1
|
Raza A, Raina J, Sahu SK, Wadhwa P. Genetic mutations in kinases: a comprehensive review on marketed inhibitors and unexplored targets in Parkinson's disease. Neurol Sci 2025; 46:1509-1524. [PMID: 39760821 DOI: 10.1007/s10072-024-07970-2] [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: 10/15/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
This comprehensive review navigates the landscape of genetic mutations in kinases, offering a thorough examination of both marketed inhibitors and unexplored targets in the context of Parkinson's Disease (PD). Although existing treatments for PD primarily center on symptom management, progress in comprehending the molecular foundations of the disease has opened avenues for targeted therapeutic approaches. This review encompasses an in-depth analysis of four key kinases-PINK1, LRRK2, GAK, and PRKRA-revealing that LRRK2 has garnered the most attention with a plethora of marketed inhibitors. However, the study underscores notable gaps in the exploration of inhibitors for PINK1, GAK, and a complete absence for PRKRA. The observed scarcity of inhibitors for these kinases emphasizes a significant area of untapped potential in PD therapeutics. By drawing attention to these unexplored targets, the review highlights the urgent need for focused research and drug development efforts to diversify the therapeutic landscape, potentially providing novel interventions for halting or slowing the progression of PD.
Collapse
Affiliation(s)
- Amir Raza
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar- Grand Trunk Rd, Phagwara, Punjab, India
| | - Jeevika Raina
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar- Grand Trunk Rd, Phagwara, Punjab, India
| | - Sanjeev Kumar Sahu
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar- Grand Trunk Rd, Phagwara, Punjab, India
| | - Pankaj Wadhwa
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar- Grand Trunk Rd, Phagwara, Punjab, India.
| |
Collapse
|
2
|
Morez M, Lara Ordóñez AJ, Melnyk P, Liberelle M, Lebègue N, Taymans JM. Leucine-rich repeat kinase 2 (LRRK2) inhibitors for Parkinson's disease: a patent review of the literature to date. Expert Opin Ther Pat 2024; 34:773-788. [PMID: 39023243 DOI: 10.1080/13543776.2024.2378076] [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: 04/26/2024] [Accepted: 07/04/2024] [Indexed: 07/20/2024]
Abstract
INTRODUCTION Nearly two decades after leucine rich repeat kinase 2 (LRRK2) was discovered as a genetic determinant of Parkinson's disease (PD), LRRK2 has emerged a priority therapeutic target in PD and inhibition of its activity is hypothesized to be beneficial. AREAS COVERED LRRK2 targeting agents, in particular kinase inhibitors and agents reducing LRRK2 expression show promise in model systems and have progressed to phase I and phase II clinical testing for PD. Several additional targeting strategies for LRRK2 are emerging, based on promoting specific 'healthy' LRRK2 quaternary structures, heteromeric complexes and conformations. EXPERT OPINION It can be expected that LRRK2 targeting strategies may proceed to phase III clinical testing for PD in the next five years, allowing the field to discover the real clinical value of LRRK2 targeting strategies.
Collapse
Affiliation(s)
- Margaux Morez
- University Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | | | - Patricia Melnyk
- University Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Maxime Liberelle
- University Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Nicolas Lebègue
- University Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Jean-Marc Taymans
- University Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| |
Collapse
|
3
|
Wang Q, Gu X, Yang L, Jiang Y, Zhang J, He J. Emerging perspectives on precision therapy for Parkinson's disease: multidimensional evidence leading to a new breakthrough in personalized medicine. Front Aging Neurosci 2024; 16:1417515. [PMID: 39026991 PMCID: PMC11254646 DOI: 10.3389/fnagi.2024.1417515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
PD is a prevalent and progressive neurodegenerative disorder characterized by both motor and non-motor symptoms. Genes play a significant role in the onset and progression of the disease. While the complexity and pleiotropy of gene expression networks have posed challenges for gene-targeted therapies, numerous pathways of gene variant expression show promise as therapeutic targets in preclinical studies, with some already in clinical trials. With the recognition of the numerous genes and complex pathways that can influence PD, it may be possible to take a novel approach to choose a treatment for the condition. This approach would be based on the symptoms, genomics, and underlying mechanisms of the disease. We discuss the utilization of emerging genetic and pathological knowledge of PD patients to categorize the disease into subgroups. Our long-term objective is to generate new insights for the therapeutic approach to the disease, aiming to delay and treat it more effectively, and ultimately reduce the burden on individuals and society.
Collapse
Affiliation(s)
- Qiaoli Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xuan Gu
- Department of Trauma center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Le Yang
- Department of Endocrinology, The People’s Hospital of Jilin Province, Changchun, China
| | - Yan Jiang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiao Zhang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinting He
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Lubben N, Brynildsen JK, Webb CM, Li HL, Leyns CEG, Changolkar L, Zhang B, Meymand ES, O'Reilly M, Madaj Z, DeWeerd D, Fell MJ, Lee VMY, Bassett DS, Henderson MX. LRRK2 kinase inhibition reverses G2019S mutation-dependent effects on tau pathology progression. Transl Neurodegener 2024; 13:13. [PMID: 38438877 PMCID: PMC10910783 DOI: 10.1186/s40035-024-00403-2] [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: 10/09/2023] [Accepted: 02/07/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common cause of familial Parkinson's disease (PD). These mutations elevate the LRRK2 kinase activity, making LRRK2 kinase inhibitors an attractive therapeutic. LRRK2 kinase activity has been consistently linked to specific cell signaling pathways, mostly related to organelle trafficking and homeostasis, but its relationship to PD pathogenesis has been more difficult to define. LRRK2-PD patients consistently present with loss of dopaminergic neurons in the substantia nigra but show variable development of Lewy body or tau tangle pathology. Animal models carrying LRRK2 mutations do not develop robust PD-related phenotypes spontaneously, hampering the assessment of the efficacy of LRRK2 inhibitors against disease processes. We hypothesized that mutations in LRRK2 may not be directly related to a single disease pathway, but instead may elevate the susceptibility to multiple disease processes, depending on the disease trigger. To test this hypothesis, we have previously evaluated progression of α-synuclein and tau pathologies following injection of proteopathic seeds. We demonstrated that transgenic mice overexpressing mutant LRRK2 show alterations in the brain-wide progression of pathology, especially at older ages. METHODS Here, we assess tau pathology progression in relation to long-term LRRK2 kinase inhibition. Wild-type or LRRK2G2019S knock-in mice were injected with tau fibrils and treated with control diet or diet containing LRRK2 kinase inhibitor MLi-2 targeting the IC50 or IC90 of LRRK2 for 3-6 months. Mice were evaluated for tau pathology by brain-wide quantitative pathology in 844 brain regions and subsequent linear diffusion modeling of progression. RESULTS Consistent with our previous work, we found systemic alterations in the progression of tau pathology in LRRK2G2019S mice, which were most pronounced at 6 months. Importantly, LRRK2 kinase inhibition reversed these effects in LRRK2G2019S mice, but had minimal effect in wild-type mice, suggesting that LRRK2 kinase inhibition is likely to reverse specific disease processes in G2019S mutation carriers. Additional work may be necessary to determine the potential effect in non-carriers. CONCLUSIONS This work supports a protective role of LRRK2 kinase inhibition in G2019S carriers and provides a rational workflow for systematic evaluation of brain-wide phenotypes in therapeutic development.
Collapse
Affiliation(s)
- Noah Lubben
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Julia K Brynildsen
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Connor M Webb
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Institute On Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Howard L Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Institute On Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Cheryl E G Leyns
- Neuroscience Discovery, Merck & Co., Inc., Boston, MA, 02115, USA
| | - Lakshmi Changolkar
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Institute On Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Bin Zhang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Institute On Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Emily S Meymand
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Institute On Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mia O'Reilly
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Zach Madaj
- Bioinformatics and Biostatistics Core, Van Andel Institute, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503, USA
| | - Daniella DeWeerd
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Matthew J Fell
- Neuroscience Discovery, Merck & Co., Inc., Boston, MA, 02115, USA
| | - Virginia M Y Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Institute On Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dani S Bassett
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Santa Fe Institute, Santa Fe, NM, 87501, USA
| | - Michael X Henderson
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
5
|
Pena N, Richbourg T, Gonzalez-Hunt CP, Qi R, Wren P, Barlow C, Shanks NF, Carlisle HJ, Sanders LH. G2019S selective LRRK2 kinase inhibitor abrogates mitochondrial DNA damage. NPJ Parkinsons Dis 2024; 10:49. [PMID: 38429321 PMCID: PMC10907374 DOI: 10.1038/s41531-024-00660-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/14/2024] [Indexed: 03/03/2024] Open
Abstract
Pathogenic mutations in LRRK2 cause Parkinson's disease (PD). The G2019S variant is the most common, which results in abnormally high kinase activity. Compounds that target LRRK2 kinase activity are currently being developed and tested in clinical trials. We recently found that G2019S LRRK2 causes mitochondrial DNA (mtDNA) damage and treatment with multiple classes of LRRK2 kinase inhibitors at concentrations associated with dephosphorylation of LRRK2 reversed mtDNA damage to healthy control levels. Because maintaining the normal function of LRRK2 in heterozygous G2019S LRRK2 carriers while specifically targeting the G2019S LRRK2 activity could have an advantageous safety profile, we explored the efficacy of a G2019S mutant selective LRRK2 inhibitor to reverse mtDNA damage in G2019S LRRK2 models and patient cells relative to non-selective LRRK2 inhibitors. Potency of LRRK2 kinase inhibition by EB-42168, a G2019S mutant LRRK2 kinase inhibitor, and MLi-2, a non-selective inhibitor, was determined by measuring phosphorylation of LRRK2 at Ser935 and/or Ser1292 using quantitative western immunoblot analysis. The Mito DNADX assay, which allows for the accurate real-time quantification of mtDNA damage in a 96-well platform, was performed in parallel. We confirmed that EB-42168 selectively inhibits LRRK2 phosphorylation on G2019S LRRK2 relative to wild-type LRRK2. On the other hand, MLi-2 was equipotent for wild-type and G2019S LRRK2. Acute treatment with EB-42168 inhibited LRRK2 phosphorylation and also restored mtDNA damage to healthy control levels. We further investigated the relationship between LRRK2 kinase activity, mtDNA damage and mitophagy. Levels of mtDNA damage caused by G2019S LRRK2 were fully re-established within 2 h of a LRRK2 inhibitor wash out and recovery experiment, indicating the mtDNA damage phenotype is highly dynamic. G2019S LRRK2 mitophagy defects were not alleviated with LRRK2 kinase inhibition, suggesting that mitophagy is not mechanistically regulating LRRK2 kinase-mediated reversal of mtDNA damage in this acute timeframe. Abrogation of mtDNA damage with the mutant selective tool inhibitor EB-42168 demonstrates the potential of a precision medicine approach for LRRK2 G2019S PD. Levels of mtDNA damage may serve as a potential pharmacodynamic biomarker of altered kinase activity that could be useful for small molecule development and clinical trials.
