1
|
Maayan Eshed G, Alcalay RN. Precision Medicine in Parkinson's Disease. Neurol Clin 2025; 43:365-381. [PMID: 40185526 DOI: 10.1016/j.ncl.2024.12.011] [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: 04/07/2025]
Abstract
The continually accumulating knowledge of Parkinson's disease (PD) genetics presents potential disease modification opportunities through targeting specific genes and associated metabolic pathways. Glucosylceramidase beta 1-associated PD and leucine-rich repeat kinase 2-associated PD are attractive drug targets, since their respective mutations significantly increase PD risk and, at the same time, are relatively prevalent in the PD population. Here, we review clinical trials and preclinical efforts whose mechanisms target genetic forms of PD, focusing on these 2 genes and their metabolic pathways. Such therapies could also potentially modify sporadic (ie, without a clear genetic risk factor) PD.
Collapse
Affiliation(s)
- Gadi Maayan Eshed
- Movement Disorders Division, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Roy N Alcalay
- Movement Disorders Division, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Tel Aviv Faculty of Medicine, Tel Aviv University; Department of Neurology, Columbia University Irving Medical Center, New York, USA.
| |
Collapse
|
2
|
Bellini G, D'Antongiovanni V, Palermo G, Antonioli L, Fornai M, Ceravolo R, Bernardini N, Derkinderen P, Pellegrini C. α-Synuclein in Parkinson's Disease: From Bench to Bedside. Med Res Rev 2025; 45:909-946. [PMID: 39704040 PMCID: PMC11976381 DOI: 10.1002/med.22091] [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: 05/12/2024] [Revised: 09/24/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024]
Abstract
α-Synuclein (α-syn), a pathological hallmark of PD, is emerging as a bridging element at the crossroads between neuro/immune-inflammatory responses and neurodegeneration in PD. Several evidence show that pathological α-syn accumulates in neuronal and non-neuronal cells (i.e., neurons, microglia, macrophages, skin cells, and intestinal cells) in central and peripheral tissues since the prodromal phase of the disease, contributing to brain pathology. Indeed, pathological α-syn deposition can promote neurogenic/immune-inflammatory responses that contribute to systemic and central neuroinflammation associated with PD. After providing an overview of the structure and functions of physiological α-syn as well as its pathological forms, we review current studies about the role of neuronal and non-neuronal α-syn at the crossroads between neuroinflammation and neurodegeneration in PD. In addition, we provide an overview of the correlation between the accumulation of α-syn in central and peripheral tissues and PD, related symptoms, and neuroinflammation. Special attention was paid to discussing whether targeting α-syn can represent a suitable therapeutical approach for PD.
Collapse
Affiliation(s)
- Gabriele Bellini
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
- Department of NeurologyThe Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, NYU Langone HealthNew York CityNew YorkUSA
| | - Vanessa D'Antongiovanni
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Giovanni Palermo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Roberto Ceravolo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Nunzia Bernardini
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Pascal Derkinderen
- Department of NeurologyNantes Université, CHU Nantes, INSERMNantesFrance
| | - Carolina Pellegrini
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| |
Collapse
|
3
|
Yaribash S, Mohammadi K, Sani MA. Alpha-Synuclein Pathophysiology in Neurodegenerative Disorders: A Review Focusing on Molecular Mechanisms and Treatment Advances in Parkinson's Disease. Cell Mol Neurobiol 2025; 45:30. [PMID: 40140103 PMCID: PMC11947388 DOI: 10.1007/s10571-025-01544-2] [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: 02/11/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025]
Abstract
Worldwide aging has contributed to the growth of prevalence of neurodegenerative diseases (NDDs), including Parkinson's disease among the elderlies. The advanced destruction of dopaminergic neurons in the substantia nigra, due to many accelerator factors in the brain is the main mechanism of Parkinson's disease. The pathological aggregated alpha-synuclein (α-syn), a protein implicated in multiple neurodegenerative disorders, is one of the critical factors in this neurodegenerative disease and other similar disorders. The misfolding and aggregation of α-syn may interrupt critical processes, including functions of synaptic vesicles and can lead to neuronal death. This protein is encoded by Alpha-Synuclein Gene (SNCA) and mutation in this gene can lead to dysfunctions of the protein structure. Since, therapeutic policies that aim α-syn are promising approaches. Advances in immunotherapies, molecular chaperones, gene therapy targeting SNCA, and DNA aptamers are some examples of this strategy. This review aims to comprehensively assess the current knowledge and evidence on α-syn pathology, genetic determinants, and novel therapeutic methods in Parkinson,'s disease and other synucleinopathies. Continued investigation to discover interventions in this system could result in finding of effective and safe treatments for NDDs.
