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
|
Zhao P, Tian R, Song D, Zhu Q, Ding X, Zhang J, Cao B, Zhang M, Xu Y, Fang J, Tan J, Yi C, Xia H, Liu W, Zou W, Sun Q. Rab GTPases are evolutionarily conserved signals mediating selective autophagy. J Cell Biol 2025; 224:e202410150. [PMID: 40197538 PMCID: PMC11977514 DOI: 10.1083/jcb.202410150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/31/2024] [Accepted: 01/21/2025] [Indexed: 04/10/2025] Open
Abstract
Selective autophagy plays a crucial role in maintaining cellular homeostasis by specifically targeting unwanted cargo labeled with "autophagy cues" signals for autophagic degradation. In this study, we identify Rab GTPases as a class of such autophagy cues signals involved in selective autophagy. Through biochemical and imaging screens, we reveal that human Rab GTPases are common autophagy substrates. Importantly, we confirm the conservation of Rab GTPase autophagic degradation in different model organisms. Rab GTPases translocate to damaged mitochondria, lipid droplets, and invading Salmonella-containing vacuoles (SCVs) to serve as degradation signals. Furthermore, they facilitate mitophagy, lipophagy, and xenophagy, respectively, by recruiting receptors. This interplay between Rab GTPases and receptors may ensure the de novo synthesis of isolation membranes around Rab-GTPase-labeled cargo, thereby mediating selective autophagy. These processes are further influenced by upstream regulators such as LRRK2, GDIs, and RabGGTase. In conclusion, this study unveils a conserved mechanism involving Rab GTPases as autophagy cues signals and proposes a model for the spatiotemporal control of selective autophagy.
Collapse
Affiliation(s)
- Pengwei Zhao
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Rui Tian
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Dandan Song
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Qi Zhu
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Xianming Ding
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Jianqin Zhang
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Beibei Cao
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Mengyuan Zhang
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Yilu Xu
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Jie Fang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Jieqiong Tan
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Cong Yi
- Department of Biochemistry, and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongguang Xia
- Department of Biochemistry, and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Liu
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
- Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Zou
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Qiming Sun
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
- Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Hangzhou, China
| |
Collapse
|
3
|
Xu L, Wang A, Guan H. microRNA-106b-5p and Rab10: Potential Markers of Acute Myeloid Leukemia. Cancer Biother Radiopharm 2024; 39:492-501. [PMID: 38949985 DOI: 10.1089/cbr.2023.0191] [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] [Indexed: 07/03/2024] Open
Abstract
This study focuses on acute myeloid leukemia (AML), a condition with a 5-year survival rate below 30% despite various treatment options. Recent strides in targeted therapies have shown promise, leading to better outcomes with minimal toxicity. These advances underscore the importance of discovering new diagnostic and prognostic targets for AML. In this context, the authors investigated the expression of microRNA-106b-5p (miR-106b-5p), Rab10 mRNA, and Rab10 proteins in peripheral blood and bone marrow (BM) samples from both healthy individuals and AML patients at different stages of the disease (initial diagnosis, recurrence, and complete remission). This examination aimed to identify potential biomarkers for AML diagnosis, treatment, and prognosis. From June 2021 to December 2022, they collected 100 BM and peripheral blood samples. The relative expression of miR-106b-5p and Rab10 mRNA in the BM of AML patients was measured using Real-time polymerase chain reaction (qRT-PCR), while the relative expression of Rab10 protein in serum was determined using the ELISA method. The chromosomal karyotype of initially diagnosed patients was analyzed using the R tape. The qRT-PCR results revealed that the expression of miR-106b-5p and Rab10 mRNA were significantly higher in patients at initial diagnosis and recurrence compared with healthy individuals and those in complete remission (p < 0.001). They observed a significant reduction in the expression of miR-106b-5p, Rab10 mRNA, and Rab10 protein in the BM and peripheral blood of patients during complete remission (p < 0.05), as demonstrated by dynamic monitoring of five patients in the initial group. Furthermore, they found a close association between the expression of miR-106b-5p and the number of white blood cells at the initial diagnosis in AML patients (p < 0.05). Spearman correlation analysis revealed a positive correlation among miR-106b-5p, Rab10 mRNA, and Rab10 proteins (p < 0.05). The diagnostic potential of miR-106b-5p and Rab10 proteins was underscored by Receiver Operating Characteristic (ROC) curve analysis, which demonstrated their high accuracy in AML diagnosis (AUC: 0.944 and 0.853, respectively; p < 0.0001). Additionally, Kaplan-Meier survival analysis suggested that lower expression of these markers was associated with better prognoses (p < 0.05). In summary, their findings propose miR-106b-5p and Rab10 proteins as promising biomarkers for AML, offering insights for diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Lingyue Xu
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ailing Wang
- Shibei District People's Hospital, Qingdao, China
| | - Hongzai Guan
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
4
|
Zhang T, Linghu KG, Tan J, Wang M, Chen D, Shen Y, Wu J, Shi M, Zhou Y, Tang L, Liu L, Qin ZH, Guo B. TIGAR exacerbates obesity by triggering LRRK2-mediated defects in macroautophagy and chaperone-mediated autophagy in adipocytes. Autophagy 2024; 20:1741-1761. [PMID: 38686804 PMCID: PMC11262232 DOI: 10.1080/15548627.2024.2338576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 03/22/2024] [Accepted: 03/31/2024] [Indexed: 05/02/2024] Open
Abstract
Obesity is one of the most common metabolic diseases around the world, which is distinguished by the abnormal buildup of triglycerides within adipose cells. Recent research has revealed that autophagy regulates lipid mobilization to maintain energy balance. TIGAR (Trp53 induced glycolysis regulatory phosphatase) has been identified as a glycolysis inhibitor, whether it plays a role in the metabolism of lipids is unknown. Here, we found that TIGAR transgenic (TIGAR+/+) mice exhibited increased fat mass and trended to obesity phenotype. Non-target metabolomics showed that TIGAR caused the dysregulation of the metabolism profile. The quantitative transcriptome sequencing identified an increased levels of LRRK2 and RAB7B in the adipose tissue of TIGAR+/+ mice. It was confirmed in vitro that TIGAR overexpression increased the levels of LRRK2 by inhibiting polyubiquitination degradation, thereby suppressing macroautophagy and chaperone-mediated autophagy (CMA) while increasing lipid accumulation which were reversed by the LRRK2 inhibitor DNL201. Furthermore, TIGAR drove LRRK2 to interact with RAB7B for suppressing lysosomal degradation of lipid droplets, while the increased lipid droplets in adipocytes were blocked by the RAB7B inhibitor ML282. Additionally, fat-specific TIGAR knockdown of TIGAR+/+ mice alleviated the symptoms of obesity, and adipose tissues-targeting superiority DNL201 nano-emulsion counteracted the obesity phenotype in TIGAR+/+ mice. In summary, the current results indicated that TIGAR performed a vital function in the lipid metabolism through LRRK2-mediated negative regulation of macroautophagy and CMA in adipocyte. The findings suggest that TIGAR has the potential to serve as a viable therapeutic target for treating obesity and its associated metabolic dysfunction.