Collapse
Affiliation(s)
- Nicholas Pena
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, NC, USA
| | - Tara Richbourg
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, NC, USA
| | - Claudia P Gonzalez-Hunt
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, NC, USA
| | - Rui Qi
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, NC, USA
| | - Paul Wren
- ESCAPE Bio, Inc., South San Francisco, CA, 94080, USA
| | | | | | | | - Laurie H Sanders
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, NC, USA.
| |
Collapse
|
6
|
Østergaard FG. Knocking out the LRRK2 gene increases sensitivity to wavelength information in rats. Sci Rep 2024; 14:4984. [PMID: 38424139 PMCID: PMC10904730 DOI: 10.1038/s41598-024-55350-9] [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: 02/08/2023] [Accepted: 02/21/2024] [Indexed: 03/02/2024] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a gene related to familial Parkinson's disease (PD). It has been associated with nonmotor symptoms such as disturbances in the visual system affecting colour discrimination and contrast sensitivity. This study examined how deficiency of LRRK2 impacts visual processing in adult rats. Additionally, we investigated whether these changes can be modelled in wild-type rats by administering the LRRK2 inhibitor PFE360. Visual evoked potentials (VEPs) and steady-state visual evoked potentials (SSVEPs) were recorded in the visual cortex and superior colliculus of female LRRK2-knockout and wild-type rats to study how the innate absence of LRRK2 changes visual processing. Exposing the animals to stimulation at five different wavelengths revealed an interaction between genotype and the response to stimulation at different wavelengths. Differences in VEP amplitudes and latencies were robust and barely impacted by the presence of the LRRK2 inhibitor PFE360, suggesting a developmental effect. Taken together, these results indicate that alterations in visual processing were related to developmental deficiency of LRRK2 and not acute deficiency of LRRK2, indicating a role of LRRK2 in the functional development of the visual system and synaptic transmission.
Collapse
Affiliation(s)
- Freja Gam Østergaard
- H. Lundbeck A/S, Ottiliavej 9, 2500, Valby, Denmark.
- GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands.
| |
Collapse
|
7
|
Cao R, Chen C, Wen J, Zhao W, Zhang C, Sun L, Yuan L, Wu C, Shan L, Xi M, Sun H. Recent advances in targeting leucine-rich repeat kinase 2 as a potential strategy for the treatment of Parkinson's disease. Bioorg Chem 2023; 141:106906. [PMID: 37837728 DOI: 10.1016/j.bioorg.2023.106906] [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: 08/17/2023] [Revised: 09/24/2023] [Accepted: 10/06/2023] [Indexed: 10/16/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. Several single gene mutations involved in PD have been identified such as leucine-rich repeat kinase 2 (LRRK2), the most common cause of sporadic and familial PD. Its mutations have attracted much attention to therapeutically targeting this kinase. To date, many compounds including small chemical molecules with diverse scaffolds and RNA agents have been developed with significant amelioration in preclinical PD models. Currently, five candidates, DNL201, DNL151, WXWH0226, NEU-723 and BIIB094, have advanced to clinical trials for PD treatment. In this review, we describe the structure, pathogenic mutations and the mechanism of LRRK2, and summarize the development of LRRK2 inhibitors in preclinical and clinical studies, trying to provide an insight into targeting LRRK2 for PD intervention in future.
Collapse
Affiliation(s)
- Ruiwei Cao
- Zhejiang Engineering Research Center of Fat-soluble Vitamin, Shaoxing University, Shaoxing 312000, China; Zhejiang Medicine Co. Ltd., Shaoxing 312500, China
| | - Caiping Chen
- Zhejiang Engineering Research Center of Fat-soluble Vitamin, Shaoxing University, Shaoxing 312000, China; Zhejiang Medicine Co. Ltd., Shaoxing 312500, China
| | - Jing Wen
- Zhejiang Medicine Co. Ltd., Shaoxing 312500, China
| | - Weihe Zhao
- Zhejiang Medicine Co. Ltd., Shaoxing 312500, China
| | | | - Longhui Sun
- Zhejiang Medicine Co. Ltd., Shaoxing 312500, China
| | - Liyan Yuan
- Zhejiang Medicine Co. Ltd., Shaoxing 312500, China
| | - Chunlei Wu
- Zhejiang Engineering Research Center of Fat-soluble Vitamin, Shaoxing University, Shaoxing 312000, China; College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing 312000, China
| | - Lei Shan
- Zhejiang Engineering Research Center of Fat-soluble Vitamin, Shaoxing University, Shaoxing 312000, China
| | - Meiyang Xi
- Zhejiang Engineering Research Center of Fat-soluble Vitamin, Shaoxing University, Shaoxing 312000, China; College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing 312000, China.
| | - Haopeng Sun
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
8
|
Kwon MJ, Kim JK, Kim JH, Kim JH, Kim MJ, Kim NY, Choi HG, Kim ES. Exploring the Link between Chronic Kidney Disease and Parkinson's Disease: Insights from a Longitudinal Study Using a National Health Screening Cohort. Nutrients 2023; 15:3205. [PMID: 37513623 PMCID: PMC10385674 DOI: 10.3390/nu15143205] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Chronic kidney disease (CKD) and Parkinson's disease (PD) are common illnesses found in the geriatric population. A potential link between CKD and PD emergence has been hypothesized; however, existing conclusions are disputed. In this longitudinal research, we analyzed data acquired from the Korean National Health Insurance Service-Health Screening Cohort. The dataset comprised the health information of 16,559 individuals clinically diagnosed with CKD and 66,236 control subjects of comparable ages, all aged ≥40 years. These subjects participated in health examinations from 2002 to 2019. To assess the correlation between CKD and PD, we employed overlap-weighted Cox proportional hazard regression models. The unadjusted, crude hazard ratio for PD was greater in the CKD group than in the control group (crude hazard ration (HR) 1.20; 95% confidence interval (CI) = 1.04-1.39; p = 0.011). However, the Cox proportional hazard regression analysis, incorporating propensity score overlap weighting, revealed no significant discrepancy after considering confounding variables such as demographic factors, socio-economic status, lifestyle, and concurrent health conditions (adjusted HR (aHR), 1.09; 95% CI = 0.97-1.22; p = 0.147). Subgroup analyses showed a higher probability of PD development among certain CKD individuals, including those who resided in rural areas (aHR, 1.19; 95% CI = 1.03-1.37; p = 0.022), maintained a normal weight (aHR, 1.29; 95% CI = 1.08-1.56; p = 0.006), or had fasting blood glucose levels ≥100 mg/dL (aHR, 1.18; 95% CI = 1.00-1.39; p = 0.046). Therefore, these clinical or environmental factors may influence the incidence of PD in CKD patients. In conclusion, our results suggest that the general CKD population may not exhibit a greater propensity for PD than their non-CKD counterparts. However, this might be contingent upon specific lifestyle and comorbid conditions. Thus, certain lifestyle alterations could be crucial in mitigating the potential manifestation of PD in patients diagnosed with CKD.
Collapse
Affiliation(s)
- Mi Jung Kwon
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Republic of Korea
| | - Jwa-Kyung Kim
- Division of Nephrology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Republic of Korea
| | - Ji Hee Kim
- Department of Neurosurgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Republic of Korea
| | - Joo-Hee Kim
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Republic of Korea
| | - Min-Jeong Kim
- Department of Radiology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Republic of Korea
| | - Nan Young Kim
- Hallym Institute of Translational Genomics and Bioinformatics, Hallym University Medical Center, Anyang 14068, Republic of Korea
| | - Hyo Geun Choi
- Suseo Seoul E.N.T. Clinic and MD Analytics, 10, Bamgogae-ro 1-gil, Gangnam-gu, Seoul 06349, Republic of Korea
| | - Eun Soo Kim
- Department of Radiology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Republic of Korea
| |
Collapse
|
9
|
Hu J, Zhang D, Tian K, Ren C, Li H, Lin C, Huang X, Liu J, Mao W, Zhang J. Small-molecule LRRK2 inhibitors for PD therapy: Current achievements and future perspectives. Eur J Med Chem 2023; 256:115475. [PMID: 37201428 DOI: 10.1016/j.ejmech.2023.115475] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/08/2023] [Accepted: 05/08/2023] [Indexed: 05/20/2023]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a multifunctional protein that orchestrates a diverse array of cellular processes, including vesicle transport, autophagy, lysosome degradation, neurotransmission, and mitochondrial activity. Hyperactivation of LRRK2 triggers vesicle transport dysfunction, neuroinflammation, accumulation of α-synuclein, mitochondrial dysfunction, and the loss of cilia, ultimately leading to Parkinson's disease (PD). Therefore, targeting LRRK2 protein is a promising therapeutic strategy for PD. The clinical translation of LRRK2 inhibitors was historically impeded by issues surrounding tissue specificity. Recent studies have identified LRRK2 inhibitors that have no effect on peripheral tissues. Currently, there are four small-molecule LRRK2 inhibitors undergoing clinical trials. This review provides a summary of the structure and biological functions of LRRK2, along with an overview of the binding modes and structure-activity relationships (SARs) of small-molecule inhibitors targeting LRRK2. It offers valuable references for developing novel drugs targeting LRRK2.
Collapse
Affiliation(s)
- Jiarui Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dan Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Keyue Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Changyu Ren
- Chengdu Fifth People's Hospital, Chengdu, 611130, Sichuan, China
| | - Heng Li
- Chengdu Fifth People's Hospital, Chengdu, 611130, Sichuan, China
| | - Congcong Lin
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiaoli Huang
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Wuyu Mao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Jifa Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Gu YZ, Vlasakova K, Miller G, Gatto NT, Ciaccio PJ, Kuruvilla S, Besteman EG, Smith R, Reynolds SJ, Amin RP, Glaab WE, Wollenberg G, Lebron J, Sistare FD. Early-Onset albuminuria and Associated Renal Pathology in Leucine-Rich Repeat Kinase 2 Knockout Rats. Toxicol Pathol 2023; 51:15-26. [PMID: 37078689 DOI: 10.1177/01926233231162809] [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: 04/21/2023]
Abstract
Activating mutations of the leucine-rich repeat kinase 2 (LRRK2) gene are associated with Parkinson disease (PD), prompting development of LRRK2 inhibitors as potential treatment for PD. However, kidney safety concerns have surfaced from LRRK2 knockout (KO) mice and rats and from repeat-dose studies in rodents administered LRRK2 inhibitors. To support drug development of this therapeutic target, we conducted a study of 26 weeks' duration in 2-month-old wild-type and LRRK2 KO Long-Evans Hooded rats to systematically examine the performance of urinary safety biomarkers and to characterize the nature of the morphological changes in the kidneys by light microscopy and by ultrastructural evaluation. Our data reveal the time course of early-onset albuminuria at 3 and 4 months in LRRK2 KO female and male rats, respectively. The increases in urine albumin were not accompanied by concurrent increases in serum creatinine, blood urea nitrogen, or renal safety biomarkers such as kidney injury molecule 1 or clusterin, although morphological alterations in both glomerular and tubular structure were identified by light and transmission electron microscopy at 8 months of age. Diet optimization with controlled food intake attenuated the progression of albuminuria and associated renal changes.