Collapse
Affiliation(s)
- Shakila Yaribash
- Faculty of Pharmacy, Tehran University of Medical Sciences, Enghelab Square, 16 Azar Street, Tehran, 1417614411, Iran
| | - Keyhan Mohammadi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Enghelab Square, 16 Azar Street, Tehran, 1417614411, Iran.
- Research Center for Antibiotics Stewardship and Antimicrobial Resistance, Infectious Diseases Department, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahmood Alizadeh Sani
- Division of Food Safety and Hygiene, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Enghelab Square, 16 Azar Street, Tehran, 1417613151, Iran
| |
Collapse
|
4
|
Fu Y, Zhang J, Qin R, Ren Y, Zhou T, Han B, Liu B. Activating autophagy to eliminate toxic protein aggregates with small molecules in neurodegenerative diseases. Pharmacol Rev 2025; 77:100053. [PMID: 40187044 DOI: 10.1016/j.pharmr.2025.100053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 12/05/2024] [Indexed: 04/07/2025] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer disease, Parkinson disease, Huntington disease, amyotrophic lateral sclerosis, and frontotemporal dementia, are well known to pose formidable challenges for their treatment due to their intricate pathogenesis and substantial variability among patients, including differences in environmental exposures and genetic predispositions. One of the defining characteristics of NDs is widely reported to be the buildup of misfolded proteins. For example, Alzheimer disease is marked by amyloid beta and hyperphosphorylated Tau aggregates, whereas Parkinson disease exhibits α-synuclein aggregates. Amyotrophic lateral sclerosis and frontotemporal dementia exhibit TAR DNA-binding protein 43, superoxide dismutase 1, and fused-in sarcoma protein aggregates, and Huntington disease involves mutant huntingtin and polyglutamine aggregates. These misfolded proteins are the key biomarkers of NDs and also serve as potential therapeutic targets, as they can be addressed through autophagy, a process that removes excess cellular inclusions to maintain homeostasis. Various forms of autophagy, including macroautophagy, chaperone-mediated autophagy, and microautophagy, hold a promise in eliminating toxic proteins implicated in NDs. In this review, we focus on elucidating the regulatory connections between autophagy and toxic proteins in NDs, summarizing the cause of the aggregates, exploring their impact on autophagy mechanisms, and discussing how autophagy can regulate toxic protein aggregation. Moreover, we underscore the activation of autophagy as a potential therapeutic strategy across different NDs and small molecules capable of activating autophagy pathways, such as rapamycin targeting the mTOR pathway to clear α-synuclein and Sertraline targeting the AMPK/mTOR/RPS6KB1 pathway to clear Tau, to further illustrate their potential in NDs' therapeutic intervention. Together, these findings would provide new insights into current research trends and propose small-molecule drugs targeting autophagy as promising potential strategies for the future ND therapies. SIGNIFICANCE STATEMENT: This review provides an in-depth overview of the potential of activating autophagy to eliminate toxic protein aggregates in the treatment of neurodegenerative diseases. It also elucidates the fascinating interrelationships between toxic proteins and the process of autophagy of "chasing and escaping" phenomenon. Moreover, the review further discusses the progress utilizing small molecules to activate autophagy to improve the efficacy of therapies for neurodegenerative diseases by removing toxic protein aggregates.
