1
|
Filippini F, Galli T. Unveiling defects of secretion mechanisms in Parkinson's disease. J Biol Chem 2024; 300:107603. [PMID: 39059489 PMCID: PMC11378209 DOI: 10.1016/j.jbc.2024.107603] [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: 01/30/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Neurodegenerative diseases are characterized by progressive dysfunction and loss of specific sets of neurons. While extensive research has focused on elucidating the genetic and epigenetic factors and molecular mechanisms underlying these disorders, emerging evidence highlights the critical role of secretion in the pathogenesis, possibly even onset, and progression of neurodegenerative diseases, suggesting the occurrence of non-cell-autonomous mechanisms. Secretion is a fundamental process that regulates intercellular communication, supports cellular homeostasis, and orchestrates various physiological functions in the body. Defective secretion can impair the release of neurotransmitters and other signaling molecules, disrupting synaptic transmission and compromising neuronal survival. It can also contribute to the accumulation, misfolding, and aggregation of disease-associated proteins, leading to neurotoxicity and neuronal dysfunction. In this review, we discuss the implications of defective secretion in the context of Parkinson's disease, emphasizing its role in protein aggregation, synaptic dysfunction, extracellular vesicle secretion, and neuroinflammation. We propose a multiple-hit model whereby protein accumulation and secretory defects must be combined for the onset and progression of the disease.
Collapse
Affiliation(s)
- Francesca Filippini
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Thierry Galli
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Membrane Traffic in Healthy & Diseased Brain, Paris, France; Groupe Hospitalier Universitaire Paris Psychiatrie & Neurosciences, Paris, France.
| |
Collapse
|
2
|
Yuan Y, Li H, Sreeram K, Malankhanova T, Boddu R, Strader S, Chang A, Bryant N, Yacoubian TA, Standaert DG, Erb M, Moore DJ, Sanders LH, Lutz MW, Velmeshev D, West AB. Single molecule array measures of LRRK2 kinase activity in serum link Parkinson's disease severity to peripheral inflammation. Mol Neurodegener 2024; 19:47. [PMID: 38862989 PMCID: PMC11167795 DOI: 10.1186/s13024-024-00738-4] [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: 02/22/2024] [Accepted: 06/02/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND LRRK2-targeting therapeutics that inhibit LRRK2 kinase activity have advanced to clinical trials in idiopathic Parkinson's disease (iPD). LRRK2 phosphorylates Rab10 on endolysosomes in phagocytic cells to promote some types of immunological responses. The identification of factors that regulate LRRK2-mediated Rab10 phosphorylation in iPD, and whether phosphorylated-Rab10 levels change in different disease states, or with disease progression, may provide insights into the role of Rab10 phosphorylation in iPD and help guide therapeutic strategies targeting this pathway. METHODS Capitalizing on past work demonstrating LRRK2 and phosphorylated-Rab10 interact on vesicles that can shed into biofluids, we developed and validated a high-throughput single-molecule array assay to measure extracellular pT73-Rab10. Ratios of pT73-Rab10 to total Rab10 measured in biobanked serum samples were compared between informative groups of transgenic mice, rats, and a deeply phenotyped cohort of iPD cases and controls. Multivariable and weighted correlation network analyses were used to identify genetic, transcriptomic, clinical, and demographic variables that predict the extracellular pT73-Rab10 to total Rab10 ratio. RESULTS pT73-Rab10 is absent in serum from Lrrk2 knockout mice but elevated by LRRK2 and VPS35 mutations, as well as SNCA expression. Bone-marrow transplantation experiments in mice show that serum pT73-Rab10 levels derive primarily from circulating immune cells. The extracellular ratio of pT73-Rab10 to total Rab10 is dynamic, increasing with inflammation and rapidly decreasing with LRRK2 kinase inhibition. The ratio of pT73-Rab10 to total Rab10 is elevated in iPD patients with greater motor dysfunction, irrespective of disease duration, age, sex, or the usage of PD-related or anti-inflammatory medications. pT73-Rab10 to total Rab10 ratios are associated with neutrophil degranulation, antigenic responses, and suppressed platelet activation. CONCLUSIONS The extracellular serum ratio of pT73-Rab10 to total Rab10 is a novel pharmacodynamic biomarker for LRRK2-linked innate immune activation associated with disease severity in iPD. We propose that those iPD patients with higher serum pT73-Rab10 levels may benefit from LRRK2-targeting therapeutics that mitigate associated deleterious immunological responses.