Collapse
Affiliation(s)
- Yi-Zhong Gu
- Merck & Co., Inc., West Point, Pennsylvania, USA
| | | | - Glen Miller
- Merck & Co., Inc., West Point, Pennsylvania, USA
| | | | | | | | | | - Roger Smith
- Merck & Co., Inc., West Point, Pennsylvania, USA
| | | | | | | | | | - Jose Lebron
- Merck & Co., Inc., West Point, Pennsylvania, USA
| | | |
Collapse
|
11
|
Liu X, Kalogeropulou AF, Domingos S, Makukhin N, Nirujogi RS, Singh F, Shpiro N, Saalfrank A, Sammler E, Ganley IG, Moreira R, Alessi DR, Ciulli A. Discovery of XL01126: A Potent, Fast, Cooperative, Selective, Orally Bioavailable, and Blood-Brain Barrier Penetrant PROTAC Degrader of Leucine-Rich Repeat Kinase 2. J Am Chem Soc 2022; 144:16930-16952. [PMID: 36007011 PMCID: PMC9501899 DOI: 10.1021/jacs.2c05499] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Indexed: 12/20/2022]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is one of the most promising targets for Parkinson's disease. LRRK2-targeting strategies have primarily focused on type 1 kinase inhibitors, which, however, have limitations as the inhibited protein can interfere with natural mechanisms, which could lead to undesirable side effects. Herein, we report the development of LRRK2 proteolysis targeting chimeras (PROTACs), culminating in the discovery of degrader XL01126, as an alternative LRRK2-targeting strategy. Initial designs and screens of PROTACs based on ligands for E3 ligases von Hippel-Lindau (VHL), Cereblon (CRBN), and cellular inhibitor of apoptosis (cIAP) identified the best degraders containing thioether-conjugated VHL ligand VH101. A second round of medicinal chemistry exploration led to qualifying XL01126 as a fast and potent degrader of LRRK2 in multiple cell lines, with DC50 values within 15-72 nM, Dmax values ranging from 82 to 90%, and degradation half-lives spanning from 0.6 to 2.4 h. XL01126 exhibits high cell permeability and forms a positively cooperative ternary complex with VHL and LRRK2 (α = 5.7), which compensates for a substantial loss of binary binding affinities to VHL and LRRK2, underscoring its strong degradation performance in cells. Remarkably, XL01126 is orally bioavailable (F = 15%) and can penetrate the blood-brain barrier after either oral or parenteral dosing in mice. Taken together, these experiments qualify XL01126 as a suitable degrader probe to study the noncatalytic and scaffolding functions of LRRK2 in vitro and in vivo and offer an attractive starting point for future drug development.
Collapse
Affiliation(s)
- Xingui Liu
- Centre
for Targeted Protein Degradation, Division of Biological Chemistry
and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Alexia F. Kalogeropulou
- Medical
Research Council (MRC) Protein Phosphorylation and Ubiquitylation
Unit, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Sofia Domingos
- Centre
for Targeted Protein Degradation, Division of Biological Chemistry
and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Nikolai Makukhin
- Centre
for Targeted Protein Degradation, Division of Biological Chemistry
and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Raja S. Nirujogi
- Medical
Research Council (MRC) Protein Phosphorylation and Ubiquitylation
Unit, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Francois Singh
- Medical
Research Council (MRC) Protein Phosphorylation and Ubiquitylation
Unit, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Natalia Shpiro
- Medical
Research Council (MRC) Protein Phosphorylation and Ubiquitylation
Unit, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Anton Saalfrank
- Medical
Research Council (MRC) Protein Phosphorylation and Ubiquitylation
Unit, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Esther Sammler
- Medical
Research Council (MRC) Protein Phosphorylation and Ubiquitylation
Unit, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Ian G. Ganley
- Medical
Research Council (MRC) Protein Phosphorylation and Ubiquitylation
Unit, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Rui Moreira
- Research
Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Dario R. Alessi
- Medical
Research Council (MRC) Protein Phosphorylation and Ubiquitylation
Unit, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Alessio Ciulli
- Centre
for Targeted Protein Degradation, Division of Biological Chemistry
and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| |
Collapse
|
12
|
Smith LJ, Lee CY, Menozzi E, Schapira AHV. Genetic variations in GBA1 and LRRK2 genes: Biochemical and clinical consequences in Parkinson disease. Front Neurol 2022; 13:971252. [PMID: 36034282 PMCID: PMC9416236 DOI: 10.3389/fneur.2022.971252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/25/2022] [Indexed: 11/24/2022] Open
Abstract
Variants in the GBA1 and LRRK2 genes are the most common genetic risk factors associated with Parkinson disease (PD). Both genes are associated with lysosomal and autophagic pathways, with the GBA1 gene encoding for the lysosomal enzyme, glucocerebrosidase (GCase) and the LRRK2 gene encoding for the leucine-rich repeat kinase 2 enzyme. GBA1-associated PD is characterized by earlier age at onset and more severe non-motor symptoms compared to sporadic PD. Mutations in the GBA1 gene can be stratified into severe, mild and risk variants depending on the clinical presentation of disease. Both a loss- and gain- of function hypothesis has been proposed for GBA1 variants and the functional consequences associated with each variant is often linked to mutation severity. On the other hand, LRRK2-associated PD is similar to sporadic PD, but with a more benign disease course. Mutations in the LRRK2 gene occur in several structural domains and affect phosphorylation of GTPases. Biochemical studies suggest a possible convergence of GBA1 and LRRK2 pathways, with double mutant carriers showing a milder phenotype compared to GBA1-associated PD. This review compares GBA1 and LRRK2-associated PD, and highlights possible genotype-phenotype associations for GBA1 and LRRK2 separately, based on biochemical consequences of single variants.
Collapse
Affiliation(s)
- Laura J. Smith
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London (UCL), London, United Kingdom
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Chiao-Yin Lee
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London (UCL), London, United Kingdom
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Elisa Menozzi
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London (UCL), London, United Kingdom
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Anthony H. V. Schapira
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London (UCL), London, United Kingdom
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| |
Collapse
|
13
|
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
|
14
|
Kluss JH, Lewis PA, Greggio E. Leucine-rich repeat kinase 2 (LRRK2): an update on the potential therapeutic target for Parkinson's disease. Expert Opin Ther Targets 2022; 26:537-546. [PMID: 35642531 DOI: 10.1080/14728222.2022.2082937] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
AREAS COVERED In this review, we will provide an update on the current status of drugs and other technologies that have emerged in recent years and provide an overview of their efficacy in ameliorating LRRK2 kinase activity and overall safety in animal models and humans. EXPERT OPINION The growth of both target discovery and innovative drug design has sparked a lot of excitement for the future of how we treat Parkinson's disease. Given the immense focus on LRRK2 as a therapeutic target, it is expected within the next decade to determine its therapeutic properties, or lack thereof, for PD.
Collapse
Affiliation(s)
- Jillian H Kluss
- School of Pharmacy, University of Reading, Whiteknights, Reading, UK.,Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Patrick A Lewis
- School of Pharmacy, University of Reading, Whiteknights, Reading, UK.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.,Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Elisa Greggio
- Department of Biology, University of Padova, Padova, Italy.,Centro Studi per la Neurodegenerazione (CESNE), University of Padova, Padova, Italy
| |
Collapse
|
15
|
Rocha EM, Keeney MT, Di Maio R, De Miranda BR, Greenamyre JT. LRRK2 and idiopathic Parkinson's disease. Trends Neurosci 2022; 45:224-236. [PMID: 34991886 PMCID: PMC8854345 DOI: 10.1016/j.tins.2021.12.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/30/2021] [Accepted: 12/09/2021] [Indexed: 12/22/2022]
Abstract
The etiology of idiopathic Parkinson's disease (iPD) is multifactorial, and both genetics and environmental exposures are risk factors. While mutations in leucine-rich repeat kinase-2 (LRRK2) that are associated with increased kinase activity are the most common cause of autosomal dominant PD, the role of LRRK2 in iPD, independent of mutations, remains uncertain. In this review, we discuss how the architecture of LRRK2 influences kinase activation and how enhanced LRRK2 substrate phosphorylation might contribute to pathogenesis. We describe how oxidative stress and endolysosomal dysfunction, both of which occur in iPD, can activate non-mutated LRRK2 to a similar degree as pathogenic mutations. Similarly, environmental toxicants that are linked epidemiologically to iPD risk can also activate LRRK2. In aggregate, current evidence suggests an important role for LRRK2 in iPD.
Collapse
Affiliation(s)
- Emily M Rocha
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Matthew T Keeney
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Molecular Pharmacology Graduate Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Roberto Di Maio
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Briana R De Miranda
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Molecular Pharmacology Graduate Program, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
16
|
Singh RK, Soliman A, Guaitoli G, Störmer E, von Zweydorf F, Dal Maso T, Oun A, Van Rillaer L, Schmidt SH, Chatterjee D, David JA, Pardon E, Schwartz TU, Knapp S, Kennedy EJ, Steyaert J, Herberg FW, Kortholt A, Gloeckner CJ, Versées W. Nanobodies as allosteric modulators of Parkinson's disease-associated LRRK2. Proc Natl Acad Sci U S A 2022; 119:e2112712119. [PMID: 35217606 PMCID: PMC8892280 DOI: 10.1073/pnas.2112712119] [Citation(s) in RCA: 27] [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: 07/14/2021] [Accepted: 01/24/2022] [Indexed: 02/07/2023] Open
Abstract
Mutations in the gene coding for leucine-rich repeat kinase 2 (LRRK2) are a leading cause of the inherited form of Parkinson's disease (PD), while LRRK2 overactivation is also associated with the more common idiopathic form of PD. LRRK2 is a large multidomain protein, including a GTPase as well as a Ser/Thr protein kinase domain. Common, disease-causing mutations increase LRRK2 kinase activity, presenting LRRK2 as an attractive target for drug discovery. Currently, drug development has mainly focused on ATP-competitive kinase inhibitors. Here, we report the identification and characterization of a variety of nanobodies that bind to different LRRK2 domains and inhibit or activate LRRK2 in cells and in in vitro. Importantly, nanobodies were identified that inhibit LRRK2 kinase activity while binding to a site that is topographically distinct from the active site and thus act through an allosteric inhibitory mechanism that does not involve binding to the ATP pocket or even to the kinase domain. Moreover, while certain nanobodies completely inhibit the LRRK2 kinase activity, we also identified nanobodies that specifically inhibit the phosphorylation of Rab protein substrates. Finally, in contrast to current type I kinase inhibitors, the studied kinase-inhibitory nanobodies did not induce LRRK2 microtubule association. These comprehensively characterized nanobodies represent versatile tools to study the LRRK2 function and mechanism and can pave the way toward novel diagnostic and therapeutic strategies for PD.
Collapse
Affiliation(s)
- Ranjan K Singh
- VIB-VUB Center for Structural Biology, 1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Ahmed Soliman
- Department of Cell Biochemistry, University of Groningen, 9747AG Groningen, The Netherlands
| | | | - Eliza Störmer
- Department of Biochemistry, Institute for Biology, University of Kassel, 34132 Kassel, Germany
| | | | - Thomas Dal Maso
- VIB-VUB Center for Structural Biology, 1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Asmaa Oun
- Department of Cell Biochemistry, University of Groningen, 9747AG Groningen, The Netherlands
- Groningen Research Institute of Pharmacy, Molecular Pharmacology XB10, 9700AD Groningen, The Netherlands
| | - Laura Van Rillaer
- VIB-VUB Center for Structural Biology, 1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Sven H Schmidt
- Department of Biochemistry, Institute for Biology, University of Kassel, 34132 Kassel, Germany
| | - Deep Chatterjee
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt, 60438 Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, 60438 Frankfurt, Germany
| | - Joshua A David
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Els Pardon
- VIB-VUB Center for Structural Biology, 1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Thomas U Schwartz
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt, 60438 Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, 60438 Frankfurt, Germany
| | - Eileen J Kennedy
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602
| | - Jan Steyaert
- VIB-VUB Center for Structural Biology, 1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Friedrich W Herberg
- Department of Biochemistry, Institute for Biology, University of Kassel, 34132 Kassel, Germany
| | - Arjan Kortholt
- Department of Cell Biochemistry, University of Groningen, 9747AG Groningen, The Netherlands
| | - Christian Johannes Gloeckner
- German Center for Neurodegenerative Diseases, D-72076 Tübingen, Germany
- Core Facility for Medical Bioanalytics, Center for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany
| | - Wim Versées
- VIB-VUB Center for Structural Biology, 1050 Brussels, Belgium;
- Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| |
Collapse
|
17
|
Guadagnolo D, Piane M, Torrisi MR, Pizzuti A, Petrucci S. Genotype-Phenotype Correlations in Monogenic Parkinson Disease: A Review on Clinical and Molecular Findings. Front Neurol 2021; 12:648588. [PMID: 34630269 PMCID: PMC8494251 DOI: 10.3389/fneur.2021.648588] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/08/2021] [Indexed: 12/30/2022] Open
Abstract
Parkinson disease (PD) is a complex neurodegenerative disorder, usually with multifactorial etiology. It is characterized by prominent movement disorders and non-motor symptoms. Movement disorders commonly include bradykinesia, rigidity, and resting tremor. Non-motor symptoms can include behavior disorders, sleep disturbances, hyposmia, cognitive impairment, and depression. A fraction of PD cases instead is due to Parkinsonian conditions with Mendelian inheritance. The study of the genetic causes of these phenotypes has shed light onto common pathogenetic mechanisms underlying Parkinsonian conditions. Monogenic Parkinsonisms can present autosomal dominant, autosomal recessive, or even X-linked inheritance patterns. Clinical presentations vary from forms indistinguishable from idiopathic PD to severe childhood-onset conditions with other neurological signs. We provided a comprehensive description of each condition, discussing current knowledge on genotype-phenotype correlations. Despite the broad clinical spectrum and the many genes involved, the phenotype appears to be related to the disrupted cell function and inheritance pattern, and several assumptions about genotype-phenotype correlations can be made. The interest in these assumptions is not merely speculative, in the light of novel promising targeted therapies currently under development.