Collapse
Affiliation(s)
- Yuqi Fu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; School of Pharmaceutical Sciences of Medical School, Shenzhen University, Shenzhen, China
| | - Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueting Ren
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Brain Science, Faculty of Medicine, Imperial College, London, UK
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Bo Liu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
5
|
Bashir B, Vishwas S, Gupta G, Paudel KR, Dureja H, Kumar P, Cho H, Sugandhi VV, Kumbhar PS, Disouza J, Dhanasekaran M, Goh BH, Gulati M, Dua K, Singh SK. Does drug repurposing bridge the gaps in management of Parkinson's disease? Unravelling the facts and fallacies. Ageing Res Rev 2025; 105:102693. [PMID: 39961372 DOI: 10.1016/j.arr.2025.102693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
Repurposing the existing drugs for the management of both common and rare diseases is increasingly appealing due to challenges such as high attrition rates, the economy, and the slow pace of discovering new drugs. Drug repurposing involves the utilization of existing medications to treat diseases for which they were not originally intended. Despite encountering scientific and economic challenges, the pharmaceutical industry is intrigued by the potential to uncover new indications for medications. Medication repurposing is applicable across different stages of drug development, with the greatest potential observed when the drug has undergone prior safety testing. In this review, strategies for repurposing drugs for Parkinson's disease (PD) are outlined, a neurodegenerative disorder predominantly impacting dopaminergic neurons in the substantia nigra pars compacta region. PD is a debilitating neurodegenerative condition marked by an amalgam of motor and non-motor symptoms. Despite the availability of certain symptomatic treatments, particularly targeting motor symptoms, there remains a lack of established drugs capable of modifying the clinical course of PD, leading to its unchecked progression. Although standard drug discovery initiatives focusing on treatments that relieve diseases have yielded valuable understanding into the underlying mechanisms of PD, none of the numerous promising candidates identified in preclinical studies have successfully transitioned into clinically effective medications. Due to the substantial expenses associated with drug discovery endeavors, it is understandable that there has been a notable shift towards drug reprofiling strategies. Assessing the efficacy of an existing medication offers the additional advantage of circumventing the requirement for preclinical safety assessments and formulation enhancements, consequently streamlining the process and reducing both the duration of time and financial investments involved in bringing a treatment to clinical fruition. Furthermore, repurposed drugs may benefit from lower rates of failure, presenting an additional potential advantage. Various strategies for repurposing drugs are available to researchers in the field of PD. Some of these strategies have demonstrated effectiveness in identifying appropriate drugs for clinical trials, thereby providing validation for such strategies. This review provides an overview of the diverse strategies employed for drug reprofiling from approaches that place emphasis on single-gene transcriptional investigations to comprehensive epidemiological correlation analysis. Additionally, instances of previous or current research endeavors employing each strategy have been discussed. For the strategies that have not been yet implemented in PD research, their strategic efficacy is demonstrated using examples involving other disorders. In this review, we assess the safety and efficacy potential of prominent candidates repurposed as potential treatments for modifying the course of PD undergoing advanced clinical trials.
Collapse
Affiliation(s)
- Bushra Bashir
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Keshav Raj Paudel
- Centre of Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia
| | - Harish Dureja
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana 124001, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Punjab, India
| | - Hyunah Cho
- College of Pharmacy & Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Vrashabh V Sugandhi
- College of Pharmacy & Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Popat S Kumbhar
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Kolhapur, Maharashtra, 416113, India.
| | - John Disouza
- Bombay Institute of Pharmacy and Research, Dombivli, Mumbai, Maharashtra, 421 203, India..
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University Auburn, AL 36849, USA
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India; Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Sunway, Malaysia.
| |
Collapse
|
6
|
Gurra P, Babu R, Pancholi B, Mohanta BC, Garabadu D. Current opinion on pluripotent stem cell technology in Gaucher's disease: challenges and future prospects. Cytotechnology 2025; 77:26. [PMID: 39735330 PMCID: PMC11680541 DOI: 10.1007/s10616-024-00687-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024] Open
Abstract
Gaucher's disease (GD) is a rare autosomal recessive genetic disorder caused by mutations in the GBA1 gene. Mutations in the gene lead to the deficiency of glucocerebrosidase, an enzyme that helps in the breakdown of glucosylceramide (GlcCer) into ceramide and glucose. The lack of the enzyme causes GlcCer accumulation in macrophages, resulting in various phenotypic characteristics of GD. The currently available therapies, including enzyme replacement therapy and substrate reduction therapy, only provide symptomatic relief. However, they grapple with limitations in efficacy, accessibility, and potential side effects. These observations laid the foundation to search for new approaches in the management of GD. Induced pluripotent stem cells (iPSCs) technology emerges as a beacon of hope, offering novel avenues for future GD therapies. The true magic of iPSCs lies in their ability to differentiate into various cell types. By reprogramming patient-derived cells into iPSCs, researchers can generate personalized models that recapitulate the genetic and phenotypic characteristics of the GD. These models are valuable tools for dissecting intricate disease pathways, developing novel therapeutic targets, and enhancing the drug development process for GD. This review emphasizes the significance of iPSCs technology in GD management. Further, it addresses several challenges that are being encountered in the application of iPSC technology in the management of GD. In addition, it provides several insights into the future aspects of iPSC technology in the management of GD.