Collapse
Affiliation(s)
- Yuan Yuan
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Huizhong Li
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Kashyap Sreeram
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Tuyana Malankhanova
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Ravindra Boddu
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Samuel Strader
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Allison Chang
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Nicole Bryant
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Talene A Yacoubian
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David G Standaert
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Madalynn Erb
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Darren J Moore
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Laurie H Sanders
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Neurology, Duke University, Durham, NC, USA
- Department of Pathology, Duke University, Durham, NC, USA
| | - Michael W Lutz
- Department of Neurology, Duke University, Durham, NC, USA
- Department of Pathology, Duke University, Durham, NC, USA
| | | | - Andrew B West
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA.
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Neurology, Duke University, Durham, NC, USA.
- Department of Neurobiology, Duke University, Durham, NC, USA.
| |
Collapse
|
3
|
Yuan Y, Li H, Sreeram K, Malankhanova T, Boddu R, Strader S, Chang A, Bryant N, Yacoubian TA, Standaert DG, Erb M, Moore DJ, Sanders LH, Lutz MW, Velmeshev D, West AB. Single molecule array measures of LRRK2 kinase activity in serum link Parkinson's disease severity to peripheral inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589570. [PMID: 38659797 PMCID: PMC11042295 DOI: 10.1101/2024.04.15.589570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Background LRRK2-targeting therapeutics that inhibit LRRK2 kinase activity have advanced to clinical trials in idiopathic Parkinson's disease (iPD). LRRK2 phosphorylates Rab10 on endolysosomes in phagocytic cells to promote some types of immunological responses. The identification of factors that regulate LRRK2-mediated Rab10 phosphorylation in iPD, and whether phosphorylated-Rab10 levels change in different disease states, or with disease progression, may provide insights into the role of Rab10 phosphorylation in iPD and help guide therapeutic strategies targeting this pathway. Methods Capitalizing on past work demonstrating LRRK2 and phosphorylated-Rab10 interact on vesicles that can shed into biofluids, we developed and validated a high-throughput single-molecule array assay to measure extracellular pT73-Rab10. Ratios of pT73-Rab10 to total Rab10 measured in biobanked serum samples were compared between informative groups of transgenic mice, rats, and a deeply phenotyped cohort of iPD cases and controls. Multivariable and weighted correlation network analyses were used to identify genetic, transcriptomic, clinical, and demographic variables that predict the extracellular pT73-Rab10 to total Rab10 ratio. Results pT73-Rab10 is absent in serum from Lrrk2 knockout mice but elevated by LRRK2 and VPS35 mutations, as well as SNCA expression. Bone-marrow transplantation experiments in mice show that serum pT73-Rab10 levels derive primarily from circulating immune cells. The extracellular ratio of pT73-Rab10 to total Rab10 is dynamic, increasing with inflammation and rapidly decreasing with LRRK2 kinase inhibition. The ratio of pT73-Rab10 to total Rab10 is elevated in iPD patients with greater motor dysfunction, irrespective of disease duration, age, sex, or the usage of PD-related or anti-inflammatory medications. pT73-Rab10 to total Rab10 ratios are associated with neutrophil activation, antigenic responses, and the suppression of platelet activation. Conclusions The extracellular ratio of pT73-Rab10 to total Rab10 in serum is a novel pharmacodynamic biomarker for LRRK2-linked innate immune activation associated with disease severity in iPD. We propose that those iPD patients with higher serum pT73-Rab10 levels may benefit from LRRK2-targeting therapeutics to mitigate associated deleterious immunological responses.
Collapse
|
4
|
Naaldijk Y, Fernández B, Fasiczka R, Fdez E, Leghay C, Croitoru I, Kwok JB, Boulesnane Y, Vizeneux A, Mutez E, Calvez C, Destée A, Taymans JM, Aragon AV, Yarza AB, Padmanabhan S, Delgado M, Alcalay RN, Chatterton Z, Dzamko N, Halliday G, Ruiz-Martínez J, Chartier-Harlin MC, Hilfiker S. A potential patient stratification biomarker for Parkinson´s disease based on LRRK2 kinase-mediated centrosomal alterations in peripheral blood-derived cells. NPJ Parkinsons Dis 2024; 10:12. [PMID: 38191886 PMCID: PMC10774440 DOI: 10.1038/s41531-023-00624-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 12/14/2023] [Indexed: 01/10/2024] Open
Abstract
Parkinson´s disease (PD) is a common neurodegenerative movement disorder and leucine-rich repeat kinase 2 (LRRK2) is a promising therapeutic target for disease intervention. However, the ability to stratify patients who will benefit from such treatment modalities based on shared etiology is critical for the success of disease-modifying therapies. Ciliary and centrosomal alterations are commonly associated with pathogenic LRRK2 kinase activity and can be detected in many cell types. We previously found centrosomal deficits in immortalized lymphocytes from G2019S-LRRK2 PD patients. Here, to investigate whether such deficits may serve as a potential blood biomarker for PD which is susceptible to LRKK2 inhibitor treatment, we characterized patient-derived cells from distinct PD cohorts. We report centrosomal alterations in peripheral cells from a subset of early-stage idiopathic PD patients which is mitigated by LRRK2 kinase inhibition, supporting a role for aberrant LRRK2 activity in idiopathic PD. Centrosomal defects are detected in R1441G-LRRK2 and G2019S-LRRK2 PD patients and in non-manifesting LRRK2 mutation carriers, indicating that they accumulate prior to a clinical PD diagnosis. They are present in immortalized cells as well as in primary lymphocytes from peripheral blood. These findings indicate that analysis of centrosomal defects as a blood-based patient stratification biomarker may help nominate idiopathic PD patients who will benefit from LRRK2-related therapeutics.
