction. Moreover, an impressive list of small molecule compounds have been identified as effective in reversing pathogenic phenotypes, paving the way to explore these for therapeutic interventions.
Collapse
Affiliation(s)
- V L Hewitt
- Medical Research Council Mitochondrial Biology Unit, Cambridge, United Kingdom
| | - A J Whitworth
- Medical Research Council Mitochondrial Biology Unit, Cambridge, United Kingdom.
| |
Collapse
|
22
|
Han L, Ni Y, Cao M, Zhu L, Dai A, Xu Z, Liu X, Chen R, Ning X, Ke K. A New Role Discovered for IGTP: The Protective Effect of IGTP in ICH-Induced Neuronal Apoptosis. Cell Mol Neurobiol 2016; 36:713-24. [PMID: 26242173 PMCID: PMC11482458 DOI: 10.1007/s10571-015-0251-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/29/2015] [Indexed: 10/23/2022]
Abstract
Interferon gamma-induced GTPase (IGTP), which is also named Irgm3, has been widely described in regulating host resistance against intracellular pathogens. Previous researches have demonstrated that IGTP exerts beneficial function during coxsackievirus B3 (CVB3) infection. However, little information is available regarding the role of IGTP in central nervous system. Here, our study revealed that IGTP may have an essential role during ICH-induced neuronal apoptosis. We found the expression level of IGTP adjacent to hematoma was strongly increased after ICH, accompanied with the up-regulation of proliferating cell nuclear antigen (PCNA), active-caspase-3, p-GSK-3β, and Bax. IGTP was also observed to be co-localized with PCNA in astrocytes and active-caspase-3 in neurons, indicating its association with astrocyte proliferation and neuronal apoptosis after ICH. Finally, in vitro study, knocking down IGTP with IGTP-specific siRNA promoted active-caspase-3, p-GSK-3β, and Bax expression, and led to more severe neuronal apoptosis after ICH. All these results above suggested that IGTP might play a critical role in protecting neurons from apoptosis after ICH.
Collapse
Affiliation(s)
- Lijian Han
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yaohui Ni
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Maohong Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Liang Zhu
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Aihua Dai
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Zhiwei Xu
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Xiaorong Liu
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Rongrong Chen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Xiaojin Ning
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Kaifu Ke
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
23
|
Peiris TH, Ramirez D, Barghouth PG, Oviedo NJ. The Akt signaling pathway is required for tissue maintenance and regeneration in planarians. BMC DEVELOPMENTAL BIOLOGY 2016; 16:7. [PMID: 27068018 PMCID: PMC4827215 DOI: 10.1186/s12861-016-0107-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 03/24/2016] [Indexed: 12/20/2022]
Abstract
Background Akt (PKB) is a serine threonine protein kinase downstream of the phosphoinositide 3-kinase (PI3K) pathway. In mammals, Akt is ubiquitously expressed and is associated with regulation of cellular proliferation, metabolism, cell growth and cell death. Akt has been widely studied for its central role in physiology and disease, in particular cancer where it has become an attractive pharmacological target. However, the mechanisms by which Akt signaling regulates stem cell behavior in the complexity of the whole body are poorly understood. Planarians are flatworms with large populations of stem cells capable of dividing to support adult tissue renewal and regeneration. The planarian ortholog Smed-Akt is molecularly conserved providing unique opportunities to analyze the function of Akt during cellular turnover and repair of adult tissues. Results Our findings abrogating Smed-Akt with RNA-interference in the planarian Schmidtea mediterranea led to a gradual decrease in stem cell (neoblasts) numbers. The reduced neoblast numbers largely affected the maintenance of adult tissues including the nervous and excretory systems and ciliated structures in the ventral epithelia, which impaired planarian locomotion. Downregulation of Smed-Akt function also resulted in an increase of cell death throughout the animal. However, in response to amputation, levels of cell death were decreased and failed to localize near the injury site. Interestingly, the neoblast mitotic response was increased around the amputation area but the regenerative blastema failed to form. Conclusions We demonstrate Akt signaling is essential for organismal physiology and in late stages of the Akt phenotype the reduction in neoblast numbers may impair regeneration in planarians. Functional disruption of Smed-Akt alters the balance between cell proliferation and cell death leading to systemic impairment of adult tissue renewal. Our results also reveal novel roles for Akt signaling during regeneration, specifically for the timely localization of cell death near the injury site. Thus, Akt signaling regulates neoblast biology and mediates in the distribution of injury-mediated cell death during tissue repair in planarians. Electronic supplementary material The online version of this article (doi:10.1186/s12861-016-0107-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- T Harshani Peiris
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, 5200 North Lake Road, Merced, CA, 95343, USA.,Quantitative and Systems Biology Graduate Program, University of California, Merced, CA, 95343, USA
| | - Daniel Ramirez
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, 5200 North Lake Road, Merced, CA, 95343, USA.,Quantitative and Systems Biology Graduate Program, University of California, Merced, CA, 95343, USA
| | - Paul G Barghouth
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, 5200 North Lake Road, Merced, CA, 95343, USA.,Quantitative and Systems Biology Graduate Program, University of California, Merced, CA, 95343, USA
| | - Néstor J Oviedo
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, 5200 North Lake Road, Merced, CA, 95343, USA. .,Quantitative and Systems Biology Graduate Program, University of California, Merced, CA, 95343, USA. .,Health Sciences Research Institute, University of California, Merced, CA, 95343, USA.