Collapse
Affiliation(s)
- Pankaj Gurra
- Department of Pharmacy, Central University of South Bihar, Gaya, 824236 India
| | - Raja Babu
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151001 India
| | - Bhaskaranand Pancholi
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151001 India
| | | | - Debapriya Garabadu
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151001 India
| |
Collapse
|
7
|
Menozzi E, Schapira AHV. Prospects for Disease Slowing in Parkinson Disease. Annu Rev Pharmacol Toxicol 2025; 65:237-258. [PMID: 39088860 DOI: 10.1146/annurev-pharmtox-022124-033653] [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: 08/03/2024]
Abstract
The increasing prevalence of Parkinson disease (PD) highlights the need to develop interventions aimed at slowing or halting its progression. As a result of sophisticated disease modeling in preclinical studies, and refinement of specific clinical/genetic/pathological profiles, our understanding of PD pathogenesis has grown over the years, leading to the identification of several targets for disease modification. This has translated to the development of targeted therapies, many of which have entered clinical trials. Nonetheless, up until now, none of these treatments have satisfactorily shown disease-modifying effects in PD. In this review, we present the most up-to-date disease-modifying pharmacological interventions in the clinical trial pipeline for PD. We focus on agents that have reached more advanced stages of clinical trials testing, highlighting both positive and negative results, and critically reflect on strengths, weaknesses, and challenges of current disease-modifying therapeutic avenues in PD.
Collapse
Affiliation(s)
- Elisa Menozzi
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom;
| | - Anthony H V Schapira
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom;
| |
Collapse
|
8
|
Jiao F, Meng L, Du K, Li X. The autophagy-lysosome pathway: a potential target in the chemical and gene therapeutic strategies for Parkinson's disease. Neural Regen Res 2025; 20:139-158. [PMID: 38767483 PMCID: PMC11246151 DOI: 10.4103/nrr.nrr-d-23-01195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 11/14/2023] [Accepted: 12/06/2023] [Indexed: 05/22/2024] Open
Abstract
Parkinson's disease is a common neurodegenerative disease with movement disorders associated with the intracytoplasmic deposition of aggregate proteins such as α-synuclein in neurons. As one of the major intracellular degradation pathways, the autophagy-lysosome pathway plays an important role in eliminating these proteins. Accumulating evidence has shown that upregulation of the autophagy-lysosome pathway may contribute to the clearance of α-synuclein aggregates and protect against degeneration of dopaminergic neurons in Parkinson's disease. Moreover, multiple genes associated with the pathogenesis of Parkinson's disease are intimately linked to alterations in the autophagy-lysosome pathway. Thus, this pathway appears to be a promising therapeutic target for treatment of Parkinson's disease. In this review, we briefly introduce the machinery of autophagy. Then, we provide a description of the effects of Parkinson's disease-related genes on the autophagy-lysosome pathway. Finally, we highlight the potential chemical and genetic therapeutic strategies targeting the autophagy-lysosome pathway and their applications in Parkinson's disease.
Collapse
Affiliation(s)
- Fengjuan Jiao
- School of Mental Health, Jining Medical University, Jining, Shandong Province, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, Shandong Province, China
| | - Lingyan Meng
- School of Mental Health, Jining Medical University, Jining, Shandong Province, China
| | - Kang Du
- School of Mental Health, Jining Medical University, Jining, Shandong Province, China
| | - Xuezhi Li
- School of Mental Health, Jining Medical University, Jining, Shandong Province, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, Shandong Province, China
| |
Collapse
|
9
|
Lin SC, Chang CC, Tsou SH, Chiu PY, Cheng JF, Hung HC, Chen WJ, Ho YJ, Lin CL. Protective Effects of Ambroxol on Aβ and α-Synuclein-Induced Neurotoxicity Through Glucocerebrosidase Activation in HT-22 Hippocampal Neuronal Cells. Int J Mol Sci 2024; 25:12114. [PMID: 39596182 PMCID: PMC11593818 DOI: 10.3390/ijms252212114] [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: 10/15/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Dementia with Lewy bodies (DLB) is a progressive neurodegenerative disorder marked by the accumulation of α-synuclein (αSyn), often co-existing with amyloid β (Aβ) pathology. Current treatments are largely symptomatic, highlighting a critical need for disease-modifying therapies. Evidence suggests that αSyn aggregates contribute to neuronal death in DLB, particularly when exacerbated by Aβ. Given the role of autophagy in clearing misfolded proteins, exploring agents that promote this pathway is essential for developing effective treatments. Ambroxol (AMBX), a mucolytic drug, has demonstrated potential in activating glucocerebrosidase (GCase), an enzyme that enhances lysosomal function and facilitates the autophagic clearance of toxic protein aggregates, including αSyn. This study aims to evaluate AMBX's neuroprotective effects in a cellular model of DLB, with the goal of identifying new therapeutic agents that target the underlying pathology of DLB. In this study, HT-22 hippocampal neuronal cells were exposed to αSyn and Aβ, followed by AMBX treatment. Our results showed that AMBX significantly improved cell viability and reduced apoptosis in cells co-treated with αSyn and Aβ. Additionally, AMBX restored GCase activity, promoted autophagy, and reduced oxidative stress, which in turn mitigated αSyn aggregation and phosphorylation. These findings suggest that by activating GCase and enhancing autophagy, AMBX may help alleviate DLB-associated neurodegeneration. This study underscores the potential of AMBX as a therapeutic agent for DLB and supports further investigation in animal models and clinical trials to validate its efficacy in neurodegenerative disease contexts.