Collapse
Affiliation(s)
- Tian Zhang
- Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Ke-Gang Linghu
- Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jia Tan
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Mingming Wang
- Department of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Diao Chen
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yan Shen
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Junchao Wu
- Department of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Mingjun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yuxia Zhou
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Lei Tang
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, Guizhou, China
| | - Lirong Liu
- Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
- Institute of Health Technology, Global Institute of Software Technology, Suzhou, Jiangsu, China
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
5
|
Li Y, Wen Y, Li Y, Tan X, Gao S, Fan P, Tian W, Wong CC, Chen Y. Rab10-CAV1 mediated intraluminal vesicle transport to migrasomes. Proc Natl Acad Sci U S A 2024; 121:e2319267121. [PMID: 39008679 PMCID: PMC11287133 DOI: 10.1073/pnas.2319267121] [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: 11/08/2023] [Accepted: 06/12/2024] [Indexed: 07/17/2024] Open
Abstract
Migrasomes, vesicular organelles generated on the retraction fibers of migrating cells, play a crucial role in migracytosis, mediating intercellular communication. The cargoes determine the functional specificity of migrasomes. Migrasomes harbor numerous intraluminal vesicles, a pivotal component of their cargoes. The mechanism underlying the transportation of these intraluminal vesicles to the migrasomes remains enigmatic. In this study, we identified that Rab10 and Caveolin-1 (CAV1) mark the intraluminal vesicles in migrasomes. Transport of Rab10-CAV1 vesicles to migrasomes required the motor protein Myosin Va and adaptor proteins RILPL2. Notably, the phosphorylation of Rab10 by the kinase LRRK2 regulated this process. Moreover, CSF-1 can be transported to migrasomes through this mechanism, subsequently fostering monocyte-macrophage differentiation in skin wound healing, which served as a proof of the physiological importance of this transporting mechanism.
Collapse
Affiliation(s)
- Yong Li
- Peking‐Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing100084, China
- Center for Precision Medicine Multi-Omics Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing100191, China
| | - Yiling Wen
- Center for Precision Medicine Multi-Omics Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing100191, China
| | - Ying Li
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Xinyi Tan
- The Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing100101, China
| | - Shuaixin Gao
- Department of Human Sciences & James Comprehensive Cancer Center, The Ohio State University, Columbus, OH43210
| | - Peiyao Fan
- Center for Precision Medicine Multi-Omics Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing100191, China
| | - Wenmin Tian
- Center for Precision Medicine Multi-Omics Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing100191, China
| | - Catherine C.L. Wong
- Peking‐Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing100084, China
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing100730, China
| | - Yang Chen
- Center for Precision Medicine Multi-Omics Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing100191, China
| |
Collapse
|
6
|
Davis GH, Zaya A, Pearce MMP. Impairment of the Glial Phagolysosomal System Drives Prion-Like Propagation in a Drosophila Model of Huntington's Disease. J Neurosci 2024; 44:e1256232024. [PMID: 38589228 PMCID: PMC11097281 DOI: 10.1523/jneurosci.1256-23.2024] [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: 07/06/2023] [Revised: 01/31/2024] [Accepted: 02/26/2024] [Indexed: 04/10/2024] Open
Abstract
Protein misfolding, aggregation, and spread through the brain are primary drivers of neurodegenerative disease pathogenesis. Phagocytic glia are responsible for regulating the load of pathological proteins in the brain, but emerging evidence suggests that glia may also act as vectors for aggregate spread. Accumulation of protein aggregates could compromise the ability of glia to eliminate toxic materials from the brain by disrupting efficient degradation in the phagolysosomal system. A better understanding of phagocytic glial cell deficiencies in the disease state could help to identify novel therapeutic targets for multiple neurological disorders. Here, we report that mutant huntingtin (mHTT) aggregates impair glial responsiveness to injury and capacity to degrade neuronal debris in male and female adult Drosophila expressing the gene that causes Huntington's disease (HD). mHTT aggregate formation in neurons impairs engulfment and clearance of injured axons and causes accumulation of phagolysosomes in glia. Neuronal mHTT expression induces upregulation of key innate immunity and phagocytic genes, some of which were found to regulate mHTT aggregate burden in the brain. A forward genetic screen revealed Rab10 as a novel component of Draper-dependent phagocytosis that regulates mHTT aggregate transmission from neurons to glia. These data suggest that glial phagocytic defects enable engulfed mHTT aggregates to evade lysosomal degradation and acquire prion-like characteristics. Together, our findings uncover new mechanisms that enhance our understanding of the beneficial and harmful effects of phagocytic glia in HD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Graham H Davis
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, New Jersey 08028
- Department of Biology, Saint Joseph's University, Philadelphia, Pennsylvania 19131
- Department of Biological Sciences, University of the Sciences, Philadelphia, Pennsylvania 19104
| | - Aprem Zaya
- Department of Biological Sciences, University of the Sciences, Philadelphia, Pennsylvania 19104
| | - Margaret M Panning Pearce
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, New Jersey 08028
- Department of Biology, Saint Joseph's University, Philadelphia, Pennsylvania 19131
- Department of Biological Sciences, University of the Sciences, Philadelphia, Pennsylvania 19104
| |
Collapse
|
7
|
Davis GH, Zaya A, Pearce MMP. Impairment of the glial phagolysosomal system drives prion-like propagation in a Drosophila model of Huntington's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.04.560952. [PMID: 38370619 PMCID: PMC10871239 DOI: 10.1101/2023.10.04.560952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Protein misfolding, aggregation, and spread through the brain are primary drivers of neurodegenerative diseases pathogenesis. Phagocytic glia are responsible for regulating the load of pathogenic protein aggregates in the brain, but emerging evidence suggests that glia may also act as vectors for aggregate spread. Accumulation of protein aggregates could compromise the ability of glia to eliminate toxic materials from the brain by disrupting efficient degradation in the phagolysosomal system. A better understanding of phagocytic glial cell deficiencies in the disease state could help to identify novel therapeutic targets for multiple neurological disorders. Here, we report that mutant huntingtin (mHTT) aggregates impair glial responsiveness to injury and capacity to degrade neuronal debris in male and female adult Drosophila expressing the gene that causes Huntington's disease (HD). mHTT aggregate formation in neurons impairs engulfment and clearance of injured axons and causes accumulation of phagolysosomes in glia. Neuronal mHTT expression induces upregulation of key innate immunity and phagocytic genes, some of which were found to regulate mHTT aggregate burden in the brain. Finally, a forward genetic screen revealed Rab10 as a novel component of Draper-dependent phagocytosis that regulates mHTT aggregate transmission from neurons to glia. These data suggest that glial phagocytic defects enable engulfed mHTT aggregates to evade lysosomal degradation and acquire prion-like characteristics. Together, our findings reveal new mechanisms that enhance our understanding of the beneficial and potentially harmful effects of phagocytic glia in HD and potentially other neurodegenerative diseases.