| |
Collapse
|
24
|
Lin CH, Lin HI, Chen ML, Lai TT, Cao LP, Farrer MJ, Wu RM, Chien CT. Lovastatin protects neurite degeneration inLRRK2-G2019Sparkinsonism through activating the Akt/Nrf pathway and inhibiting GSK3β activity. Hum Mol Genet 2016; 25:1965-1978. [DOI: 10.1093/hmg/ddw068] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 02/22/2016] [Indexed: 11/13/2022] Open
|
25
|
Abstract
Disrupted brain iron homeostasis is a common feature of neurodegenerative disease. To begin to understand how neuronal iron handling might be involved, we focused on dopaminergic neurons and asked how inactivation of transport proteins affected iron homeostasis in vivo in mice. Loss of the cellular iron exporter, ferroportin, had no apparent consequences. However, loss of transferrin receptor 1, involved in iron uptake, caused neuronal iron deficiency, age-progressive degeneration of a subset of dopaminergic neurons, and motor deficits. There was gradual depletion of dopaminergic projections in the striatum followed by death of dopaminergic neurons in the substantia nigra. Damaged mitochondria accumulated, and gene expression signatures indicated attempted axonal regeneration, a metabolic switch to glycolysis, oxidative stress, and the unfolded protein response. We demonstrate that loss of transferrin receptor 1, but not loss of ferroportin, can cause neurodegeneration in a subset of dopaminergic neurons in mice.
Collapse
|
26
|
Langston RG, Rudenko IN, Cookson MR. The function of orthologues of the human Parkinson's disease gene LRRK2 across species: implications for disease modelling in preclinical research. Biochem J 2016; 473:221-32. [PMID: 26811536 PMCID: PMC5165698 DOI: 10.1042/bj20150985] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In the period since LRRK2 (leucine-rich repeat kinase 2) was identified as a causal gene for late-onset autosomal dominant parkinsonism, a great deal of work has been aimed at understanding whether the LRRK2 protein might be a druggable target for Parkinson's disease (PD). As part of this effort, animal models have been developed to explore both the normal and the pathophysiological roles of LRRK2. However, LRRK2 is part of a wider family of proteins whose functions in different organisms remain poorly understood. In this review, we compare the information available on biochemical properties of LRRK2 homologues and orthologues from different species from invertebrates (e.g. Caenorhabditis elegans and Drosophila melanogaster) to mammals. We particularly discuss the mammalian LRRK2 homologue, LRRK1, and those species where there is only a single LRRK homologue, discussing examples where each of the LRRK family of proteins has distinct properties as well as those cases where there appear to be functional redundancy. We conclude that uncovering the function of LRRK2 orthologues will help to elucidate the key properties of human LRRK2 as well as to improve understanding of the suitability of different animal models for investigation of LRRK2-related PD.
Collapse
Affiliation(s)
- Rebekah G. Langston
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Bethesda, MD, 20892
| | - Iakov N. Rudenko
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Bethesda, MD, 20892
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, NIA, NIH, Bethesda, MD, 20892
| |
Collapse
|
27
|
Roles of NlAKTIP in the Growth and Eclosion of the Rice Brown Planthopper, Nilaparvata lugens Stål, as Revealed by RNA Interference. Int J Mol Sci 2015; 16:22888-903. [PMID: 26402675 PMCID: PMC4613341 DOI: 10.3390/ijms160922888] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 09/14/2015] [Accepted: 09/16/2015] [Indexed: 01/15/2023] Open
Abstract
AKT-interacting protein (AKTIP) interacts with serine/threonine protein kinase B (PKB)/AKT. AKTIP modulates AKT’s activity by enhancing the phosphorylation of the regulatory site and plays a crucial role in multiple biological processes. In this study, the full length cDNA of NlAKTIP, a novel AKTIP gene in the brown planthopper (BPH) Nilaparvata lugens, was cloned. The reverse transcription quantitive PCR (RT-qPCR) results showed that the NlAKTIP gene was strongly expressed in gravid female adults, but was relatively weakly expressed in nymphs and male adult BPH. In female BPH, treatment with dsAKTIP resulted in the efficient silencing of NlAKTIP, leading to a significant reduction of mRNA levels, about 50% of those of the untreated control group at day 7 of the study. BPH fed with dsAKTIP had reduced growth with lower body weights and smaller sizes, and the body weight of BPH treated with dsAKTIP at day 7 decreased to about 30% of that of the untreated control. Treatment of dsAKTIP significantly delayed the eclosion for over 7 days relative to the control group and restricted ovarian development to Grade I (transparent stage), whereas the controls developed to Grade IV (matured stage). These results indicated that NlAKTIP is crucial to the growth and development of female BPH. This study provided a valuable clue of a potential target NlAKTIP for inhibiting the BPH, and also provided a new point of view on the interaction between BPH and resistant rice.
Collapse
|