Collapse
Affiliation(s)
- Yahaira Naaldijk
- Department. of Anesthesiology and Department. of Physiology, Pharmacology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Belén Fernández
- Institute of Parasitology and Biomedicine ´López-Neyra¨, Consejo Superior de Investigaciones Científicas (CSIC), 18016, Granada, Spain
| | - Rachel Fasiczka
- Department. of Anesthesiology and Department. of Physiology, Pharmacology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Elena Fdez
- Institute of Parasitology and Biomedicine ´López-Neyra¨, Consejo Superior de Investigaciones Científicas (CSIC), 18016, Granada, Spain
| | - Coline Leghay
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - LilNCog - Lille Neuroscience & Cognition, F-59000, Lille, France
| | - Ioana Croitoru
- Biodonostia Health Research Institute (IIS Biodonostia), San Sebastain, Spain
| | - John B Kwok
- School of Medical Sciences, Faculty of Medicine and Health and the Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Yanisse Boulesnane
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - LilNCog - Lille Neuroscience & Cognition, F-59000, Lille, France
| | - Amelie Vizeneux
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - LilNCog - Lille Neuroscience & Cognition, F-59000, Lille, France
| | - Eugenie Mutez
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - LilNCog - Lille Neuroscience & Cognition, F-59000, Lille, France
| | - Camille Calvez
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - LilNCog - Lille Neuroscience & Cognition, F-59000, Lille, France
| | - Alain Destée
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - LilNCog - Lille Neuroscience & Cognition, F-59000, Lille, France
| | - Jean-Marc Taymans
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - LilNCog - Lille Neuroscience & Cognition, F-59000, Lille, France
| | | | - Alberto Bergareche Yarza
- Biodonostia Health Research Institute (IIS Biodonostia), San Sebastain, Spain
- Donostia University Hospital, San Sebastian, Spain
| | | | - Mario Delgado
- Institute of Parasitology and Biomedicine ´López-Neyra¨, Consejo Superior de Investigaciones Científicas (CSIC), 18016, Granada, Spain
| | - Roy N Alcalay
- Department. of Neurology, Colsumbia University Medical Center, New York, NY, USA
- Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Zac Chatterton
- School of Medical Sciences, Faculty of Medicine and Health and the Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Nicolas Dzamko
- School of Medical Sciences, Faculty of Medicine and Health and the Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Glenda Halliday
- School of Medical Sciences, Faculty of Medicine and Health and the Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Javier Ruiz-Martínez
- Biodonostia Health Research Institute (IIS Biodonostia), San Sebastain, Spain
- Donostia University Hospital, San Sebastian, Spain
| | | | - Sabine Hilfiker
- Department. of Anesthesiology and Department. of Physiology, Pharmacology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA.
| |
Collapse
|
5
|
Komori T, Kuwahara T. An Update on the Interplay between LRRK2, Rab GTPases and Parkinson's Disease. Biomolecules 2023; 13:1645. [PMID: 38002327 PMCID: PMC10669493 DOI: 10.3390/biom13111645] [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: 10/19/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
Over the last decades, research on the pathobiology of neurodegenerative diseases has greatly evolved, revealing potential targets and mechanisms linked to their pathogenesis. Parkinson's disease (PD) is no exception, and recent studies point to the involvement of endolysosomal defects in PD. The endolysosomal system, which tightly controls a flow of endocytosed vesicles targeted either for degradation or recycling, is regulated by a number of Rab GTPases. Their associations with leucine-rich repeat kinase 2 (LRRK2), a major causative and risk protein of PD, has also been one of the hot topics in the field. Understanding their interactions and functions is critical for unraveling their contribution to PD pathogenesis. In this review, we summarize recent studies on LRRK2 and Rab GTPases and attempt to provide more insight into the interaction of LRRK2 with each Rab and its relationship to PD.