Collapse
Affiliation(s)
- Daniele Guadagnolo
- Department of Experimental Medicine, Policlinico Umberto i Hospital, Sapienza University of Rome, Rome, Italy
| | - Maria Piane
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Medical Genetics and Advanced Cell Diagnostics Unit, S. Andrea University Hospital, Rome, Italy
| | - Maria Rosaria Torrisi
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Medical Genetics and Advanced Cell Diagnostics Unit, S. Andrea University Hospital, Rome, Italy
| | - Antonio Pizzuti
- Department of Experimental Medicine, Policlinico Umberto i Hospital, Sapienza University of Rome, Rome, Italy
| | - Simona Petrucci
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Medical Genetics and Advanced Cell Diagnostics Unit, S. Andrea University Hospital, Rome, Italy
| |
Collapse
|
18
|
Senkevich K, Rudakou U, Gan-Or Z. New therapeutic approaches to Parkinson's disease targeting GBA, LRRK2 and Parkin. Neuropharmacology 2021; 202:108822. [PMID: 34626666 DOI: 10.1016/j.neuropharm.2021.108822] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 01/23/2023]
Abstract
Parkinson's disease (PD) is defined as a complex disorder with multifactorial pathogenesis, yet a more accurate definition could be that PD is not a single entity, but rather a mixture of different diseases with similar phenotypes. Attempts to classify subtypes of PD have been made based on clinical phenotypes or biomarkers. However, the most practical approach, at least for a portion of the patients, could be to classify patients based on genes involved in PD. GBA and LRRK2 mutations are the most common genetic causes or risk factors of PD, and PRKN is the most common cause of autosomal recessive form of PD. Patients carrying variants in GBA, LRRK2 or PRKN differ in some of their clinical characteristics, pathology and biochemical parameters. Thus, these three PD-associated genes are of special interest for drug development. Existing therapeutic approaches in PD are strictly symptomatic, as numerous clinical trials aimed at modifying PD progression or providing neuroprotection have failed over the last few decades. The lack of precision medicine approach in most of these trials could be one of the reasons why they were not successful. In the current review we discuss novel therapeutic approaches targeting GBA, LRRK2 and PRKN and discuss different aspects related to these genes and clinical trials.
Collapse
Affiliation(s)
- Konstantin Senkevich
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada; Department of Neurology and neurosurgery, McGill University, Montréal, QC, Canada; First Pavlov State Medical University of St. Petersburg, Saint-Petersburg, Russia
| | - Uladzislau Rudakou
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada; Department of Neurology and neurosurgery, McGill University, Montréal, QC, Canada; Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Ziv Gan-Or
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada; Department of Neurology and neurosurgery, McGill University, Montréal, QC, Canada; Department of Human Genetics, McGill University, Montréal, QC, Canada.
| |
Collapse
|
19
|
Zhang K, Zhu S, Li J, Jiang T, Feng L, Pei J, Wang G, Ouyang L, Liu B. Targeting autophagy using small-molecule compounds to improve potential therapy of Parkinson's disease. Acta Pharm Sin B 2021; 11:3015-3034. [PMID: 34729301 PMCID: PMC8546670 DOI: 10.1016/j.apsb.2021.02.016] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/28/2021] [Accepted: 02/19/2021] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD), known as one of the most universal neurodegenerative diseases, is a serious threat to the health of the elderly. The current treatment has been demonstrated to relieve symptoms, and the discovery of new small-molecule compounds has been regarded as a promising strategy. Of note, the homeostasis of the autolysosome pathway (ALP) is closely associated with PD, and impaired autophagy may cause the death of neurons and thereby accelerating the progress of PD. Thus, pharmacological targeting autophagy with small-molecule compounds has been drawn a rising attention so far. In this review, we focus on summarizing several autophagy-associated targets, such as AMPK, mTORC1, ULK1, IMPase, LRRK2, beclin-1, TFEB, GCase, ERRα, C-Abelson, and as well as their relevant small-molecule compounds in PD models, which will shed light on a clue on exploiting more potential targeted small-molecule drugs tracking PD treatment in the near future.
Collapse
Key Words
- 3-MA, 3-methyladenine
- 5-HT2A, Serotonin 2A
- 5-HT2C, serotonin 2C
- A2A, adenosine 2A
- AADC, aromatic amino acid decarboxylase
- ALP, autophagy-lysosomal pathway
- AMPK, 5ʹAMP-activated protein kinase
- ATG, autophagy related protein
- ATP13A2, ATPase cation transporting 13A2
- ATTEC, autophagosome-tethering compound
- AUC, the area under the curve
- AUTAC, autophagy targeting chimera
- Autophagy
- BAF, bafilomycinA1
- BBB, blood−brain barrier
- CL, clearance rate
- CMA, chaperone-mediated autophagy
- CNS, central nervous system
- COMT, catechol-O-methyltransferase
- DA, dopamine
- DAT, dopamine transporter
- DJ-1, Parkinson protein 7
- DR, dopamine receptor
- ER, endoplasmic reticulum
- ERRα, estrogen-related receptor alpha
- F, oral bioavailability
- GAPDH, glyceraldehyde 3-phosphate dehydrogenase
- GBA, glucocerebrosidase β acid
- GWAS, genome-wide association study
- HDAC6, histone deacetylase 6
- HSC70, heat shock cognate 71 kDa protein
- HSPA8, heat shock 70 kDa protein 8
- IMPase, inositol monophosphatase
- IPPase, inositol polyphosphate 1-phosphatase
- KI, knockin
- LAMP2A, lysosome-associated membrane protein 2 A
- LC3, light chain 3
- LIMP-2, lysosomal integrated membrane protein-2
- LRRK2, leucine-rich repeat sequence kinase 2
- LRS, leucyl-tRNA synthetase
- LUHMES, lund human mesencephalic
- Lamp2a, type 2A lysosomal-associated membrane protein
- MAO-B, monoamine oxidase B
- MPP+, 1-methyl-4-phenylpyridinium
- MPTP, 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine
- MYCBP2, MYC-binding protein 2
- NMDA, N-methyl-d-aspartic acid
- ONRs, orphan nuclear receptors
- PD therapy
- PD, Parkinson's disease
- PDE4, phosphodiesterase 4
- PI3K, phosphatidylinositol 3-kinase
- PI3P, phosphatidylinositol 3-phosphate
- PINK1, PTEN-induced kinase 1
- PLC, phospholipase C
- PREP, prolyl oligopeptidase
- Parkin, parkin RBR E3 ubiquitin−protein ligase
- Parkinson's disease (PD)
- ROS, reactive oxygen species
- SAR, structure–activity relationship
- SAS, solvent accessible surface
- SN, substantia nigra
- SNCA, α-synuclein gene
- SYT11, synaptotagmin 11
- Small-molecule compound
- TFEB, transcription factor EB
- TSC2, tuberous sclerosis complex 2
- Target
- ULK1, UNC-51-like kinase 1
- UPS, ubiquitin−proteasome system
- mAChR, muscarinic acetylcholine receptor
- mTOR, the mammalian target of rapamycin
- α-syn, α-synuclein
Collapse
|
20
|
Verma A, Ebanks K, Fok CY, Lewis PA, Bettencourt C, Bandopadhyay R. In silico comparative analysis of LRRK2 interactomes from brain, kidney and lung. Brain Res 2021; 1765:147503. [PMID: 33915162 PMCID: PMC8212912 DOI: 10.1016/j.brainres.2021.147503] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/06/2021] [Accepted: 04/23/2021] [Indexed: 01/11/2023]
Abstract
Mutations in LRRK2 are the most frequent cause of familial Parkinson's disease (PD), with common LRRK2 non-coding variants also acting as risk factors for idiopathic PD. Currently, therapeutic agents targeting LRRK2 are undergoing advanced clinical trials in humans, however, it is important to understand the wider implications of LRRK2 targeted treatments given that LRRK2 is expressed in diverse tissues including the brain, kidney and lungs. This presents challenges to treatment in terms of effects on peripheral organ functioning, thus, protein interactors of LRRK2 could be targeted in lieu to optimize therapeutic effects. Herein an in-silico analysis of LRRK2 direct interactors in brain tissue from various brain regionswas conducted along with a comparative analysis of the LRRK2 interactome in the brain, kidney, and lung tissues. This was carried out based on curated protein-protein interaction (PPI) data from protein interaction databases such as HIPPIE, human gene/protein expression databases and Gene ontology (GO) enrichment analysis using Bingo. Seven targets (MAP2K6, MATK, MAPT, PAK6, SH3GL2, CDC42EP3 and CHGB) were found to be viable objectives for LRRK2 based investigations for PD that would have minimal impact on optimal functioning within peripheral organs. Specifically, MAPT, CHGB, PAK6, and SH3GL2 interacted with LRRK2 in the brain and kidney but not in lung tissue whilst LRRK2-MAP2K6 interacted only in the cerebellum and MATK-LRRK2 interaction was absent in kidney tissues. CDC42EP3 expression levels were low in brain tissues compared to kidney/lung. The results of this computational analysis suggest new avenues for experimental investigations towards LRRK2-targeted therapeutics.
Collapse
Affiliation(s)
- Amrita Verma
- Reta Lila Weston Institute of Neurological Studies, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom
| | - Kirsten Ebanks
- Reta Lila Weston Institute of Neurological Studies, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom
| | - Chi-Yee Fok
- Reta Lila Weston Institute of Neurological Studies, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom
| | - Patrick A Lewis
- Royal Veterinary College, Royal College Street, London NW10TV, United Kingdom; Department of Neurodegenerative Disease and Queen Square Brain Bank, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Conceicao Bettencourt
- Department of Neurodegenerative Disease and Queen Square Brain Bank, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom
| | - Rina Bandopadhyay
- Reta Lila Weston Institute of Neurological Studies, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London WC1N 1PJ, United Kingdom.
| |
Collapse
|
21
|
Araki M, Ito K, Takatori S, Ito G, Tomita T. BORCS6 is involved in the enlargement of lung lamellar bodies in Lrrk2 knockout mice. Hum Mol Genet 2021; 30:1618-1631. [PMID: 34077533 DOI: 10.1093/hmg/ddab146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) has been implicated in the pathogenesis of Parkinson disease. It has been shown that Lrrk2 knockout (KO) rodents have enlarged lamellar bodies (LBs) in their alveolar epithelial type II cells, although the underlying mechanisms remain unclear. Here we performed proteomic analyses on LBs isolated from Lrrk2 KO mice and found that the LB proteome is substantially different in Lrrk2 KO mice compared with wild-type mice. In Lrrk2 KO LBs, several Rab proteins were increased, and subunit proteins of BLOC-1-related complex (BORC) were decreased. The amount of surfactant protein C was significantly decreased in the bronchoalveolar lavage fluid obtained from Lrrk2 KO mice, suggesting that LB exocytosis is impaired in Lrrk2 KO mice. We also found that the enlargement of LBs is recapitulated in A549 cells upon KO of LRRK2 or by treating cells with LRRK2 inhibitors. Using this model, we show that KO of BORCS6, a BORC subunit gene, but not other BORC genes, causes LB enlargement. Our findings implicate the LRRK2-BORCS6 pathway in the maintenance of LB morphology.