Collapse
Affiliation(s)
- Sheng-Chieh Lin
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (S.-C.L.); (C.-C.C.)
- Department of Orthopedics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Ching-Chi Chang
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (S.-C.L.); (C.-C.C.)
- Department of Psychiatry, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Sing-Hua Tsou
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan;
| | - Pai-Yi Chiu
- Department of Neurology, Show Chwan Memorial Hospital, Changhua 500, Taiwan;
| | - Ju-Fang Cheng
- Division of Pediatric Neurology, Changhua Christian Hospital, Changhua 500, Taiwan;
| | - Hui-Chih Hung
- Department of Life Sciences and Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan;
| | - Wei-Jen Chen
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung 402, Taiwan;
| | - Ying-Jui Ho
- Department of Psychology, Chung Shan Medical University, Taichung 402, Taiwan
| | - Chih-Li Lin
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan;
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| |
Collapse
|
10
|
Patzwaldt K, Castaneda-Vega S. Ambroxol, the cough expectorant with neuroprotective effects. Neural Regen Res 2024; 19:2345-2346. [PMID: 38526267 PMCID: PMC11090448 DOI: 10.4103/nrr.nrr-d-23-01664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/13/2023] [Accepted: 12/23/2023] [Indexed: 03/26/2024] Open
Affiliation(s)
- Kristin Patzwaldt
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Salvador Castaneda-Vega
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Clinic Tuebingen, Tuebingen, Germany
| |
Collapse
|
11
|
Kuppuramalingam A, Cabasso O, Horowitz M. Functional Analysis of Human GBA1 Missense Mutations in Drosophila: Insights into Gaucher Disease Pathogenesis and Phenotypic Consequences. Cells 2024; 13:1619. [PMID: 39404383 PMCID: PMC11475061 DOI: 10.3390/cells13191619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
The human GBA1 gene encodes lysosomal acid β-glucocerebrosidase, whose activity is deficient in Gaucher disease (GD). In Drosophila, there are two GBA1 orthologs, Gba1a and Gba1b, and Gba1b is the bona fide GCase encoding gene. Several fly lines with different deletions in the Gba1b were studied in the past. However, since most GD-associated GBA1 mutations are point mutations, we created missense mutations homologous to the two most common GD mutations: the mild N370S mutation (D415S in Drosophila) and the severe L444P mutation (L494P in Drosophila), using the CRISPR-Cas9 technology. Flies homozygous for the D415S mutation (dubbed D370S hereafter) presented low GCase activity and substrate accumulation, which led to lysosomal defects, activation of the Unfolded Protein Response (UPR), inflammation/neuroinflammation, and neurodegeneration along with earlier death compared to control flies. Surprisingly, the L494P (called L444P hereafter) flies presented higher GCase activity with fewer lysosomal defects and milder disease in comparison to that presented by the D370S homozygous flies. Treatment with ambroxol had a limited effect on all homozygous fly lines tested. Overall, our results underscore the differences between the fly and human GCase enzymes, as evidenced by the distinct phenotypic outcomes of mutations in flies compared to those observed in human GD patients.
Collapse
Affiliation(s)
- Aparna Kuppuramalingam
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (A.K.); (O.C.)
| | - Or Cabasso
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (A.K.); (O.C.)
| | - Mia Horowitz
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (A.K.); (O.C.)