Collapse
Affiliation(s)
| | - Tomoki Kuwahara
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| |
Collapse
|
6
|
Jennings D, Huntwork-Rodriguez S, Vissers MFJM, Daryani VM, Diaz D, Goo MS, Chen JJ, Maciuca R, Fraser K, Mabrouk OS, van de Wetering de Rooij J, Heuberger JAAC, Groeneveld GJ, Borin MT, Cruz-Herranz A, Graham D, Scearce-Levie K, De Vicente J, Henry AG, Chin P, Ho C, Troyer MD. LRRK2 Inhibition by BIIB122 in Healthy Participants and Patients with Parkinson's Disease. Mov Disord 2023; 38:386-398. [PMID: 36807624 DOI: 10.1002/mds.29297] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 11/16/2022] [Accepted: 12/01/2022] [Indexed: 02/20/2023] Open
Abstract
BACKGROUND Leucine-rich repeat kinase 2 (LRRK2) inhibition is a promising therapeutic approach for the treatment of Parkinson's disease (PD). OBJECTIVE The aim of this study was to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of the potent, selective, CNS-penetrant LRRK2 inhibitor BIIB122 (DNL151) in healthy participants and patients with PD. METHODS Two randomized, double-blind, placebo-controlled studies were completed. The phase 1 study (DNLI-C-0001) evaluated single and multiple doses of BIIB122 for up to 28 days in healthy participants. The phase 1b study (DNLI-C-0003) evaluated BIIB122 for 28 days in patients with mild to moderate PD. The primary objectives were to investigate the safety, tolerability, and plasma pharmacokinetics of BIIB122. Pharmacodynamic outcomes included peripheral and central target inhibition and lysosomal pathway engagement biomarkers. RESULTS A total of 186/184 healthy participants (146/145 BIIB122, 40/39 placebo) and 36/36 patients (26/26 BIIB122, 10/10 placebo) were randomized/treated in the phase 1 and phase 1b studies, respectively. In both studies, BIIB122 was generally well tolerated; no serious adverse events were reported, and the majority of treatment-emergent adverse events were mild. BIIB122 cerebrospinal fluid/unbound plasma concentration ratio was ~1 (range, 0.7-1.8). Dose-dependent median reductions from baseline were observed in whole-blood phosphorylated serine 935 LRRK2 (≤98%), peripheral blood mononuclear cell phosphorylated threonine 73 pRab10 (≤93%), cerebrospinal fluid total LRRK2 (≤50%), and urine bis (monoacylglycerol) phosphate (≤74%). CONCLUSIONS At generally safe and well-tolerated doses, BIIB122 achieved substantial peripheral LRRK2 kinase inhibition and modulation of lysosomal pathways downstream of LRRK2, with evidence of CNS distribution and target inhibition. These studies support continued investigation of LRRK2 inhibition with BIIB122 for the treatment of PD. © 2023 Denali Therapeutics Inc and The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Danna Jennings
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| | - Sarah Huntwork-Rodriguez
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| | - Maurits F J M Vissers
- Centre for Human Drug Research, Leiden, the Netherlands
- Department of Clinical Neuropharmacology, Leiden University Medical Center, Leiden, the Netherlands
| | - Vinay M Daryani
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| | - Dolores Diaz
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| | - Marisa S Goo
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| | - John J Chen
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| | - Romeo Maciuca
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| | | | | | - Jeroen van de Wetering de Rooij
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
- PRA Health Sciences, Groningen, the Netherlands
| | | | - Geert Jan Groeneveld
- Centre for Human Drug Research, Leiden, the Netherlands
- Department of Clinical Neuropharmacology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marie T Borin
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| | - Andrés Cruz-Herranz
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| | | | - Kimberly Scearce-Levie
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| | - Javier De Vicente
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| | - Anastasia G Henry
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| | - Peter Chin
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| | - Carole Ho
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| | - Matthew D Troyer
- SeniorClinical Scientist - Clinical Development, Denali Therapeutics Inc, South San Francisco, California, USA
| |
Collapse
|
7
|
West AB, Schwarzschild MA. LRRK2-Targeting Therapies March Through the Valley of Death. Mov Disord 2023; 38:361-365. [PMID: 36942368 PMCID: PMC11076002 DOI: 10.1002/mds.29343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 01/24/2023] [Indexed: 03/23/2023] Open
Affiliation(s)
- Andrew B. West
- Duke Center for Neurodegeneration Research, Department of Pharmacology and Cancer Biology, Duke University, 3 Genome court, Durham, NC, 27710, USA
| | - Michael A. Schwarzschild
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, 114 16th Street, Charlestown, MA 02129 USA
| |
Collapse
|
8
|