Collapse
Affiliation(s)
- Miho Araki
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Kyohei Ito
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Sho Takatori
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Genta Ito
- Social Cooperation Program of Brain and Neurological Disorders, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
- Social Cooperation Program of Brain and Neurological Disorders, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| |
Collapse
|
22
|
Allosteric inhibition of LRRK2, where are we now. Biochem Soc Trans 2021; 48:2185-2194. [PMID: 33079169 PMCID: PMC7609032 DOI: 10.1042/bst20200424] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 12/17/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. In recent years, it has been shown that leucine-rich repeat kinase 2 (LRRK2) has a crucial function in both familial and sporadic forms of PD. LRRK2 pathogenic mutations are thought to result in an increase in LRRK2 kinase activity. Thus, inhibiting LRRK2 kinase activity has become a main therapeutic target. Many compounds capable of inhibiting LRRK2 kinase activity with high selectivity and brain availability have been described. However, the safety of long-term use of these ATP-competitive LRRK2 kinase inhibitors has been challenged by several studies. Therefore, alternative ways of targeting LRRK2 activity will have a great benefit. In this review, we discuss the recent progress in the development of allosteric inhibitors of LRRK2, mainly via interfering with GTPase activity, and propose potential new intra and interprotein interactions targets that can lead to open doors toward new therapeutics.
Collapse
|
23
|
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
|
24
|
Day JO, Mullin S. The Genetics of Parkinson's Disease and Implications for Clinical Practice. Genes (Basel) 2021; 12:genes12071006. [PMID: 34208795 PMCID: PMC8304082 DOI: 10.3390/genes12071006] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/21/2021] [Accepted: 06/28/2021] [Indexed: 12/17/2022] Open
Abstract
The genetic landscape of Parkinson’s disease (PD) is characterised by rare high penetrance pathogenic variants causing familial disease, genetic risk factor variants driving PD risk in a significant minority in PD cases and high frequency, low penetrance variants, which contribute a small increase of the risk of developing sporadic PD. This knowledge has the potential to have a major impact in the clinical care of people with PD. We summarise these genetic influences and discuss the implications for therapeutics and clinical trial design.
Collapse
Affiliation(s)
- Jacob Oliver Day
- Faculty of Health, University of Plymouth, Plymouth PL4 8AA, UK;
| | - Stephen Mullin
- Faculty of Health, University of Plymouth, Plymouth PL4 8AA, UK;
- Department of Clinical and Movement Neurosciences, University College London Institute of Neurology, London WC1N 3BG, UK
- Correspondence:
| |
Collapse
|
25
|
Baptista MAS, Merchant K, Barrett T, Bhargava S, Bryce DK, Ellis JM, Estrada AA, Fell MJ, Fiske BK, Fuji RN, Galatsis P, Henry AG, Hill S, Hirst W, Houle C, Kennedy ME, Liu X, Maddess ML, Markgraf C, Mei H, Meier WA, Needle E, Ploch S, Royer C, Rudolph K, Sharma AK, Stepan A, Steyn S, Trost C, Yin Z, Yu H, Wang X, Sherer TB. LRRK2 inhibitors induce reversible changes in nonhuman primate lungs without measurable pulmonary deficits. Sci Transl Med 2021; 12:12/540/eaav0820. [PMID: 32321864 DOI: 10.1126/scitranslmed.aav0820] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 07/22/2019] [Indexed: 12/27/2022]
Abstract
The kinase-activating mutation G2019S in leucine-rich repeat kinase 2 (LRRK2) is one of the most common genetic causes of Parkinson's disease (PD) and has spurred development of LRRK2 inhibitors. Preclinical studies have raised concerns about the safety of LRRK2 inhibitors due to histopathological changes in the lungs of nonhuman primates treated with two of these compounds. Here, we investigated whether these lung effects represented on-target pharmacology and whether they were reversible after drug withdrawal in macaques. We also examined whether treatment was associated with pulmonary function deficits. We conducted a 2-week repeat-dose toxicology study in macaques comparing three different LRRK2 inhibitors: GNE-7915 (30 mg/kg, twice daily as a positive control), MLi-2 (15 and 50 mg/kg, once daily), and PFE-360 (3 and 6 mg/kg, once daily). Subsets of animals dosed with GNE-7915 or MLi-2 were evaluated 2 weeks after drug withdrawal for lung function. All compounds induced mild cytoplasmic vacuolation of type II lung pneumocytes without signs of lung degeneration, implicating on-target pharmacology. At low doses of PFE-360 or MLi-2, there was ~50 or 100% LRRK2 inhibition in brain tissue, respectively, but histopathological lung changes were either absent or minimal. The lung effect was reversible after dosing ceased. Lung function tests demonstrated that the histological changes in lung tissue induced by MLi-2 and GNE-7915 did not result in pulmonary deficits. Our results suggest that the observed lung effects in nonhuman primates in response to LRRK2 inhibitors should not preclude clinical testing of these compounds for PD.
Collapse
Affiliation(s)
- Marco A S Baptista
- The Michael J. Fox Foundation for Parkinson's Research, Grand Central Station, P.O. Box 4777, New York, NY 10001, USA.
| | - Kalpana Merchant
- Northwestern University School of Medicine, Chicago, IL 60611, USA
| | - Ted Barrett
- Lovelace Respiratory Research Institute, Albuquerque, NM 87108, USA
| | - Sakshi Bhargava
- Pfizer Inc., Neuroscience Research Unit, Cambridge, MA 02139, USA
| | - Dianne K Bryce
- Merck Research Laboratories, Early Discovery Neuroscience, Boston, MA 02115, USA
| | - J Michael Ellis
- Merck Research Laboratories, Early Discovery Neuroscience, Boston, MA 02115, USA
| | | | - Matthew J Fell
- Merck Research Laboratories, Early Discovery Neuroscience, Boston, MA 02115, USA
| | - Brian K Fiske
- The Michael J. Fox Foundation for Parkinson's Research, Grand Central Station, P.O. Box 4777, New York, NY 10001, USA
| | - Reina N Fuji
- Genentech, Inc., South San Francisco, CA 94080, USA
| | - Paul Galatsis
- Pfizer Inc., Neuroscience Research Unit, Cambridge, MA 02139, USA
| | | | - Sue Hill
- Merck Research Laboratories, Early Discovery Neuroscience, Boston, MA 02115, USA
| | - Warren Hirst
- Pfizer Inc., Neuroscience Research Unit, Cambridge, MA 02139, USA
| | | | - Matthew E Kennedy
- Merck Research Laboratories, Early Discovery Neuroscience, Boston, MA 02115, USA
| | - Xingrong Liu
- Genentech, Inc., South San Francisco, CA 94080, USA
| | - Matthew L Maddess
- Merck Research Laboratories, Early Discovery Neuroscience, Boston, MA 02115, USA
| | - Carrie Markgraf
- Merck Research Laboratories, Early Discovery Neuroscience, Boston, MA 02115, USA
| | - Hong Mei
- Merck Research Laboratories, Early Discovery Neuroscience, Boston, MA 02115, USA
| | | | - Elie Needle
- Pfizer Inc., Neuroscience Research Unit, Cambridge, MA 02139, USA
| | | | | | - Karin Rudolph
- Lovelace Respiratory Research Institute, Albuquerque, NM 87108, USA
| | | | - Antonia Stepan
- Pfizer Inc., Neuroscience Research Unit, Cambridge, MA 02139, USA
| | - Stefan Steyn
- Pfizer Inc., Neuroscience Research Unit, Cambridge, MA 02139, USA
| | - Craig Trost
- Covance Laboratories, Inc., Madison, WI 53704, USA
| | - Zhizhang Yin
- Merck Research Laboratories, Early Discovery Neuroscience, Boston, MA 02115, USA
| | - Hongshi Yu
- Merck Research Laboratories, Early Discovery Neuroscience, Boston, MA 02115, USA
| | - Xiang Wang
- Denali Therapeutics, South San Francisco, CA 94080, USA
| | - Todd B Sherer
- The Michael J. Fox Foundation for Parkinson's Research, Grand Central Station, P.O. Box 4777, New York, NY 10001, USA
| |
Collapse
|
26
|
Bryce DK, Ware CM, Woodhouse JD, Ciaccio PJ, Ellis JM, Hegde LG, Kuruvilla S, Maddess ML, Markgraf CG, Otte KM, Poulet FM, Timmins LM, Kennedy ME, Fell MJ. Characterization of the Onset, Progression, and Reversibility of Morphological Changes in Mouse Lung after Pharmacological Inhibition of Leucine-Rich Kinase 2 Kinase Activity. J Pharmacol Exp Ther 2021; 377:11-19. [PMID: 33509901 DOI: 10.1124/jpet.120.000217] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 01/20/2021] [Indexed: 11/22/2022] Open
Abstract
Gain-of-function mutations in leucine-rich kinase 2 (LRRK2) are associated with increased incidence of Parkinson disease (PD); thus, pharmacological inhibition of LRRK2 kinase activity is postulated as a disease-modifying treatment of PD. Histomorphological changes in lungs of nonhuman primates (NHPs) treated with small-molecule LRRK2 kinase inhibitors have brought the safety of this treatment approach into question. Although it remains unclear how LRRK2 kinase inhibition affects the lung, continued studies in NHPs prove to be both cost- and resource-prohibitive. To develop a tractable alternative animal model platform, we dosed male mice in-diet with the potent, highly selective LRRK2 kinase inhibitor MLi-2 and induced histomorphological changes in lung within 1 week. Oral bolus dosing of MLi-2 at a frequency modeled to provide steady-state exposure equivalent to that achieved with in-diet dosing induced type II pneumocyte vacuolation, suggesting pulmonary changes require sustained LRRK2 kinase inhibition. Treating mice with MLi-2 in-diet for up to 6 months resulted in type II pneumocyte vacuolation that progressed only modestly over time and was fully reversible after withdrawal of MLi-2. Immunohistochemical analysis of lung revealed a significant increase in prosurfactant protein C staining within type II pneumocytes. In the present study, we demonstrated the kinetics for onset, progression, and rapid reversibility of chronic LRRK2 kinase inhibitor effects on lung histomorphology in rodents and provide further evidence for the derisking of safety and tolerability concerns for chronic LRRK2 kinase inhibition in PD. SIGNIFICANCE STATEMENT: We have defined a mouse model by which the on-target lung effects of leucine-rich kinase 2 (LRRK2) kinase inhibition can be monitored, whereas previous in vivo testing relied solely on nonhuman primates. Data serve to derisk long-term treatment with LRRK2 kinase inhibitors, as all lung changes were mild and readily reversible.