Collapse
Affiliation(s)
- Graham H. Davis
- Rowan University, Department of Biological and Biomedical Sciences, Glassboro, NJ 08028
- Saint Joseph’s University, Department of Biology, Philadelphia, PA 19131
- University of the Sciences, Department of Biological Sciences, Philadelphia, PA 19104
| | - Aprem Zaya
- University of the Sciences, Department of Biological Sciences, Philadelphia, PA 19104
| | - Margaret M. Panning Pearce
- Rowan University, Department of Biological and Biomedical Sciences, Glassboro, NJ 08028
- Saint Joseph’s University, Department of Biology, Philadelphia, PA 19131
- University of the Sciences, Department of Biological Sciences, Philadelphia, PA 19104
| |
Collapse
|
8
|
Baidya AT, Deshwal S, Das B, Mathew AT, Devi B, Sandhir R, Kumar R. Catalyzing a Cure: Discovery and development of LRRK2 inhibitors for the treatment of Parkinson's disease. Bioorg Chem 2024; 143:106972. [PMID: 37995640 DOI: 10.1016/j.bioorg.2023.106972] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023]
Abstract
Parkinson's disease (PD) is an age-related second most common progressive neurodegenerative disorder that affects millions of people worldwide. Despite decades of research, no effective disease modifying therapeutics have reached clinics for treatment/management of PD. Leucine-rich repeat kinase 2 (LRRK2) which controls membrane trafficking and lysosomal function and its variant LRRK2-G2019S are involved in the development of both familial and sporadic PD. LRRK2, is therefore considered as a legitimate target for the development of therapeutics against PD. During the last decade, efforts have been made to develop effective, safe and selective LRRK2 inhibitors and also our understanding about LRRK2 has progressed. However, there is an urge to learn from the previously designed and reported LRRK2 inhibitors in order to effectively approach designing of new LRRK2 inhibitors. In this review, we have aimed to cover the pre-clinical studies undertaken to develop small molecule LRRK2 inhibitors by screening the patents and other available literature in the last decade. We have highlighted LRRK2 as targets in the progress of PD and subsequently covered detailed design, synthesis and development of diverse scaffolds as LRRK2 inhibitors. Moreover, LRRK2 inhibitors under clinical development has also been discussed. LRRK2 inhibitors seem to be potential targets for future therapeutic interventions in the treatment and management of PD and this review can act as a cynosure for guiding discovery, design, and development of selective and non-toxic LRRK2 inhibitors. Although, there might be challenges in developing effective LRRK2 inhibitors, the opportunity to successfully develop novel therapeutics targeting LRRK2 against PD has never been greater.
Collapse
Affiliation(s)
- Anurag Tk Baidya
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005, UP, India
| | - Sonam Deshwal
- Department of Biochemistry, Panjab University, Chandigarh 160014, India
| | - Bhanuranjan Das
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005, UP, India
| | - Alen T Mathew
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005, UP, India
| | - Bharti Devi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005, UP, India
| | - Rajat Sandhir
- Department of Biochemistry, Panjab University, Chandigarh 160014, India
| | - Rajnish Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005, UP, India.
| |
Collapse
|
9
|
Zhou ZD, Yi LX, Wang DQ, Lim TM, Tan EK. Role of dopamine in the pathophysiology of Parkinson's disease. Transl Neurodegener 2023; 12:44. [PMID: 37718439 PMCID: PMC10506345 DOI: 10.1186/s40035-023-00378-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/08/2023] [Indexed: 09/19/2023] Open
Abstract
A pathological feature of Parkinson's disease (PD) is the progressive loss of dopaminergic neurons and decreased dopamine (DA) content in the substantia nigra pars compacta in PD brains. DA is the neurotransmitter of dopaminergic neurons. Accumulating evidence suggests that DA interacts with environmental and genetic factors to contribute to PD pathophysiology. Disturbances of DA synthesis, storage, transportation and metabolism have been shown to promote neurodegeneration of dopaminergic neurons in various PD models. DA is unstable and can undergo oxidation and metabolism to produce multiple reactive and toxic by-products, including reactive oxygen species, DA quinones, and 3,4-dihydroxyphenylacetaldehyde. Here we summarize and highlight recent discoveries on DA-linked pathophysiologic pathways, and discuss the potential protective and therapeutic strategies to mitigate the complications associated with DA.
Collapse
Affiliation(s)
- Zhi Dong Zhou
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore.
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore, 169857, Singapore.
| | - Ling Xiao Yi
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
| | - Dennis Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Tit Meng Lim
- Department of Biological Science, National University of Singapore, Singapore, 119077, Singapore
| | - Eng King Tan
- National Neuroscience Institute of Singapore, 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 Behavioral Disorders, Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore, 169857, Singapore.
| |
Collapse
|
10
|
Pirone A, Ciregia F, Lazzarini G, Miragliotta V, Ronci M, Zuccarini M, Zallocco L, Beghelli D, Mazzoni MR, Lucacchini A, Giusti L. Proteomic Profiling Reveals Specific Molecular Hallmarks of the Pig Claustrum. Mol Neurobiol 2023; 60:4336-4358. [PMID: 37095366 PMCID: PMC10293365 DOI: 10.1007/s12035-023-03347-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 04/13/2023] [Indexed: 04/26/2023]
Abstract
The present study, employing a comparative proteomic approach, analyzes the protein profile of pig claustrum (CLA), putamen (PU), and insula (IN). Pig brain is an interesting model whose key translational features are its similarities with cortical and subcortical structures of human brain. A greater difference in protein spot expression was observed in CLA vs PU as compared to CLA vs IN. The deregulated proteins identified in CLA resulted to be deeply implicated in neurodegenerative (i.e., sirtuin 2, protein disulfide-isomerase 3, transketolase) and psychiatric (i.e., copine 3 and myelin basic protein) disorders in humans. Metascape analysis of differentially expressed proteins in CLA vs PU comparison suggested activation of the α-synuclein pathway and L1 recycling pathway corroborating the involvement of these anatomical structures in neurodegenerative diseases. The expression of calcium/calmodulin-dependent protein kinase and dihydropyrimidinase like 2, which are linked to these pathways, was validated using western blot analysis. Moreover, the protein data set of CLA vs PU comparison was analyzed by Ingenuity Pathways Analysis to obtain a prediction of most significant canonical pathways, upstream regulators, human diseases, and biological functions. Interestingly, inhibition of presenilin 1 (PSEN1) upstream regulator and activation of endocannabinoid neuronal synapse pathway were observed. In conclusion, this is the first study presenting an extensive proteomic analysis of pig CLA in comparison with adjacent areas, IN and PUT. These results reinforce the common origin of CLA and IN and suggest an interesting involvement of CLA in endocannabinoid circuitry, neurodegenerative, and psychiatric disorders in humans.
Collapse
Affiliation(s)
- Andrea Pirone
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy.