Niotis K, West AB, Saunders-Pullman R. Who to Enroll in Parkinson Disease Prevention Trials? The Case for Genetically At-Risk Cohorts. Neurology 2022; 99:10-18. [PMID: 35970585 DOI: 10.1212/wnl.0000000000200812] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 04/15/2022] [Indexed: 12/15/2022] Open
Abstract
Therapies that prevent the occurrence of Parkinson disease (PD) (primary prevention) or mitigate the progression of symptoms in those with early disease (secondary prevention) are a critical unmet need in disease management. Despite great promise, PD prevention trials have not yet demonstrated success. Initiation of treatment too late in the disease course and the heterogeneity of disease are obstacles that may have contributed to the failure. Genetically stratified groups offer many advantages to primary and secondary prevention trials. In addition to their ease of identification, they decrease disease heterogeneity on several levels. Particularly, they comprise a phenotypically and pathologically enriched group with defined clinical features, pathogenic mechanisms and associated proteins that may serve as specific trial endpoints, therapeutic targets and biomarkers for disease state, and pharmacodynamic and pharmacokinetic status. However, challenges arise from genetic variant heterogeneity, from reduced penetrance whereby many carriers will not develop PD, and in recruiting a population that will meet the desired outcome in the proposed study duration. In this review, we discussed the opportunities afforded by the enrollment of genetically stratified cohorts (i.e., leucine-rich repeat kinase 2 and glucocerebrosidase 1) into prevention trials with a primary focus on primary prevention trials. We also outlined challenges surrounding the enrollment of these cohorts and offered suggestions to leverage their many advantages.
Collapse
Affiliation(s)
- Kellyann Niotis
- From the Department of Neurology (K.N., R.S.-P.), Mount Sinai Beth Israel Medical Center; Department of Neurology (K.N., R.S.-P.), Icahn School of Medicine at Mount Sinai, New York; and Duke Center for Neurodegeneration Research (A.B.W.), Departments of Pharmacology and Cancer Biology, Neurology, and Neurobiology, Duke University, Durham, NC
| | - Andrew B West
- From the Department of Neurology (K.N., R.S.-P.), Mount Sinai Beth Israel Medical Center; Department of Neurology (K.N., R.S.-P.), Icahn School of Medicine at Mount Sinai, New York; and Duke Center for Neurodegeneration Research (A.B.W.), Departments of Pharmacology and Cancer Biology, Neurology, and Neurobiology, Duke University, Durham, NC
| | - Rachel Saunders-Pullman
- From the Department of Neurology (K.N., R.S.-P.), Mount Sinai Beth Israel Medical Center; Department of Neurology (K.N., R.S.-P.), Icahn School of Medicine at Mount Sinai, New York; and Duke Center for Neurodegeneration Research (A.B.W.), Departments of Pharmacology and Cancer Biology, Neurology, and Neurobiology, Duke University, Durham, NC.
| |
Collapse
|
9
|
Wang S, Unnithan S, Bryant N, Chang A, Rosenthal LS, Pantelyat A, Dawson TM, Al‐Khalidi HR, West AB. Elevated Urinary Rab10 Phosphorylation in Idiopathic Parkinson Disease. Mov Disord 2022; 37:1454-1464. [PMID: 35521944 PMCID: PMC9308673 DOI: 10.1002/mds.29043] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Pathogenic leucine-rich repeat kinase 2 LRRK2 mutations may increase LRRK2 kinase activity and Rab substrate phosphorylation. Genetic association studies link variation in LRRK2 to idiopathic Parkinson disease (iPD) risk. OBJECTIVES Through measurements of the LRRK2 kinase substrate pT73-Rab10 in urinary extracellular vesicles, this study seeks to understand how LRRK2 kinase activity might change with iPD progression. METHODS Using an immunoblotting approach validated in LRRK2 transgenic mice, the ratio of pT73-Rab10 to total Rab10 protein was measured in extracellular vesicles from a cross-section of G2019S LRRK2 mutation carriers (N = 45 participants) as well as 485 urine samples from a novel longitudinal cohort of iPD and controls (N = 85 participants). Generalized estimating equations were used to conduct analyses with commonly used clinical scales. RESULTS Although the G2019S LRRK2 mutation did not increase pT73-Rab10 levels, the ratio of pT73-Rab10 to total Rab10 nominally increased over baseline in iPD urine vesicle samples with time, but did not increase in age-matched controls (1.34-fold vs. 1.05-fold, 95% confidence interval [CI], 0.004-0.56; P = 0.046; Welch's t test). Effect estimates adjusting for sex, age, disease duration, diagnosis, and baseline clinical scores identified increasing total Movement Disorder Society-Sponsored Revision of the Unified (MDS-UPDRS) scores (β = 0.77; CI, 0.52-1.01; P = 0.0001) with each fold increase of pT73-Rab10 to total Rab10. Lower Montreal Cognitive Assessment (MoCA) score in iPD is also associated with increased pT73-Rab10. CONCLUSIONS These results provide initial insights into peripheral LRRK2-dependent Rab phosphorylation, measured in biobanked urine, where higher levels of pT73-Rab10 are associated with worse disease progression. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson Movement Disorder Society.