Collapse
Affiliation(s)
- Dianne K Bryce
- Merck & Co., Inc., Kenilworth, New Jersey; Discovery Neuroscience (D.K.B., C.M.W., C.G.M., F.M.P., L.M.T., M.E.K., M.J.F.), Pharmacology (J.D.W., L.G.H.), Safety Assessment and Laboratory Animal Resources (P.J.C.), Discovery Chemistry (M.L.M.), and PPDM (K.M.O.), Merck & Co., Inc., Boston, Massachusetts; and Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania (S.K., C.G.M., F.M.P.)
| | - Chris M Ware
- Merck & Co., Inc., Kenilworth, New Jersey; Discovery Neuroscience (D.K.B., C.M.W., C.G.M., F.M.P., L.M.T., M.E.K., M.J.F.), Pharmacology (J.D.W., L.G.H.), Safety Assessment and Laboratory Animal Resources (P.J.C.), Discovery Chemistry (M.L.M.), and PPDM (K.M.O.), Merck & Co., Inc., Boston, Massachusetts; and Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania (S.K., C.G.M., F.M.P.)
| | - Janice D Woodhouse
- Merck & Co., Inc., Kenilworth, New Jersey; Discovery Neuroscience (D.K.B., C.M.W., C.G.M., F.M.P., L.M.T., M.E.K., M.J.F.), Pharmacology (J.D.W., L.G.H.), Safety Assessment and Laboratory Animal Resources (P.J.C.), Discovery Chemistry (M.L.M.), and PPDM (K.M.O.), Merck & Co., Inc., Boston, Massachusetts; and Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania (S.K., C.G.M., F.M.P.)
| | - Paul J Ciaccio
- Merck & Co., Inc., Kenilworth, New Jersey; Discovery Neuroscience (D.K.B., C.M.W., C.G.M., F.M.P., L.M.T., M.E.K., M.J.F.), Pharmacology (J.D.W., L.G.H.), Safety Assessment and Laboratory Animal Resources (P.J.C.), Discovery Chemistry (M.L.M.), and PPDM (K.M.O.), Merck & Co., Inc., Boston, Massachusetts; and Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania (S.K., C.G.M., F.M.P.)
| | - J Michael Ellis
- Merck & Co., Inc., Kenilworth, New Jersey; Discovery Neuroscience (D.K.B., C.M.W., C.G.M., F.M.P., L.M.T., M.E.K., M.J.F.), Pharmacology (J.D.W., L.G.H.), Safety Assessment and Laboratory Animal Resources (P.J.C.), Discovery Chemistry (M.L.M.), and PPDM (K.M.O.), Merck & Co., Inc., Boston, Massachusetts; and Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania (S.K., C.G.M., F.M.P.)
| | - Laxminarayan G Hegde
- Merck & Co., Inc., Kenilworth, New Jersey; Discovery Neuroscience (D.K.B., C.M.W., C.G.M., F.M.P., L.M.T., M.E.K., M.J.F.), Pharmacology (J.D.W., L.G.H.), Safety Assessment and Laboratory Animal Resources (P.J.C.), Discovery Chemistry (M.L.M.), and PPDM (K.M.O.), Merck & Co., Inc., Boston, Massachusetts; and Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania (S.K., C.G.M., F.M.P.)
| | - Sabu Kuruvilla
- Merck & Co., Inc., Kenilworth, New Jersey; Discovery Neuroscience (D.K.B., C.M.W., C.G.M., F.M.P., L.M.T., M.E.K., M.J.F.), Pharmacology (J.D.W., L.G.H.), Safety Assessment and Laboratory Animal Resources (P.J.C.), Discovery Chemistry (M.L.M.), and PPDM (K.M.O.), Merck & Co., Inc., Boston, Massachusetts; and Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania (S.K., C.G.M., F.M.P.)
| | - Matthew L Maddess
- Merck & Co., Inc., Kenilworth, New Jersey; Discovery Neuroscience (D.K.B., C.M.W., C.G.M., F.M.P., L.M.T., M.E.K., M.J.F.), Pharmacology (J.D.W., L.G.H.), Safety Assessment and Laboratory Animal Resources (P.J.C.), Discovery Chemistry (M.L.M.), and PPDM (K.M.O.), Merck & Co., Inc., Boston, Massachusetts; and Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania (S.K., C.G.M., F.M.P.)
| | - Carrie G Markgraf
- Merck & Co., Inc., Kenilworth, New Jersey; Discovery Neuroscience (D.K.B., C.M.W., C.G.M., F.M.P., L.M.T., M.E.K., M.J.F.), Pharmacology (J.D.W., L.G.H.), Safety Assessment and Laboratory Animal Resources (P.J.C.), Discovery Chemistry (M.L.M.), and PPDM (K.M.O.), Merck & Co., Inc., Boston, Massachusetts; and Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania (S.K., C.G.M., F.M.P.)
| | - Karin M Otte
- Merck & Co., Inc., Kenilworth, New Jersey; Discovery Neuroscience (D.K.B., C.M.W., C.G.M., F.M.P., L.M.T., M.E.K., M.J.F.), Pharmacology (J.D.W., L.G.H.), Safety Assessment and Laboratory Animal Resources (P.J.C.), Discovery Chemistry (M.L.M.), and PPDM (K.M.O.), Merck & Co., Inc., Boston, Massachusetts; and Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania (S.K., C.G.M., F.M.P.)
| | - Frederique M Poulet
- Merck & Co., Inc., Kenilworth, New Jersey; Discovery Neuroscience (D.K.B., C.M.W., C.G.M., F.M.P., L.M.T., M.E.K., M.J.F.), Pharmacology (J.D.W., L.G.H.), Safety Assessment and Laboratory Animal Resources (P.J.C.), Discovery Chemistry (M.L.M.), and PPDM (K.M.O.), Merck & Co., Inc., Boston, Massachusetts; and Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania (S.K., C.G.M., F.M.P.)
| | - Lauren M Timmins
- Merck & Co., Inc., Kenilworth, New Jersey; Discovery Neuroscience (D.K.B., C.M.W., C.G.M., F.M.P., L.M.T., M.E.K., M.J.F.), Pharmacology (J.D.W., L.G.H.), Safety Assessment and Laboratory Animal Resources (P.J.C.), Discovery Chemistry (M.L.M.), and PPDM (K.M.O.), Merck & Co., Inc., Boston, Massachusetts; and Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania (S.K., C.G.M., F.M.P.)
| | - Matthew E Kennedy
- Merck & Co., Inc., Kenilworth, New Jersey; Discovery Neuroscience (D.K.B., C.M.W., C.G.M., F.M.P., L.M.T., M.E.K., M.J.F.), Pharmacology (J.D.W., L.G.H.), Safety Assessment and Laboratory Animal Resources (P.J.C.), Discovery Chemistry (M.L.M.), and PPDM (K.M.O.), Merck & Co., Inc., Boston, Massachusetts; and Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania (S.K., C.G.M., F.M.P.)
| | - Matthew J Fell
- Merck & Co., Inc., Kenilworth, New Jersey; Discovery Neuroscience (D.K.B., C.M.W., C.G.M., F.M.P., L.M.T., M.E.K., M.J.F.), Pharmacology (J.D.W., L.G.H.), Safety Assessment and Laboratory Animal Resources (P.J.C.), Discovery Chemistry (M.L.M.), and PPDM (K.M.O.), Merck & Co., Inc., Boston, Massachusetts; and Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania (S.K., C.G.M., F.M.P.)
| |
Collapse
|
27
|
Kluss JH, Mazza MC, Li Y, Manzoni C, Lewis PA, Cookson MR, Mamais A. Preclinical modeling of chronic inhibition of the Parkinson's disease associated kinase LRRK2 reveals altered function of the endolysosomal system in vivo. Mol Neurodegener 2021; 16:17. [PMID: 33741046 PMCID: PMC7977595 DOI: 10.1186/s13024-021-00441-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/04/2021] [Indexed: 12/26/2022] Open
Abstract
The most common mutation in the Leucine-rich repeat kinase 2 gene (LRRK2), G2019S, causes familial Parkinson's Disease (PD) and renders the encoded protein kinase hyperactive. While targeting LRRK2 activity is currently being tested in clinical trials as a therapeutic avenue for PD, to date, the molecular effects of chronic LRRK2 inhibition have not yet been examined in vivo. We evaluated the utility of newly available phospho-antibodies for Rab substrates and LRRK2 autophosphorylation to examine the pharmacodynamic response to treatment with the potent and specific LRRK2 inhibitor, MLi-2, in brain and peripheral tissue in G2019S LRRK2 knock-in mice. We report higher sensitivity of LRRK2 autophosphorylation to MLi-2 treatment and slower recovery in washout conditions compared to Rab GTPases phosphorylation, and we identify pS106 Rab12 as a robust readout of downstream LRRK2 activity across tissues. The downstream effects of long-term chronic LRRK2 inhibition in vivo were evaluated in G2019S LRRK2 knock-in mice by phospho- and total proteomic analyses following an in-diet administration of MLi-2 for 10 weeks. We observed significant alterations in endolysosomal and trafficking pathways in the kidney that were sensitive to MLi-2 treatment and were validated biochemically. Furthermore, a subtle but distinct biochemical signature affecting mitochondrial proteins was observed in brain tissue in the same animals that, again, was reverted by kinase inhibition. Proteomic analysis in the lung did not detect any major pathway of dysregulation that would be indicative of pulmonary impairment. This is the first study to examine the molecular underpinnings of chronic LRRK2 inhibition in a preclinical in vivo PD model and highlights cellular processes that may be influenced by therapeutic strategies aimed at restoring LRRK2 physiological activity in PD patients.
Collapse
Affiliation(s)
- Jillian H Kluss
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.,School of Pharmacy, University of Reading, Whiteknights Campus, Reading, UK
| | - Melissa Conti Mazza
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Yan Li
- Proteomic Core Facility, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Claudia Manzoni
- School of Pharmacy, University of Reading, Whiteknights Campus, Reading, UK.,UCL School of Pharmacy, Brunswick Square, London, UK
| | - Patrick A Lewis
- School of Pharmacy, University of Reading, Whiteknights Campus, Reading, UK.,Royal Veterinary College, Royal College Street, London, UK.,Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
| | - Adamantios Mamais
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
28
|
Kelly K, Chang A, Hastings L, Abdelmotilib H, West AB. Genetic background influences LRRK2-mediated Rab phosphorylation in the rat brain. Brain Res 2021; 1759:147372. [PMID: 33600829 DOI: 10.1016/j.brainres.2021.147372] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 01/11/2021] [Accepted: 02/09/2021] [Indexed: 11/16/2022]
Abstract
Pathogenic missense mutations in the leucine-rich repeat kinase 2 gene, encoding LRRK2, results in the upregulation of Rab10 and Rab12 phosphorylation in different cells and tissues. Here, we evaluate levels of the LRRK2 kinase substrates pT73-Rab10 and pS106-Rab12 proteins in rat brain tissues from different genetic backgrounds. Whereas lines of Sprague Dawley rats have equivalent levels of pT73-Rab10 and pS106-Rab12 similar to Lrrk2 knockout rats, Long-Evans rats have levels of pT73-Rab10 and pS106-Rab12 comparable to G2019S-LRRK2 BAC transgenic rats. Strong LRRK2 kinase inhibitors are ineffective at reducing pT73-Rab10 and pS106-Rab12 levels in the Sprague Dawley rats, but potently reduce pT73-Rab10 and pS106-Rab12 levels in Long-Evans rats. Oral administration of the PFE-360 LRRK2 kinase inhibitor fails to provide neuroprotection from dopaminergic neurodegeneration caused by rAAV2/1-mediated overexpression of A53T-αsynuclein in Sprague Dawley rats. These results highlight substantial differences in LRRK2-mediated Rab10 and Rab12 phosphorylation in commonly utilized rat genetic backgrounds and suggest LRRK2 may not play a central role in Rab phosphorylation or mutant αsynuclein toxicity in Sprague Dawley rats.