| | - Federica Ciregia
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giulia Lazzarini
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | | | - Maurizio Ronci
- Department of Medical, Oral and Biotechnological Sciences, University G. D'Annunzio of Chieti-Pescara, Chieti, Italy
- Interuniversitary Consortium for Engineering and Medicine, COIIM, Campobasso, Italy
| | - Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University G. D'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Lorenzo Zallocco
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Daniela Beghelli
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | | | - Antonio Lucacchini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Laura Giusti
- School of Pharmacy, University of Camerino, Camerino, Italy
| |
Collapse
|
11
|
Hadisurya M, Li L, Kuwaranancharoen K, Wu X, Lee ZC, Alcalay RN, Padmanabhan S, Tao WA, Iliuk A. Quantitative proteomics and phosphoproteomics of urinary extracellular vesicles define putative diagnostic biosignatures for Parkinson's disease. COMMUNICATIONS MEDICINE 2023; 3:64. [PMID: 37165152 PMCID: PMC10172329 DOI: 10.1038/s43856-023-00294-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/27/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene have been recognized as genetic risk factors for Parkinson's disease (PD). However, compared to cancer, fewer genetic mutations contribute to the cause of PD, propelling the search for protein biomarkers for early detection of the disease. METHODS Utilizing 138 urine samples from four groups, healthy individuals (control), healthy individuals with G2019S mutation in the LRRK2 gene (non-manifesting carrier/NMC), PD individuals without G2019S mutation (idiopathic PD/iPD), and PD individuals with G2019S mutation (LRRK2 PD), we applied a proteomics strategy to determine potential diagnostic biomarkers for PD from urinary extracellular vesicles (EVs). RESULTS After efficient isolation of urinary EVs through chemical affinity followed by mass spectrometric analyses of EV peptides and enriched phosphopeptides, we identify and quantify 4476 unique proteins and 2680 unique phosphoproteins. We detect multiple proteins and phosphoproteins elevated in PD EVs that are known to be involved in important PD pathways, in particular the autophagy pathway, as well as neuronal cell death, neuroinflammation, and formation of amyloid fibrils. We establish a panel of proteins and phosphoproteins as novel candidates for disease biomarkers and substantiate the biomarkers using machine learning, ROC, clinical correlation, and in-depth network analysis. Several putative disease biomarkers are further partially validated in patients with PD using parallel reaction monitoring (PRM) and immunoassay for targeted quantitation. CONCLUSIONS These findings demonstrate a general strategy of utilizing biofluid EV proteome/phosphoproteome as an outstanding and non-invasive source for a wide range of disease exploration.
Collapse
Affiliation(s)
- Marco Hadisurya
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Li Li
- Tymora Analytical Operations, West Lafayette, IN, 47906, USA
| | | | - Xiaofeng Wu
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Zheng-Chi Lee
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47907, USA
- West Lafayette Junior/Senior High School, West Lafayette, IN, 47906, USA
| | - Roy N Alcalay
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Shalini Padmanabhan
- The Michael J. Fox Foundation for Parkinson's Research, New York City, NY, 10163, USA
| | - W Andy Tao
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47907, USA.
- Tymora Analytical Operations, West Lafayette, IN, 47906, USA.
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA.
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA.
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA.
| | - Anton Iliuk
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47907, USA.
- Tymora Analytical Operations, West Lafayette, IN, 47906, USA.
| |
Collapse
|
12
|
Identification of LRRK2 gene related to sarcopenia and neuroticism using weighted gene co-expression network analysis. J Affect Disord 2023; 325:675-681. [PMID: 36690080 DOI: 10.1016/j.jad.2023.01.042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 12/26/2022] [Accepted: 01/08/2023] [Indexed: 01/22/2023]
Abstract
BACKGROUND Sarcopenia is reported to be associated with neuroticism, but the mechanisms are not fully understood. Thus, it's of vital importance to elucidate the molecular mechanism of sarcopenia and neuroticism and to explore the potential molecular target of medical therapies for sarcopenia and neuroticism. METHODS The expression datasets (sarcopenia: GSE111006 and neuroticism: GSE60491) were downloaded from the Gene Expression Omnibus. Weighted gene co-expression network analysis (WGCNA) was used to build the gene co-expression network, screen important modules, and filter the hub genes. Genes with significance over 0.2 and a module membership over 0.8 were hub genes. The overlapped hub genes between sarcopenia and neuroticism were defined as key genes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed for the genes in modules with clinical interest. RESULTS In this study, we identified 28 gene modules for sarcopenia and 7 for neuroticism by WGCNA. The key modules of sarcopenia and neuroticism were the tan and turquoise modules, respectively. Hub genes of sarcopenia and neuroticism were 20 genes and 107 genes, respectively. The function enrichment found that apoptosis was the common pathway for sarcopenia and neuroticism. Finally, LRRK2 was identified as key genes. LIMITATIONS The sarcopenia dataset contained fewer samples. CONCLUSION Based on WGCNA, our study identified apoptosis pathway and LRRK2 that acted as essential components in the etiology of sarcopenia and neuroticism, which may enhance our fundamental knowledge of the molecular mechanisms underlying the disease.
Collapse
|
13
|
Zhao Y, Vavouraki N, Lovering RC, Escott-Price V, Harvey K, Lewis PA, Manzoni C. Tissue specific LRRK2 interactomes reveal a distinct striatal functional unit. PLoS Comput Biol 2023; 19:e1010847. [PMID: 36716346 PMCID: PMC9910798 DOI: 10.1371/journal.pcbi.1010847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 02/09/2023] [Accepted: 01/03/2023] [Indexed: 02/01/2023] Open
Abstract
Mutations in LRRK2 are the most common genetic cause of Parkinson's disease. Despite substantial research efforts, the physiological and pathological role of this multidomain protein remains poorly defined. In this study, we used a systematic approach to construct the general protein-protein interactome around LRRK2, which was then evaluated taking into consideration the differential expression patterns and the co-expression behaviours of the LRRK2 interactors in 15 different healthy tissue types. The LRRK2 interactors exhibited distinct expression features in the brain as compared to the peripheral tissues analysed. Moreover, a high degree of similarity was found for the LRRK2 interactors in putamen, caudate and nucleus accumbens, thus defining a potential LRRK2 functional cluster within the striatum. The general LRRK2 interactome paired with the expression profiles of its members constitutes a powerful tool to generate tissue-specific LRRK2 interactomes. We exemplified the generation of the tissue-specific LRRK2 interactomes and explored the functions highlighted by the "core LRRK2 interactors" in the striatum in comparison with the cerebellum. Finally, we illustrated how the LRRK2 general interactome reported in this manuscript paired with the expression profiles can be used to trace the relationship between LRRK2 and specific interactors of interest, here focusing on the LRRK2 interactors belonging to the Rab protein family.
Collapse
Affiliation(s)
- Yibo Zhao
- University College London, School of Pharmacy, London, United Kingdom
| | | | - Ruth C. Lovering
- University College London, Institute for Cardiovascular Science, London, United Kingdom
| | - Valentina Escott-Price
- University of Cardiff, School of Medicine, Division of Psychological Medicine and Clinical Neurosciences, Cardiff, United Kingdom
| | - Kirsten Harvey
- University College London, School of Pharmacy, London, United Kingdom
| | - Patrick A. Lewis
- University of Reading, School of Pharmacy, Reading, United Kingdom
- Royal Veterinary College, London, United Kingdom
- UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Claudia Manzoni
- University College London, School of Pharmacy, London, United Kingdom
| |
Collapse
|
14
|
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
|
15
|
Zhang ZW, Tu H, Jiang M, Vanan S, Chia SY, Jang SE, Saw WT, Ong ZW, Ma DR, Zhou ZD, Xu J, Guo KH, Yu WP, Ling SC, Margolin RA, Chain DG, Zeng L, Tan EK. The APP intracellular domain promotes LRRK2 expression to enable feed-forward neurodegenerative mechanisms in Parkinson's disease. Sci Signal 2022; 15:eabk3411. [PMID: 35998231 DOI: 10.1126/scisignal.abk3411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Gain-of-function mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are common in familial forms of Parkinson's disease (PD), which is characterized by progressive neurodegeneration that impairs motor and cognitive function. We previously demonstrated that LRRK2-mediated phosphorylation of β-amyloid precursor protein (APP) triggers the production and nuclear translocation of the APP intracellular domain (AICD). Here, we connected LRRK2 to AICD in a feed-forward cycle that enhanced LRRK2-mediated neurotoxicity. In cooperation with the transcription factor FOXO3a, AICD promoted LRRK2 expression, thus increasing the abundance of LRRK2 that promotes AICD activation. APP deficiency in LRRK2G2019S mice suppressed LRRK2 expression, LRRK2-mediated mitochondrial dysfunction, α-synuclein accumulation, and tyrosine hydroxylase (TH) loss in the brain, phenotypes associated with toxicity and loss of dopaminergic neurons in PD. Conversely, AICD overexpression increased LRRK2 expression and LRRK2-mediated neurotoxicity in LRRK2G2019S mice. In LRRK2G2019S mice or cultured dopaminergic neurons from LRRK2G2019S patients, treatment with itanapraced reduced LRRK2 expression and was neuroprotective. Itanapraced showed similar effects in a neurotoxin-induced PD mouse model, suggesting that inhibiting the AICD may also have therapeutic benefits in idiopathic PD. Our findings reveal a therapeutically targetable, feed-forward mechanism through which AICD promotes LRRK2-mediated neurotoxicity in PD.