Collapse
Affiliation(s)
- Shijie Wang
- Duke Center for Neurodegeneration and NeurotherapeuticsDuke UniversityDurhamNorth CarolinaUSA
| | - Shakthi Unnithan
- Department of Biostatistics and BioinformaticsDuke UniversityDurhamNorth CarolinaUSA
| | - Nicole Bryant
- Duke Center for Neurodegeneration and NeurotherapeuticsDuke UniversityDurhamNorth CarolinaUSA
| | - Allison Chang
- Duke Center for Neurodegeneration and NeurotherapeuticsDuke UniversityDurhamNorth CarolinaUSA
| | | | | | - Ted M. Dawson
- Department of NeurologyThe Johns Hopkins UniversityBaltimoreMarylandUSA
- Neurodegeneration and Stem Cell Programs, Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Solomon H. Snyder Department of NeuroscienceJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Pharmacology and Molecular SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Hussein R. Al‐Khalidi
- Department of Biostatistics and BioinformaticsDuke UniversityDurhamNorth CarolinaUSA
| | - Andrew B. West
- Duke Center for Neurodegeneration and NeurotherapeuticsDuke UniversityDurhamNorth CarolinaUSA
| |
Collapse
|
10
|
Jennings D, Huntwork-Rodriguez S, Henry AG, Sasaki JC, Meisner R, Diaz D, Solanoy H, Wang X, Negrou E, Bondar VV, Ghosh R, Maloney MT, Propson NE, Zhu Y, Maciuca RD, Harris L, Kay A, LeWitt P, King TA, Kern D, Ellenbogen A, Goodman I, Siderowf A, Aldred J, Omidvar O, Masoud ST, Davis SS, Arguello A, Estrada AA, de Vicente J, Sweeney ZK, Astarita G, Borin MT, Wong BK, Wong H, Nguyen H, Scearce-Levie K, Ho C, Troyer MD. Preclinical and clinical evaluation of the LRRK2 inhibitor DNL201 for Parkinson's disease. Sci Transl Med 2022; 14:eabj2658. [PMID: 35675433 DOI: 10.1126/scitranslmed.abj2658] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic risk factors for Parkinson's disease (PD). Increased LRRK2 kinase activity is thought to impair lysosomal function and may contribute to the pathogenesis of PD. Thus, inhibition of LRRK2 is a potential disease-modifying therapeutic strategy for PD. DNL201 is an investigational, first-in-class, CNS-penetrant, selective, ATP-competitive, small-molecule LRRK2 kinase inhibitor. In preclinical models, DNL201 inhibited LRRK2 kinase activity as evidenced by reduced phosphorylation of both LRRK2 at serine-935 (pS935) and Rab10 at threonine-73 (pT73), a direct substrate of LRRK2. Inhibition of LRRK2 by DNL201 demonstrated improved lysosomal function in cellular models of disease, including primary mouse astrocytes and fibroblasts from patients with Gaucher disease. Chronic administration of DNL201 to cynomolgus macaques at pharmacologically relevant doses was not associated with adverse findings. In phase 1 and phase 1b clinical trials in 122 healthy volunteers and in 28 patients with PD, respectively, DNL201 at single and multiple doses inhibited LRRK2 and was well tolerated at doses demonstrating LRRK2 pathway engagement and alteration of downstream lysosomal biomarkers. Robust cerebrospinal fluid penetration of DNL201 was observed in both healthy volunteers and patients with PD. These data support the hypothesis that LRRK2 inhibition has the potential to correct lysosomal dysfunction in patients with PD at doses that are generally safe and well tolerated, warranting further clinical development of LRRK2 inhibitors as a therapeutic modality for PD.