Collapse
Affiliation(s)
- Kaela Kelly
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Allison Chang
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Lyndsay Hastings
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Hisham Abdelmotilib
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Andrew B West
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
| |
Collapse
|
29
|
Garofalo AW, Bright J, De Lombaert S, Toda AMA, Zobel K, Andreotti D, Beato C, Bernardi S, Budassi F, Caberlotto L, Gao P, Griffante C, Liu X, Mengatto L, Migliore M, Sabbatini FM, Sava A, Serra E, Vincetti P, Zhang M, Carlisle HJ. Selective Inhibitors of G2019S-LRRK2 Kinase Activity. J Med Chem 2020; 63:14821-14839. [PMID: 33197196 DOI: 10.1021/acs.jmedchem.0c01243] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pathogenic variants in the leucine-rich repeat kinase 2 (LRRK2) gene have been identified that increase the risk for developing Parkinson's disease in a dominantly inherited fashion. These pathogenic variants, of which G2019S is the most common, cause abnormally high kinase activity, and compounds that inhibit this activity are being pursued as potentially disease-modifying therapeutics. Because LRRK2 regulates important cellular processes, developing inhibitors that can selectively target the pathogenic variant while sparing normal LRRK2 activity could offer potential advantages in heterozygous carriers. We conducted a high-throughput screen and identified a single selective compound that preferentially inhibited G2019S-LRRK2. Optimization of this scaffold led to a series of novel, potent, and highly selective G2019S-LRRK2 inhibitors.
Collapse
Affiliation(s)
| | - Jessica Bright
- ESCAPE Bio, South San Francisco, California 94080, United States
| | | | - Alyssa M A Toda
- ESCAPE Bio, South San Francisco, California 94080, United States
| | - Kerry Zobel
- ESCAPE Bio, South San Francisco, California 94080, United States
| | | | | | | | | | | | - Peng Gao
- WuXi AppTec, Tianjin 300456, P. R. China
| | | | | | | | | | | | - Anna Sava
- Aptuit, an Evotec Company, Verona 37135, Italy
| | - Elena Serra
- Aptuit, an Evotec Company, Verona 37135, Italy
| | | | | | - Holly J Carlisle
- ESCAPE Bio, South San Francisco, California 94080, United States
| |
Collapse
|
30
|
Kelly K, West AB. Pharmacodynamic Biomarkers for Emerging LRRK2 Therapeutics. Front Neurosci 2020; 14:807. [PMID: 32903744 PMCID: PMC7438883 DOI: 10.3389/fnins.2020.00807] [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: 05/29/2020] [Accepted: 07/10/2020] [Indexed: 12/22/2022] Open
Abstract
Genetic studies have identified variants in the LRRK2 gene as important components of Parkinson's disease (PD) pathobiology. Biochemical and emergent biomarker studies have coalesced around LRRK2 hyperactivation in disease. Therapeutics that diminish LRRK2 activity, either with small molecule kinase inhibitors or anti-sense oligonucleotides, have recently advanced to the clinic. Historically, there have been few successes in the development of therapies that might slow or halt the progression of neurodegenerative diseases. Over the past few decades of biomedical research, retrospective analyses suggest the broad integration of informative biomarkers early in development tends to distinguish successful pipelines from those that fail early. Herein, we discuss the biomarker regulatory process, emerging LRRK2 biomarker candidates, assays, underlying biomarker biology, and clinical integration.
Collapse
Affiliation(s)
- Kaela Kelly
- Duke Center for Neurodegeneration Research, Departments of Pharmacology and Cancer Biology, Neurology, and Neurobiology, Duke University, Durham, NC, United States
| | - Andrew B West
- Duke Center for Neurodegeneration Research, Departments of Pharmacology and Cancer Biology, Neurology, and Neurobiology, Duke University, Durham, NC, United States
| |
Collapse
|
31
|
Dopaminergic neurodegeneration induced by Parkinson's disease-linked G2019S LRRK2 is dependent on kinase and GTPase activity. Proc Natl Acad Sci U S A 2020; 117:17296-17307. [PMID: 32631998 PMCID: PMC7382283 DOI: 10.1073/pnas.1922184117] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common cause of late-onset, autosomal-dominant familial Parkinson's disease (PD). LRRK2 functions as both a kinase and GTPase, and PD-linked mutations are known to influence both enzymatic activities. While PD-linked LRRK2 mutations can commonly induce neuronal damage in culture models, the mechanisms underlying these pathogenic effects remain uncertain. Rodent models containing familial LRRK2 mutations often lack robust PD-like neurodegenerative phenotypes. Here, we develop a robust preclinical model of PD in adult rats induced by the brain delivery of recombinant adenoviral vectors with neuronal-specific expression of human LRRK2 harboring the most common G2019S mutation. In this model, G2019S LRRK2 induces the robust degeneration of substantia nigra dopaminergic neurons, a pathological hallmark of PD. Introduction of a stable kinase-inactive mutation or administration of the selective kinase inhibitor, PF-360, attenuates neurodegeneration induced by G2019S LRRK2. Neuroprotection provided by pharmacological kinase inhibition is mediated by an unusual mechanism involving the robust destabilization of human LRRK2 protein in the brain relative to endogenous LRRK2. Our study further demonstrates that G2019S LRRK2-induced dopaminergic neurodegeneration critically requires normal GTPase activity, as hypothesis-testing mutations that increase GTP hydrolysis or impair GTP-binding activity provide neuroprotection although via distinct mechanisms. Taken together, our data demonstrate that G2019S LRRK2 induces neurodegeneration in vivo via a mechanism that is dependent on kinase and GTPase activity. Our study provides a robust rodent preclinical model of LRRK2-linked PD and nominates kinase inhibition and modulation of GTPase activity as promising disease-modifying therapeutic targets.
Collapse
|
32
|
Toffoli M, Vieira SRL, Schapira AHV. Genetic causes of PD: A pathway to disease modification. Neuropharmacology 2020; 170:108022. [PMID: 32119885 DOI: 10.1016/j.neuropharm.2020.108022] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/17/2020] [Accepted: 02/26/2020] [Indexed: 01/08/2023]
Abstract
The underline neuropathology of Parkinson disease is pleiomorphic and its genetic background diverse. Possibly because of this heterogeneity, no effective disease modifying therapy is available. In this paper we give an overview of the genetics of Parkinson disease and explain how this is relevant for the development of new therapies. This article is part of the special issue entitled 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- M Toffoli
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, United Kingdom
| | - S R L Vieira
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, United Kingdom
| | - A H V Schapira
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, United Kingdom.
| |
Collapse
|
33
|
Abstract
Human genetic variants predicted to cause loss-of-function of protein-coding genes (pLoF variants) provide natural in vivo models of human gene inactivation and can be valuable indicators of gene function and the potential toxicity of therapeutic inhibitors targeting these genes1,2. Gain-of-kinase-function variants in LRRK2 are known to significantly increase the risk of Parkinson's disease3,4, suggesting that inhibition of LRRK2 kinase activity is a promising therapeutic strategy. While preclinical studies in model organisms have raised some on-target toxicity concerns5-8, the biological consequences of LRRK2 inhibition have not been well characterized in humans. Here, we systematically analyze pLoF variants in LRRK2 observed across 141,456 individuals sequenced in the Genome Aggregation Database (gnomAD)9, 49,960 exome-sequenced individuals from the UK Biobank and over 4 million participants in the 23andMe genotyped dataset. After stringent variant curation, we identify 1,455 individuals with high-confidence pLoF variants in LRRK2. Experimental validation of three variants, combined with previous work10, confirmed reduced protein levels in 82.5% of our cohort. We show that heterozygous pLoF variants in LRRK2 reduce LRRK2 protein levels but that these are not strongly associated with any specific phenotype or disease state. Our results demonstrate the value of large-scale genomic databases and phenotyping of human loss-of-function carriers for target validation in drug discovery.
Collapse
|
34
|
Cunningham LA, Moore DJ. Endosomal sorting pathways in the pathogenesis of Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2020; 252:271-306. [PMID: 32247367 PMCID: PMC7206894 DOI: 10.1016/bs.pbr.2020.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The identification of Parkinson's disease (PD)-associated genes has created a powerful platform to begin to understand and nominate pathophysiological disease mechanisms. Herein, we discuss the genetic and experimental evidence supporting endolysosomal dysfunction as a major pathway implicated in PD. Well-studied familial PD-linked gene products, including LRRK2, VPS35, and α-synuclein, demonstrate how disruption of different aspects of endolysosomal sorting pathways by disease-causing mutations may manifest into PD-like phenotypes in many disease models. Newly-identified PD-linked genes, including auxilin, synaptojanin-1 and Rab39b, as well as putative risk genes for idiopathic PD (endophilinA1, Rab29, GAK), further support endosomal sorting deficits as being central to PD. LRRK2 may represent a nexus by regulating many distinct features of endosomal sorting, potentially via phosphorylation of key endocytosis machinery (i.e., auxilin, synaptojanin-1, endoA1) and Rab GTPases (i.e., Rab29, Rab8A, Rab10) that function within these pathways. In turn, LRRK2 kinase activity is critically regulated by Rab29 at the Golgi complex and retromer-associated VPS35 at endosomes. Taken together, the known functions of PD-associated gene products, the impact of disease-linked mutations, and the emerging functional interactions between these proteins points to endosomal sorting pathways as a key point of convergence in the pathogenesis of PD.
Collapse
Affiliation(s)
- Lindsey A Cunningham
- Van Andel Institute Graduate School, Grand Rapids, MI, United States; Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Darren J Moore
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States.
| |
Collapse
|
35
|
Andersen MA, Sotty F, Jensen PH, Badolo L, Jeggo R, Smith GP, Christensen KV. Long-Term Exposure to PFE-360 in the AAV-α-Synuclein Rat Model: Findings and Implications. eNeuro 2019; 6:ENEURO.0453-18.2019. [PMID: 31685675 PMCID: PMC6978918 DOI: 10.1523/eneuro.0453-18.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 10/10/2019] [Accepted: 10/21/2019] [Indexed: 12/30/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder associated with impaired motor function and several non-motor symptoms, with no available disease modifying treatment. Intracellular accumulation of pathological α-synuclein inclusions is a hallmark of idiopathic PD, whereas, dominant mutations in leucine-rich repeat kinase 2 (LRRK2) are associated with familial PD that is clinically indistinguishable from idiopathic PD. Recent evidence supports the hypothesis that an increase in LRRK2 kinase activity is associated with the development of not only familial LRRK2 PD, but also idiopathic PD. Previous reports have shown preclinical effects of LRRK2 modulation on α-synuclein-induced neuropathology. Increased subthalamic nucleus (STN) burst firing in preclinical neurotoxin models and PD patients is hypothesized to be causally involved in the development of the motor deficit in PD. To study a potential pathophysiological relationship between α-synuclein pathology and LRRK2 kinase activity in PD, we investigated the effect of chronic LRRK2 inhibition in an AAV-α-synuclein overexpression rat model. In this study, we report that chronic LRRK2 inhibition using PFE-360 only induced a marginal effect on motor function. In addition, the aberrant STN burst firing and associated neurodegenerative processes induced by α-synuclein overexpression model remained unaffected by chronic LRRK2 inhibition. Our findings do not strongly support LRRK2 inhibition for the treatment of PD. Therefore, the reported beneficial effects of LRRK2 inhibition in similar α-synuclein overexpression rodent models must be considered with prudence and additional studies are warranted in alternative α-synuclein-based models.