Collapse
Affiliation(s)
- Zhi-Wei Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Haitao Tu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Mei Jiang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore.,Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Sarivin Vanan
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Sook Yoong Chia
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Se-Eun Jang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Wuan-Ting Saw
- Research Department, National Neuroscience Institute, Singapore General Hospital (SGH) Campus, Singapore 169856, Singapore
| | - Zhi-Wei Ong
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Dong-Rui Ma
- Department of Neurology, Singapore General Hospital, Singapore 169609, Singapore
| | - Zhi-Dong Zhou
- Research Department, National Neuroscience Institute, Singapore General Hospital (SGH) Campus, Singapore 169856, Singapore.,Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Jie Xu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Kai-Hua Guo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Wei-Ping Yu
- Animal Gene Editing Laboratory, Biological Resource Center, A*STAR, Singapore 138673, Singapore.,Institute of Molecular and Cell Biology, A*STAR, Singapore 138673, Singapore
| | - Shuo-Chien Ling
- Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | | | | | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore.,Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore.,Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technology University, Novena Campus, Singapore 308232, Singapore
| | - Eng-King Tan
- Research Department, National Neuroscience Institute, Singapore General Hospital (SGH) Campus, Singapore 169856, Singapore.,Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore.,Department of Neurology, National Neuroscience Institute, Singapore 308433, Singapore
| |
Collapse
|
16
|
Thakur G, Kumar V, Lee KW, Won C. Structural Insights and Development of LRRK2 Inhibitors for Parkinson's Disease in the Last Decade. Genes (Basel) 2022; 13:1426. [PMID: 36011337 PMCID: PMC9408223 DOI: 10.3390/genes13081426] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 12/01/2022] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, characterized by the specific loss of dopaminergic neurons in the midbrain. The pathophysiology of PD is likely caused by a variety of environmental and hereditary factors. Many single-gene mutations have been linked to this disease, but a significant number of studies indicate that mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) are a potential therapeutic target for both sporadic and familial forms of PD. Consequently, the identification of potential LRRK2 inhibitors has been the focus of drug discovery. Various investigations have been conducted in academic and industrial organizations to investigate the mechanism of LRRK2 in PD and further develop its inhibitors. This review summarizes the role of LRRK2 in PD and its structural details, especially the kinase domain. Furthermore, we reviewed in vitro and in vivo findings of selected inhibitors reported to date against wild-type and mutant versions of the LRRK2 kinase domain as well as the current trends researchers are employing in the development of LRRK2 inhibitors.
Collapse
Affiliation(s)
- Gunjan Thakur
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Vikas Kumar
- Division of Life Sciences, Department of Bio & Medical Big Data (BK4 Program), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea
| | - Keun Woo Lee
- Division of Life Sciences, Department of Bio & Medical Big Data (BK4 Program), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea
| | - Chungkil Won
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| |
Collapse
|
17
|
Zhou ZD, Saw WT, Ho PGH, Zhang ZW, Zeng L, Chang YY, Sun AXY, Ma DR, Wang HY, Zhou L, Lim KL, Tan EK. The role of tyrosine hydroxylase-dopamine pathway in Parkinson's disease pathogenesis. Cell Mol Life Sci 2022; 79:599. [PMID: 36409355 PMCID: PMC9678997 DOI: 10.1007/s00018-022-04574-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by selective and progressive dopamine (DA) neuron loss in the substantia nigra and other brain regions, with the presence of Lewy body formation. Most PD cases are sporadic, whereas monogenic forms of PD have been linked to multiple genes, including Leucine kinase repeat 2 (LRRK2) and PTEN-induced kinase 1 (PINK1), two protein kinase genes involved in multiple signaling pathways. There is increasing evidence to suggest that endogenous DA and DA-dependent neurodegeneration have a pathophysiologic role in sporadic and familial PD. METHODS We generated patient-derived dopaminergic neurons and human midbrain-like organoids (hMLOs), transgenic (TG) mouse and Drosophila models, expressing both mutant and wild-type (WT) LRRK2 and PINK1. Using these models, we examined the effect of LRRK2 and PINK1 on tyrosine hydroxylase (TH)-DA pathway. RESULTS We demonstrated that PD-linked LRRK2 mutations were able to modulate TH-DA pathway, resulting in up-regulation of DA early in the disease which subsequently led to neurodegeneration. The LRRK2-induced DA toxicity and degeneration were abrogated by wild-type (WT) PINK1 (but not PINK1 mutations), and early treatment with a clinical-grade drug, α-methyl-L-tyrosine (α-MT), a TH inhibitor, was able to reverse the pathologies in human neurons and TG Drosophila models. We also identified opposing effects between LRRK2 and PINK1 on TH expression, suggesting that functional balance between these two genes may regulate the TH-DA pathway. CONCLUSIONS Our findings highlight the vital role of the TH-DA pathway in PD pathogenesis. LRRK2 and PINK1 have opposing effects on the TH-DA pathway, and its balance affects DA neuron survival. LRRK2 or PINK1 mutations can disrupt this balance, promoting DA neuron demise. Our findings provide support for potential clinical trials using TH-DA pathway inhibitors in early or prodromic PD.