Collapse
Affiliation(s)
| | | | | | | | - René Meisner
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Dolores Diaz
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Hilda Solanoy
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Xiang Wang
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Elvira Negrou
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | | | | | - Yuda Zhu
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - Laura Harris
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Angela Kay
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | - Drew Kern
- University of Colorado, School of Medicine, Aurora, CO, USA
| | - Aaron Ellenbogen
- Michigan Institute for Neurological Disorders, Farmington Hills, MI, USA
| | | | - Andrew Siderowf
- University of Pennsylvania, Penn Neurology Pennsylvania Hospital, Philadelphia, PA, USA
| | | | - Omid Omidvar
- Collaborative Neuroscience Research, Long Beach, CA, USA
| | | | | | | | | | | | | | - Giuseppe Astarita
- Denali Therapeutics Inc., South San Francisco, CA, USA.,Henry Ford Health System, Detroit, MI, USA
| | - Marie T Borin
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - Harvey Wong
- University of British Columbia, Vancouver, BC, Canada
| | - Hoang Nguyen
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - Carole Ho
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | |
Collapse
|
11
|
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: 2.0] [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
|
12
|
Udayar V, Chen Y, Sidransky E, Jagasia R. Lysosomal dysfunction in neurodegeneration: emerging concepts and methods. Trends Neurosci 2022; 45:184-199. [PMID: 35034773 PMCID: PMC8854344 DOI: 10.1016/j.tins.2021.12.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 11/23/2021] [Accepted: 12/12/2021] [Indexed: 02/06/2023]
Abstract
The understanding of lysosomes has come a long way since the initial discovery of their role in degrading cellular waste. The lysosome is now recognized as a highly dynamic organelle positioned at the crossroads of cell signaling, transcription, and metabolism. Underscoring its importance is the observation that, in addition to rare monogenic lysosomal storage disorders, genes regulating lysosomal function are implicated in common sporadic neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Developing therapies for these disorders is particularly challenging, largely due to gaps in knowledge of the underlying molecular and cellular processes. In this review, we discuss technological advances that have propelled deeper understanding of the lysosome in neurodegeneration, from elucidating the functions of lysosome-related disease risk variants at the level of the organelle, cell, and tissue, to the development of disease-specific biological models that recapitulate disease manifestations. Finally, we identify key questions to be addressed to successfully bridge the gap to the clinic.
Collapse
Affiliation(s)
- Vinod Udayar
- Roche Pharmaceutical Research and Early Development, Neuroscience and Rare Diseases Discovery & Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Yu Chen
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ellen Sidransky
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Ravi Jagasia
- Roche Pharmaceutical Research and Early Development, Neuroscience and Rare Diseases Discovery & Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| |
Collapse
|
13
|
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: 68] [Impact Index Per Article: 34.0] [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
|
14
|
Leggio L, Paternò G, Vivarelli S, Falzone GG, Giachino C, Marchetti B, Iraci N. Extracellular Vesicles as Novel Diagnostic and Prognostic Biomarkers for Parkinson's Disease. Aging Dis 2021; 12:1494-1515. [PMID: 34527424 PMCID: PMC8407885 DOI: 10.14336/ad.2021.0527] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 05/27/2021] [Indexed: 12/29/2022] Open
Abstract
The elderly population will significantly increase in the next decade and, with it, the proportion of people affected by age-related diseases. Among them, one of the most invalidating is Parkinson's disease (PD), characterized by motor- and non-motor dysfunctions which strongly impair the quality of life of affected individuals. PD is characterized by the progressive degeneration of dopaminergic neurons, with consequent dopamine depletion, and the accumulation of misfolded α-synuclein aggregates. Although 150 years have passed since PD first description, no effective therapies are currently available, but only palliative treatments. Importantly, PD is often diagnosed when the neuronal loss is elevated, making difficult any therapeutic intervention. In this context, two key challenges remain unanswered: (i) the early diagnosis to avoid the insurgence of irreversible symptoms; and (ii) the reliable monitoring of therapy efficacy. Research strives to identify novel biomarkers for PD diagnosis, prognosis, and therapeutic follow-up. One of the most promising sources of biomarkers is represented by extracellular vesicles (EVs), a heterogeneous population of nanoparticles, released by all cells in the microenvironment. Brain-derived EVs are able to cross the blood-brain barrier, protecting their payload from enzymatic degradation, and are easily recovered from biofluids. Interestingly, EV content is strongly influenced by the specific pathophysiological status of the donor cell. In this manuscript, the role of EVs as source of novel PD biomarkers is discussed, providing all recent findings concerning relevant proteins and miRNAs carried by PD patient-derived EVs, from several biological specimens. Moreover, the contribution of mitochondria-derived EVs will be dissected. Finally, the promising possibility to use EVs as source of markers to monitor PD therapy efficacy will be also examined. In the future, larger cohort studies will help to validate these EV-associated candidates, that might be effectively used as non-invasive and robust source of biomarkers for PD.
Collapse
Affiliation(s)
- Loredana Leggio
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, 95125 Catania, Italy.
| | - Greta Paternò
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, 95125 Catania, Italy.
| | - Silvia Vivarelli
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, 95125 Catania, Italy.
| | - Giovanna G Falzone
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, 95125 Catania, Italy.
| | - Carmela Giachino
- Neuropharmacology Section, OASI Research Institute-IRCCS, 94018 Troina, Italy.
| | - Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, 95125 Catania, Italy.