Collapse
Affiliation(s)
- Michael Aagaard Andersen
- Neurodegeneration, Neuroscience Drug Discovery DK, H. Lundbeck A/S, DK-2500 Valby Denmark
- Department of Biomedicine, Dandrite, Faculty of Health, Aarhus University, DK-8000 Aarhus Denmark
| | - Florence Sotty
- Neurodegeneration, Neuroscience Drug Discovery DK, H. Lundbeck A/S, DK-2500 Valby Denmark
| | - Poul Henning Jensen
- Department of Biomedicine, Dandrite, Faculty of Health, Aarhus University, DK-8000 Aarhus Denmark
| | - Lassina Badolo
- Department of Discovery DMPK, H. Lundbeck A/S, DK-2500 Valby Denmark
| | - Ross Jeggo
- Neurodegeneration, Neuroscience Drug Discovery DK, H. Lundbeck A/S, DK-2500 Valby Denmark
| | - Garrick Paul Smith
- Department of Discovery Chemistry 2, H. Lundbeck A/S, DK-2500 Valby Denmark
| | | |
Collapse
|
36
|
Abstract
Kinase activating missense mutations in leucine-rich repeat kinase 2 (LRRK2) are pathogenically linked to neurodegenerative Parkinson's disease (PD). Over the past decade, substantial effort has been devoted to the development of potent and selective small molecule inhibitors of LRRK2, as well as their preclinical testing across different Parkinson's disease models. This review outlines the genetic and biochemical evidence that pathogenic missense mutations increase LRRK2 kinase activity, which in turn provides the rationale for the development of small molecule inhibitors as potential PD therapeutics. An overview of progress in the development of LRRK2 inhibitors is provided, which in particular indicates that highly selective and potent compounds capable of clinical utility have been developed. We outline evidence from rodent- and human-induced pluripotent stem cell models that support a pathogenic role for LRRK2 kinase activity, and review the substantial experiments aimed at evaluating the safety of LRRK2 inhibitors. We address challenges still to overcome in the translational therapeutic pipeline, including biomarker development and clinical trial strategies, and finally outline the potential utility of LRRK2 inhibitors for other genetic forms of PD and ultimately sporadic PD. Collective evidence supports the ongoing clinical translation of LRRK2 inhibitors as a therapeutic intervention for PD is greatly needed.
Collapse
|
37
|
Targeting leucine-rich repeat kinase 2 (LRRK2) for the treatment of Parkinson's disease. Future Med Chem 2019; 11:1953-1977. [DOI: 10.4155/fmc-2018-0484] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a serine-threonine kinase involved in multiple cellular processes and signaling pathways. LRRK2 mutations are associated with autosomal-inherited Parkinson's disease (PD), and evidence suggests that LRRK2 pathogenic variants generally increase kinase activity. Therefore, inhibition of LRRK2 kinase function is a promising therapeutic strategy for PD treatment. The search for drug-like molecules capable of reducing LRRK2 kinase activity in PD led to the design of selective LRRK2 inhibitors predicted to be within the CNS drug-like space. This review highlights the journey that translates chemical tools for interrogating the role of LRRK2 in PD into promising drug candidates, addressing the challenges in discovering selective and brain-penetrant LRRK2 modulators and exploring the structure–activity relationship of distinct LRRK2 inhibitors.
Collapse
|
38
|
Kluss JH, Mamais A, Cookson MR. LRRK2 links genetic and sporadic Parkinson's disease. Biochem Soc Trans 2019; 47:651-661. [PMID: 30837320 PMCID: PMC6563926 DOI: 10.1042/bst20180462] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/05/2019] [Accepted: 02/07/2019] [Indexed: 12/21/2022]
Abstract
The past two decades in research has revealed the importance of leucine-rich repeat kinase 2 (LRRK2) in both monogenic and sporadic forms of Parkinson's disease (PD). In families, mutations in LRRK2 can cause PD with age-dependent but variable penetrance and genome-wide association studies have found variants of the gene that are risk factors for sporadic PD. Functional studies have suggested that the common mechanism that links all disease-associated variants is that they increase LRRK2 kinase activity, albeit in different ways. Here, we will discuss the roles of LRRK2 in areas of inflammation and vesicular trafficking in the context of monogenic and sporadic PD. We will also provide a hypothetical model that links inflammation and vesicular trafficking together in an effort to outline how these pathways might interact and eventually lead to neuronal cell death. We will also highlight the translational potential of LRRK2-specific kinase inhibitors for the treatment of PD.
Collapse
Affiliation(s)
- Jillian H Kluss
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707, U.S.A
| | - Adamantios Mamais
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707, U.S.A
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707, U.S.A.
| |
Collapse
|
39
|
Henderson MX, Sengupta M, McGeary I, Zhang B, Olufemi MF, Brown H, Trojanowski JQ, Lee VMY. LRRK2 inhibition does not impart protection from α-synuclein pathology and neuron death in non-transgenic mice. Acta Neuropathol Commun 2019; 7:28. [PMID: 30808409 PMCID: PMC6391792 DOI: 10.1186/s40478-019-0679-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 02/17/2019] [Indexed: 12/14/2022] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are one of the most common causes of familial Parkinson's disease (PD). The most common mutations in the LRRK2 gene induce elevated kinase activity of the LRRK2 protein. Recent studies have also suggested that LRRK2 kinase activity may be elevated in idiopathic PD patients, even in the absence of LRRK2 mutations. LRRK2 is therefore a prime candidate for small molecule kinase inhibitor development. However, it is currently unknown how LRRK2 influences the underlying pathogenesis of PD and how LRRK2 might influence extant pathogenesis. To understand whether LRRK2 inhibition would show some benefit in the absence of LRRK2 mutations, we treated a preclinical mouse model of PD with the potent LRRK2 inhibitor MLi-2. The inhibitor was well-tolerated by mice and dramatically reduced LRRK2 kinase activity. However, LRRK2 inhibition did not reverse motor phenotypes, pathological α-synuclein accumulation or neuron loss. The current study suggests that LRRK2 is not necessary for α-synuclein pathogenesis in this mouse model of PD and that further studies are needed to assess the likely clinical benefit of LRRK2 inhibition in idiopathic PD.
Collapse
Affiliation(s)
- Michael X Henderson
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, 3600 Spruce St, 3rd Floor Maloney, Philadelphia, PA, 19104-4283, USA
| | - Medha Sengupta
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, 3600 Spruce St, 3rd Floor Maloney, Philadelphia, PA, 19104-4283, USA
| | - Ian McGeary
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, 3600 Spruce St, 3rd Floor Maloney, Philadelphia, PA, 19104-4283, USA
| | - Bin Zhang
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, 3600 Spruce St, 3rd Floor Maloney, Philadelphia, PA, 19104-4283, USA
| | - Modupe F Olufemi
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, 3600 Spruce St, 3rd Floor Maloney, Philadelphia, PA, 19104-4283, USA
| | - Hannah Brown
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, 3600 Spruce St, 3rd Floor Maloney, Philadelphia, PA, 19104-4283, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, 3600 Spruce St, 3rd Floor Maloney, Philadelphia, PA, 19104-4283, USA
| | - Virginia M Y Lee
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, 3600 Spruce St, 3rd Floor Maloney, Philadelphia, PA, 19104-4283, USA.
| |
Collapse
|
40
|
Araki M, Ito G, Tomita T. Physiological and pathological functions of LRRK2: implications from substrate proteins. Neuronal Signal 2018; 2:NS20180005. [PMID: 32714591 PMCID: PMC7373236 DOI: 10.1042/ns20180005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 02/06/2023] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) encodes a 2527-amino acid (aa) protein composed of multiple functional domains, including a Ras of complex proteins (ROC)-type GTP-binding domain, a carboxyl terminal of ROC (COR) domain, a serine/threonine protein kinase domain, and several repeat domains. LRRK2 is genetically involved in the pathogenesis of both sporadic and familial Parkinson's disease (FPD). Parkinson's disease (PD) is the second most common neurodegenerative disorder, manifesting progressive motor dysfunction. PD is pathologically characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta, and the presence of intracellular inclusion bodies called Lewy bodies (LB) in the remaining neurons. As the most frequent PD-causing mutation in LRRK2, G2019S, increases the kinase activity of LRRK2, an abnormal increase in LRRK2 kinase activity is believed to contribute to PD pathology; however, the precise biological functions of LRRK2 involved in PD pathogenesis remain unknown. Although biochemical studies have discovered several substrate proteins of LRRK2 including Rab GTPases and tau, little is known about whether excess phosphorylation of these substrates is the cause of the neurodegeneration in PD. In this review, we summarize latest findings regarding the physiological and pathological functions of LRRK2, and discuss the possible molecular mechanisms of neurodegeneration caused by LRRK2 and its substrates.
Collapse
Affiliation(s)
- Miho Araki
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Genta Ito
- Laboratory of Brain and Neurological Disorders, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Laboratory of Brain and Neurological Disorders, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
41
|
Parkinson's disease-like burst firing activity in subthalamic nucleus induced by AAV-α-synuclein is normalized by LRRK2 modulation. Neurobiol Dis 2018; 116:13-27. [DOI: 10.1016/j.nbd.2018.04.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/10/2018] [Accepted: 04/16/2018] [Indexed: 01/13/2023] Open
|
42
|
Kelly K, Wang S, Boddu R, Liu Z, Moukha-Chafiq O, Augelli-Szafran C, West AB. The G2019S mutation in LRRK2 imparts resiliency to kinase inhibition. Exp Neurol 2018; 309:1-13. [PMID: 30048714 DOI: 10.1016/j.expneurol.2018.07.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 07/18/2018] [Accepted: 07/21/2018] [Indexed: 12/13/2022]
Abstract
The G2019S mutation in LRRK2 is one of the most common known genetic causes of neurodegeneration and Parkinson disease (PD). LRRK2 mutations are thought to enhance LRRK2 kinase activity. Efficacious small molecule LRRK2 kinase inhibitors with favorable drug properties have recently been developed for pre-clinical studies in rodent models, and inhibitors have advanced to safety trials in humans. Rats that express human G2019S-LRRK2 protein and G2019S-LRRK2 knock-in mice provide newly characterized models to better understand the ostensible target for inhibitors. Herein, we explore the relationships between LRRK2 kinase inhibition in the brain and the periphery to establish the link between LRRK2 kinase activity and protein stability, induction of lysosomal defects in kidney and lung, and how G2019S-LRRK2 expression impacts these phenotypes. Using a novel ultra-sensitive scalable assay based on protein capillary electrophoresis with LRRK2 kinase inhibitors included in-diet, G2019S-LRRK2 protein was resilient to inhibition compared to wild-type (WT)-LRRK2 protein, particularly in the brain. Whereas WT-LRRK2 kinase activity could be completed blocked without lowering LRRK2 protein levels, higher inhibitor concentrations were necessary to fully reduce G2019S-LRRK2 activity. G2019S-LRRK2 expression afforded robust protection from inhibitor-induced kidney lysosomal defects, suggesting a gain-of-function for the mutation in this phenotype. In rodents treated with inhibitors, parallel measurements of phospho-Rab10 revealed a poor correlation to phospho-LRRK2, likely due to cells that express Rab10 but poorly express LRRK2 in heterogenous tissues and cell isolates. In summary, our results highlight several challenges associated with the inhibition of the G2019S-LRRK2 kinase that might be considered in initial clinical efforts.
Collapse
Affiliation(s)
- Kaela Kelly
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Shijie Wang
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Ravindra Boddu
- Division of Nephrology, Nephrology Research and Training Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Zhiyong Liu
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | | | | | - Andrew B West
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|