Collapse
Affiliation(s)
- Zhi Dong Zhou
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioural Disorders, 8 College Road, Singapore, 169857 Singapore
| | - Wuan Ting Saw
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Patrick Ghim Hoe Ho
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Zhi Wei Zhang
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Li Zeng
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Ya Yin Chang
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioural Disorders, 8 College Road, Singapore, 169857 Singapore
| | - Alfred Xu Yang Sun
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioural Disorders, 8 College Road, Singapore, 169857 Singapore
| | - Dong Rui Ma
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Hong Yan Wang
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioural Disorders, 8 College Road, Singapore, 169857 Singapore
| | - Lei Zhou
- Ocular Proteomics Laboratory, Singapore Eye Research Institute, Singapore, 169856 Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077 Singapore
- Ophthalmology and Visual Sciences Academic Clinical Research Program, Duke-NUS Medical School, Singapore, 169857 Singapore
| | - Kah Leong Lim
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
- Developmental of Stem Cell Biology and Regenerative Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232 Singapore
| | - Eng-King Tan
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
- Department of Neurology, Singapore General Hospital, National Neuroscience Institute, Outram Road, Singapore, 169608 Singapore
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioural Disorders, 8 College Road, Singapore, 169857 Singapore
| |
Collapse
|
18
|
Brzozowski CF, Hijaz BA, Singh V, Gcwensa NZ, Kelly K, Boyden ES, West AB, Sarkar D, Volpicelli-Daley LA. Inhibition of LRRK2 kinase activity promotes anterograde axonal transport and presynaptic targeting of α-synuclein. Acta Neuropathol Commun 2021; 9:180. [PMID: 34749824 PMCID: PMC8576889 DOI: 10.1186/s40478-021-01283-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/20/2021] [Indexed: 01/18/2023] Open
Abstract
Pathologic inclusions composed of α-synuclein called Lewy pathology are hallmarks of Parkinson’s Disease (PD). Dominant inherited mutations in leucine rich repeat kinase 2 (LRRK2) are the most common genetic cause of PD. Lewy pathology is found in the majority of individuals with LRRK2-PD, particularly those with the G2019S-LRRK2 mutation. Lewy pathology in LRRK2-PD associates with increased non-motor symptoms such as cognitive deficits, anxiety, and orthostatic hypotension. Thus, understanding the relationship between LRRK2 and α-synuclein could be important for determining the mechanisms of non-motor symptoms. In PD models, expression of mutant LRRK2 reduces membrane localization of α-synuclein, and enhances formation of pathologic α-synuclein, particularly when synaptic activity is increased. α-Synuclein and LRRK2 both localize to the presynaptic terminal. LRRK2 plays a role in membrane traffic, including axonal transport, and therefore may influence α-synuclein synaptic localization. This study shows that LRRK2 kinase activity influences α-synuclein targeting to the presynaptic terminal. We used the selective LRRK2 kinase inhibitors, MLi-2 and PF-06685360 (PF-360) to determine the impact of reduced LRRK2 kinase activity on presynaptic localization of α-synuclein. Expansion microscopy (ExM) in primary hippocampal cultures and the mouse striatum, in vivo, was used to more precisely resolve the presynaptic localization of α-synuclein. Live imaging of axonal transport of α-synuclein-GFP was used to investigate the impact of LRRK2 kinase inhibition on α-synuclein axonal transport towards the presynaptic terminal. Reduced LRRK2 kinase activity increases α-synuclein overlap with presynaptic markers in primary neurons, and increases anterograde axonal transport of α-synuclein-GFP. In vivo, LRRK2 inhibition increases α-synuclein overlap with glutamatergic, cortico-striatal terminals, and dopaminergic nigral-striatal presynaptic terminals. The findings suggest that LRRK2 kinase activity plays a role in axonal transport, and presynaptic targeting of α-synuclein. These data provide potential mechanisms by which LRRK2-mediated perturbations of α-synuclein localization could cause pathology in both LRRK2-PD, and idiopathic PD.
Collapse
|
19
|
Wojewska DN, Kortholt A. LRRK2 Targeting Strategies as Potential Treatment of Parkinson's Disease. Biomolecules 2021; 11:1101. [PMID: 34439767 PMCID: PMC8392603 DOI: 10.3390/biom11081101] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023] Open
Abstract
Parkinson's Disease (PD) affects millions of people worldwide with no cure to halt the progress of the disease. Leucine-rich repeat kinase 2 (LRRK2) is the most common genetic cause of PD and, as such, LRRK2 inhibitors are promising therapeutic agents. In the last decade, great progress in the LRRK2 field has been made. This review provides a comprehensive overview of the current state of the art, presenting recent developments and challenges in developing LRRK2 inhibitors, and discussing extensively the potential targeting strategies from the protein perspective. As currently there are three LRRK2-targeting agents in clinical trials, more developments are predicted in the upcoming years.
Collapse
Affiliation(s)
- Dominika Natalia Wojewska
- Faculty of Science and Engineering, University of Groningen, Nijenborg 7, 9747AG Groningen, The Netherlands;
| | - Arjan Kortholt
- Faculty of Science and Engineering, University of Groningen, Nijenborg 7, 9747AG Groningen, The Netherlands;
- YETEM-Innovative Technologies Application and Research Center, Suleyman Demirel University, 32260 Isparta, Turkey
| |
Collapse
|
20
|
Cresto N, Gardier C, Gaillard MC, Gubinelli F, Roost P, Molina D, Josephine C, Dufour N, Auregan G, Guillermier M, Bernier S, Jan C, Gipchtein P, Hantraye P, Chartier-Harlin MC, Bonvento G, Van Camp N, Taymans JM, Cambon K, Liot G, Bemelmans AP, Brouillet E. The C-Terminal Domain of LRRK2 with the G2019S Substitution Increases Mutant A53T α-Synuclein Toxicity in Dopaminergic Neurons In Vivo. Int J Mol Sci 2021; 22:ijms22136760. [PMID: 34201785 PMCID: PMC8268201 DOI: 10.3390/ijms22136760] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/10/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022] Open
Abstract
Alpha-synuclein (α-syn) and leucine-rich repeat kinase 2 (LRRK2) play crucial roles in Parkinson's disease (PD). They may functionally interact to induce the degeneration of dopaminergic (DA) neurons via mechanisms that are not yet fully understood. We previously showed that the C-terminal portion of LRRK2 (ΔLRRK2) with the G2019S mutation (ΔLRRK2G2019S) was sufficient to induce neurodegeneration of DA neurons in vivo, suggesting that mutated LRRK2 induces neurotoxicity through mechanisms that are (i) independent of the N-terminal domains and (ii) "cell-autonomous". Here, we explored whether ΔLRRK2G2019S could modify α-syn toxicity through these two mechanisms. We used a co-transduction approach in rats with AAV vectors encoding ΔLRRK2G2019S or its "dead" kinase form, ΔLRRK2DK, and human α-syn with the A53T mutation (AAV-α-synA53T). Behavioral and histological evaluations were performed at 6- and 15-weeks post-injection. Results showed that neither form of ΔLRRK2 alone induced the degeneration of neurons at these post-injection time points. By contrast, injection of AAV-α-synA53T alone resulted in motor signs and degeneration of DA neurons. Co-injection of AAV-α-synA53T with AAV-ΔLRRK2G2019S induced DA neuron degeneration that was significantly higher than that induced by AAV-α-synA53T alone or with AAV-ΔLRRK2DK. Thus, mutated α-syn neurotoxicity can be enhanced by the C-terminal domain of LRRK2G2019 alone, through cell-autonomous mechanisms.
Collapse
Affiliation(s)
- Noémie Cresto
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Camille Gardier
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Marie-Claude Gaillard
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Francesco Gubinelli
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Pauline Roost
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Daniela Molina
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Charlène Josephine
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Noëlle Dufour
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Gwenaëlle Auregan
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Martine Guillermier
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Suéva Bernier
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Caroline Jan
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Pauline Gipchtein
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Philippe Hantraye
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Marie-Christine Chartier-Harlin
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France; (M.-C.C.-H.); (J.-M.T.)
- Brain Biology and Chemistry, LiCEND, F-59000 Lille, France
| | - Gilles Bonvento
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Nadja Van Camp
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Jean-Marc Taymans
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France; (M.-C.C.-H.); (J.-M.T.)