- Neuropharmacology Section, OASI Research Institute-IRCCS, 94018 Troina, Italy.
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, 95125 Catania, Italy.
| |
Collapse
|
15
|
Busatto S, Morad G, Guo P, Moses MA. The role of extracellular vesicles in the physiological and pathological regulation of the blood-brain barrier. FASEB Bioadv 2021; 3:665-675. [PMID: 34485835 PMCID: PMC8409556 DOI: 10.1096/fba.2021-00045] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs) are a subclass of biological nanoparticles secreted by most cell types. Once secreted, EVs can travel long distances to deliver their content to target cells thereby playing a key role in cell-to-cell communication and supporting both physiological and pathological processes. In recent years, the functional versatility of EVs has come to be more widely appreciated. Their heterogeneous structure encloses solubilized bioactive cargoes including proteins and nucleic acids. EVs mirror the secreting cell in composition therefore representing a novel source of diagnostic and prognostic biomarkers. Moreover, due to their unique structure, EVs constitute a promising class of biocompatible nanovehicles for drug delivery as well. Importantly, and of burgeoning interest, is the fact that EVs have the intrinsic ability to breach biological barriers including the complex blood-brain barrier (BBB), whose restrictive nature represents a significant therapeutic challenge. EVs have been shown to contribute to the progression of a variety of brain diseases including metastatic brain cancer, neurodegenerative diseases, and acute pathologies including infections and ischemia. In this review, the role of EVs in the maintenance and regulation of the BBB under normal physiological and pathologic conditions are discussed. Applications of EVs as therapeutic and diagnostic tools in the treatment of diseases that affect the central nervous system are presented as are limitations hindering their broad translation and potential solutions to resolve them.
Collapse
Affiliation(s)
- Sara Busatto
- Vascular Biology ProgramBoston Children's HospitalBostonMAUSA
- Department of SurgeryBoston Children's Hospital and Harvard Medical SchoolBostonMAUSA
| | - Golnaz Morad
- Department of Surgical OncologyUniversity of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Peng Guo
- Vascular Biology ProgramBoston Children's HospitalBostonMAUSA
- Department of SurgeryBoston Children's Hospital and Harvard Medical SchoolBostonMAUSA
| | - Marsha A. Moses
- Vascular Biology ProgramBoston Children's HospitalBostonMAUSA
- Department of SurgeryBoston Children's Hospital and Harvard Medical SchoolBostonMAUSA
| |
Collapse
|
16
|
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: 2.3] [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
|
17
|
Chittoor-Vinod VG, Nichols RJ, Schüle B. Genetic and Environmental Factors Influence the Pleomorphy of LRRK2 Parkinsonism. Int J Mol Sci 2021; 22:1045. [PMID: 33494262 PMCID: PMC7864502 DOI: 10.3390/ijms22031045] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/16/2021] [Accepted: 01/17/2021] [Indexed: 12/25/2022] Open
Abstract
Missense mutations in the LRRK2 gene were first identified as a pathogenic cause of Parkinson's disease (PD) in 2004. Soon thereafter, a founder mutation in LRRK2, p.G2019S (rs34637584), was described, and it is now estimated that there are approximately 100,000 people worldwide carrying this risk variant. While the clinical presentation of LRRK2 parkinsonism has been largely indistinguishable from sporadic PD, disease penetrance and age at onset can be quite variable. In addition, its neuropathological features span a wide range from nigrostriatal loss with Lewy body pathology, lack thereof, or atypical neuropathology, including a large proportion of cases with concomitant Alzheimer's pathology, hailing LRRK2 parkinsonism as the "Rosetta stone" of parkinsonian disorders, which provides clues to an understanding of the different neuropathological trajectories. These differences may result from interactions between the LRRK2 mutant protein and other proteins or environmental factors that modify LRRK2 function and, thereby, influence pathobiology. This review explores how potential genetic and biochemical modifiers of LRRK2 function may contribute to the onset and clinical presentation of LRRK2 parkinsonism. We review which genetic modifiers of LRRK2 influence clinical symptoms, age at onset, and penetrance, what LRRK2 mutations are associated with pleomorphic LRRK2 neuropathology, and which environmental modifiers can augment LRRK2 mutant pathophysiology. Understanding how LRRK2 function is influenced and modulated by other interactors and environmental factors-either increasing toxicity or providing resilience-will inform targeted therapeutic development in the years to come. This will allow the development of disease-modifying therapies for PD- and LRRK2-related neurodegeneration.
Collapse
Affiliation(s)
| | - R. Jeremy Nichols
- Department Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Birgitt Schüle
- Department Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA;
| |
Collapse
|