- Brain Biology and Chemistry, LiCEND, F-59000 Lille, France
| | - Karine Cambon
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Géraldine Liot
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Alexis-Pierre Bemelmans
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
| | - Emmanuel Brouillet
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, MIRCen, F-92265 Fontenay-aux-Roses, France; (N.C.); (C.G.); (M.-C.G.); (F.G.); (P.R.); (D.M.); (C.J.); (N.D.); (G.A.); (M.G.); (S.B.); (C.J.); (P.G.); (P.H.); (G.B.); (N.V.C.); (K.C.); (G.L.); (A.-P.B.)
- Correspondence:
| |
Collapse
|
21
|
Leucine-rich repeat kinase 2-related functions in GLIA: an update of the last years. Biochem Soc Trans 2021; 49:1375-1384. [PMID: 33960369 DOI: 10.1042/bst20201092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/13/2022]
Abstract
Missense mutations in the leucine-rich repeat kinase-2 (LRRK2) gene represent the most common cause of autosomal dominant Parkinson's disease (PD). In the years LRRK2 has been associated with several organelles and related pathways in cell. However, despite the significant amount of research done in the past decade, the contribution of LRRK2 mutations to PD pathogenesis remains unknown. Growing evidence highlights that LRRK2 controls multiple processes in brain immune cells, microglia and astrocytes, and suggests that deregulated LRRK2 activity in these cells, due to gene mutation, might be directly associated with pathological mechanisms underlying PD. In this brief review, we recapitulate and update the last LRRK2 functions dissected in microglia and astrocytes. Moreover, we discuss how dysfunctions of LRRK2-related pathways may impact glia physiology and their cross-talk with neurons, thus leading to neurodegeneration and progression of PD.
Collapse
|
22
|
Goldstein O, Gana-Weisz M, Casey F, Meltzer-Fridrich H, Yaacov O, Waldman YY, Lin D, Mordechai Y, Zhu J, Cullen PF, Omer N, Shiner T, Thaler A, Bar-Shira A, Mirelman A, John S, Giladi N, Orr-Urtreger A. PARK16 locus: Differential effects of the non-coding rs823114 on Parkinson’s disease risk, RNA expression, and DNA methylation. J Genet Genomics 2021; 48:341-345. [DOI: 10.1016/j.jgg.2020.10.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/29/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
|
23
|
Controversy of TMEM230 Associated with Parkinson's Disease. Neuroscience 2020; 453:280-286. [PMID: 33212219 DOI: 10.1016/j.neuroscience.2020.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease with movement disorders including resting tremor, bradykinesia, rigidity, and postural instability. The key pathological features of PD are selective loss of dopaminergic (DA) neurons in substantial nigra and the presence of Lewy bodies (LBs). Mutations in TMEM230 (transmembrane protein 230) have been recently reported to play a pathological role and contribute to PD pathogenesis. TMEM230 gene encodes two isoforms of TMEM230 proteins, isoform I (183 amino acids) and isoform II (120 amino acids). The function of TMEM230 is not clear, but it may be involved in vesicle trafficking and recycling, autophagy, protein aggregation, and cell toxicity. There are four reported PD-linked TMEM230 mutations (p.Y92C, p.R141L, p.*184Wext*5, p.*184PGext*5). TMEM230-linked PD cases exhibit late-onset, good-response to levodopa, and typical clinical features of sporadic PD with DA neuronal loss in substantial nigra and Lewy body pathology. In this mini review, we recap the current literature of TMEM230 in genetic, neurobiological, and pathological studies in order to further understand the potential roles of TMEM230 in PD pathogenesis.
Collapse
|
24
|
Dawson VL, Dawson TM. Promising disease-modifying therapies for Parkinson's disease. Sci Transl Med 2020; 11:11/520/eaba1659. [PMID: 31776289 DOI: 10.1126/scitranslmed.aba1659] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 11/14/2019] [Indexed: 12/11/2022]
Abstract
To date, there is no disease-modifying therapy for Parkinson's disease; however, promising new agents have advanced into clinical trials.
Collapse
Affiliation(s)
- Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
25
|
LRRK2 Regulates CPT1A to Promote β-Oxidation in HepG2 Cells. Molecules 2020; 25:molecules25184122. [PMID: 32916992 PMCID: PMC7570678 DOI: 10.3390/molecules25184122] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is involved in lipid metabolism; however, the role of LRRK2 in lipid metabolism to affect non-alcoholic fatty liver disease (NAFLD) is still unclear. In the mouse model of NAFLD induced by a high-fat diet, we observed that LRRK2 was decreased in livers. In HepG2 cells, exposure to palmitic acid (PA) down-regulated LRRK2. Overexpression and knockdown of LRRK2 in HepG2 cells were performed to further investigate the roles of LRRK2 in lipid metabolism. Our results showed that β-oxidation in HepG2 cells was promoted by LRRK2 overexpression, whereas LRRK2 knockdown inhibited β-oxidation. The critical enzyme of β-oxidation, carnitine palmitoyltransferase 1A (CPT1A), was positively regulated by LRRK2. Our data suggested that the regulation of CPT1A by LRRK2 may be via the activation of AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor α (PPARα). The overexpression of LRRK2 reduced the concentration of a pro-inflammatory cytokine, tumor necrosis factor α (TNFα), induced by PA. The increase in β-oxidation may promote lipid catabolism to suppress inflammation induced by PA. These results indicated that LRRK2 participated in the regulation of β-oxidation and suggested that the decreased LRRK2 may promote inflammation by suppressing β-oxidation in the liver.
Collapse
|
26
|
Murata T, Unno Y, Fukuda M, Utsunomiya-Tate N. The dynamic structure of Rab35 is stabilized in the presence of GTP under physiological conditions. Biochem Biophys Rep 2020; 23:100776. [PMID: 32613087 PMCID: PMC7322127 DOI: 10.1016/j.bbrep.2020.100776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
Abstract
Rab proteins, a family of small guanosine triphosphatases, play key roles in intracellular membrane trafficking and the regulation of various cellular processes. As a Rab isoform, Rab35 is crucial for recycling endosome trafficking, cytokinesis and neurite outgrowth. In this report, we analyzed dynamic structural changes and physicochemical features of Rab35 in response to different external conditions, including temperature, pH, salt concentration and guanosine triphosphate (GTP), by circular dichroism (CD) spectroscopy. CD spectra revealed that the α-helix content of Rab35 varies under different conditions considerably. The addition of GTP increases the α-helix content of Rab35 when the temperature, pH and salt concentration match physiological conditions. The results suggest that the external environment affects the secondary structure of Rab35. In particular, the presence of GTP stabilized the α-helices of Rab35 under physiological conditions. These structural changes may translate to changes in Rab35 function and relate to its role in membrane trafficking.
Collapse
Affiliation(s)
- Takuya Murata
- Faculty of Pharma-Science, Teikyo University, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Yuka Unno
- Faculty of Pharma-Science, Teikyo University, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Naoko Utsunomiya-Tate
- Faculty of Pharma-Science, Teikyo University, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| |
Collapse
|
27
|
Small GTPases of the Ras and Rho Families Switch on/off Signaling Pathways in Neurodegenerative Diseases. Int J Mol Sci 2020. [DOI: 10.3390/ijms21176312
expr 858053618 + 832508766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Small guanosine triphosphatases (GTPases) of the Ras superfamily are key regulators of many key cellular events such as proliferation, differentiation, cell cycle regulation, migration, or apoptosis. To control these biological responses, GTPases activity is regulated by guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and in some small GTPases also guanine nucleotide dissociation inhibitors (GDIs). Moreover, small GTPases transduce signals by their downstream effector molecules. Many studies demonstrate that small GTPases of the Ras family are involved in neurodegeneration processes. Here, in this review, we focus on the signaling pathways controlled by these small protein superfamilies that culminate in neurodegenerative pathologies, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). Specifically, we concentrate on the two most studied families of the Ras superfamily: the Ras and Rho families. We summarize the latest findings of small GTPases of the Ras and Rho families in neurodegeneration in order to highlight these small proteins as potential therapeutic targets capable of slowing down different neurodegenerative diseases.
Collapse
|
28
|
Arrazola Sastre A, Luque Montoro M, Gálvez-Martín P, Lacerda HM, Lucia A, Llavero F, Zugaza JL. Small GTPases of the Ras and Rho Families Switch on/off Signaling Pathways in Neurodegenerative Diseases. Int J Mol Sci 2020; 21:E6312. [PMID: 32878220 PMCID: PMC7504559 DOI: 10.3390/ijms21176312] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 12/16/2022] Open
Abstract
Small guanosine triphosphatases (GTPases) of the Ras superfamily are key regulators of many key cellular events such as proliferation, differentiation, cell cycle regulation, migration, or apoptosis. To control these biological responses, GTPases activity is regulated by guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and in some small GTPases also guanine nucleotide dissociation inhibitors (GDIs). Moreover, small GTPases transduce signals by their downstream effector molecules. Many studies demonstrate that small GTPases of the Ras family are involved in neurodegeneration processes. Here, in this review, we focus on the signaling pathways controlled by these small protein superfamilies that culminate in neurodegenerative pathologies, such as Alzheimer's disease (AD) and Parkinson's disease (PD). Specifically, we concentrate on the two most studied families of the Ras superfamily: the Ras and Rho families. We summarize the latest findings of small GTPases of the Ras and Rho families in neurodegeneration in order to highlight these small proteins as potential therapeutic targets capable of slowing down different neurodegenerative diseases.
Collapse
Affiliation(s)
- Alazne Arrazola Sastre
- Achucarro Basque Center for Neuroscience, Science Park of the Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48940 Leioa, Spain; (A.A.S.); (M.L.M.)
- Department of Genetics, Physical Anthropology, and Animal Physiology, Faculty of Science and Technology, UPV/EHU, 48940 Leioa, Spain
| | - Miriam Luque Montoro
- Achucarro Basque Center for Neuroscience, Science Park of the Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48940 Leioa, Spain; (A.A.S.); (M.L.M.)
| | - Patricia Gálvez-Martín
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, 180041 Granada, Spain;
- R&D Human Health, Bioibérica S.A.U., 08950 Barcelona, Spain
| | | | - Alejandro Lucia
- Faculty of Sport Science, European University of Madrid, 28670 Madrid, Spain;
- Research Institute of the Hospital 12 de Octubre (i+12), 28041 Madrid, Spain
| | - Francisco Llavero
- Achucarro Basque Center for Neuroscience, Science Park of the Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48940 Leioa, Spain; (A.A.S.); (M.L.M.)
- Faculty of Sport Science, European University of Madrid, 28670 Madrid, Spain;
| | - José Luis Zugaza
- Achucarro Basque Center for Neuroscience, Science Park of the Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48940 Leioa, Spain; (A.A.S.); (M.L.M.)
- Department of Genetics, Physical Anthropology, and Animal Physiology, Faculty of Science and Technology, UPV/EHU, 48940 Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|
29
|
O'Hara DM, Pawar G, Kalia SK, Kalia LV. LRRK2 and α-Synuclein: Distinct or Synergistic Players in Parkinson's Disease? Front Neurosci 2020; 14:577. [PMID: 32625052 PMCID: PMC7311858 DOI: 10.3389/fnins.2020.00577] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 05/12/2020] [Indexed: 12/19/2022] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder, characterized by prominent degeneration of dopaminergic neurons in the substantia nigra and aggregation of the protein α-synuclein within intraneuronal inclusions known as Lewy bodies. Ninety percent of PD cases are idiopathic while the remaining 10% are associated with gene mutations that affect cellular functions ranging from kinase activity to mitochondrial quality control, hinting at a multifactorial disease process. Mutations in LRRK2 and SNCA (the gene coding for α-synuclein) cause monogenic forms of autosomal dominant PD, and polymorphisms in either gene are also associated with increased risk of idiopathic PD. Although Lewy bodies are a defining neuropathological feature of PD, an appreciable subset of patients with LRRK2 mutations present with a clinical phenotype indistinguishable from idiopathic PD but lack Lewy pathology at autopsy, suggesting that LRRK2-mediated PD may occur independently of α-synuclein aggregation. Here, we examine whether LRRK2 and α-synuclein, as mediators of neurodegeneration in PD, exist in common or distinct pathways. Specifically, we review evidence from preclinical models and human neuropathological studies examining interactions between the two proteins. Elucidating the degree of interplay between LRRK2 and α-synuclein will be necessary for treatment stratification once effective targeted disease-modifying therapies are developed.
Collapse
Affiliation(s)
- Darren M O'Hara
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Grishma Pawar
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Suneil K Kalia
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Lorraine V Kalia
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Division of Neurology, Department of Medicine, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.,Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
30
|
Guevara CA, Matikainen-Ankney BA, Kezunovic N, LeClair K, Conway AP, Menard C, Flanigan ME, Pfau M, Russo SJ, Benson DL, Huntley GW. LRRK2 mutation alters behavioral, synaptic, and nonsynaptic adaptations to acute social stress. J Neurophysiol 2020; 123:2382-2389. [PMID: 32374202 DOI: 10.1152/jn.00137.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) risk is increased by stress and certain gene mutations, including the most prevalent PD-linked mutation LRRK2-G2019S. Both PD and stress increase risk for psychiatric symptoms, yet it is unclear how PD-risk genes alter neural circuitry in response to stress that may promote psychopathology. Here we show significant differences between adult G2019S knockin and wild-type (wt) mice in stress-induced behaviors, with an unexpected uncoupling of depression-like and hedonia-like responses in G2019S mice. Moreover, mutant spiny projection neurons in nucleus accumbens (NAc) lack an adaptive, stress-induced change in excitability displayed by wt neurons, and instead show stress-induced changes in synaptic properties that wt neurons lack. Some synaptic alterations in NAc are already evident early in postnatal life. Thus G2019S alters the magnitude and direction of behavioral responses to stress that may reflect unique modifications of adaptive plasticity in cells and circuits implicated in psychopathology in humans.NEW & NOTEWORTHY Depression is associated with Parkinson's disease (PD), and environmental stress is a risk factor for both. We investigated how LRRK2-G2019S PD mutation affects depression-like behaviors, synaptic function, and intrinsic neuronal excitability following stress. In response to stress, the mutation drives abnormal synaptic changes, prevents adaptive changes in intrinsic excitability, and leads to aberrant behaviors, thus defining new ways in which PD mutations derail adaptive plasticity in response to stress that may contribute to disease onset.
Collapse
Affiliation(s)
- Christopher A Guevara
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Bridget A Matikainen-Ankney
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nebojsa Kezunovic
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Katherine LeClair
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alexander P Conway
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Caroline Menard
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Meghan E Flanigan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Madeline Pfau
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Deanna L Benson
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - George W Huntley
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|