- Sagol School of Neuroscience, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
12
|
Zhang X, Wu H, Tang B, Guo J. Clinical, mechanistic, biomarker, and therapeutic advances in GBA1-associated Parkinson's disease. Transl Neurodegener 2024; 13:48. [PMID: 39267121 PMCID: PMC11391654 DOI: 10.1186/s40035-024-00437-6] [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: 01/29/2024] [Accepted: 08/17/2024] [Indexed: 09/14/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. The development of PD is closely linked to genetic and environmental factors, with GBA1 variants being the most common genetic risk. Mutations in the GBA1 gene lead to reduced activity of the coded enzyme, glucocerebrosidase, which mediates the development of PD by affecting lipid metabolism (especially sphingolipids), lysosomal autophagy, endoplasmic reticulum, as well as mitochondrial and other cellular functions. Clinically, PD with GBA1 mutations (GBA1-PD) is characterized by particular features regarding the progression of symptom severity. On the therapeutic side, the discovery of the relationship between GBA1 variants and PD offers an opportunity for targeted therapeutic interventions. In this review, we explore the genotypic and phenotypic correlations, etiologic mechanisms, biomarkers, and therapeutic approaches of GBA1-PD and summarize the current state of research and its challenges.
Collapse
Affiliation(s)
- Xuxiang Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Heng Wu
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, University of South China, Hengyang, 421001, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province, Hengyang, 421001, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, University of South China, Hengyang, 421001, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China.
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China.
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, China.
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
13
|
Dai L, Liu M, Ke W, Chen L, Fang X, Zhang Z. Lysosomal dysfunction in α-synuclein pathology: molecular mechanisms and therapeutic strategies. Cell Mol Life Sci 2024; 81:382. [PMID: 39223418 PMCID: PMC11368888 DOI: 10.1007/s00018-024-05419-5] [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: 05/15/2024] [Revised: 08/09/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
In orchestrating cell signaling, facilitating plasma membrane repair, supervising protein secretion, managing waste elimination, and regulating energy consumption, lysosomes are indispensable guardians that play a crucial role in preserving intracellular homeostasis. Neurons are terminally differentiated post-mitotic cells. Neuronal function and waste elimination depend on normal lysosomal function. Converging data suggest that lysosomal dysfunction is a critical event in the etiology of Parkinson's disease (PD). Mutations in Glucosylceramidase Beta 1 (GBA1) and leucine-rich repeat kinase 2 (LRRK2) confer an increased risk for the development of parkinsonism. Furthermore, lysosomal dysfunction has been observed in the affected neurons of sporadic PD (sPD) patients. Given that lysosomal hydrolases actively contribute to the breakdown of impaired organelles and misfolded proteins, any compromise in lysosomal integrity could incite abnormal accumulation of proteins, including α-synuclein, the major component of Lewy bodies in PD. Clinical observations have shown that lysosomal protein levels in cerebrospinal fluid may serve as potential biomarkers for PD diagnosis and as signs of lysosomal dysfunction. In this review, we summarize the current evidence regarding lysosomal dysfunction in PD and discuss the intimate relationship between lysosomal dysfunction and pathological α-synuclein. In addition, we discuss therapeutic strategies that target lysosomes to treat PD.
Collapse
Affiliation(s)
- Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Miao Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wei Ke
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liam Chen
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Xin Fang
- Department of Neurology, the First Affiliated Hospital of Nanchang University, Nanchang, 330000, China.
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- TaiKang Center for Life and Medical Science, Wuhan University, Wuhan, 430000, China.
| |
Collapse
|
14
|
Cyske Z, Gaffke L, Rintz E, Wiśniewska K, Węgrzyn G, Pierzynowska K. Molecular mechanisms of the ambroxol action in Gaucher disease and GBA1 mutation-associated Parkinson disease. Neurochem Int 2024; 178:105774. [PMID: 38797393 DOI: 10.1016/j.neuint.2024.105774] [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: 12/26/2023] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Glucocerebrosidase (GCase), encoded by the GBA1 gene, is one of the lysosomal enzymes responsible for hydrolyzing the glycosphingolipids. Deficiency in GCase activity (in patients with two defective alleles of GBA1) leads to glucosylceramide storage in lysosomes which in turn results in the development of the Gaucher diseases, a lysosomal storage disorder, while a heterozygous state may be correlated with the GBA1 mutation-associated Parkinson disease. One of the proposed forms of therapy for these two conditions is the use of pharmacological chaperones which work by facilitating the achievement of the correct conformation of abnormally folded enzymes. Several compounds with chaperone activities against GCase have already been tested, one of which turned out to be ambroxol. Studies conducted on the action of this compound have indeed indicated its effectiveness in increasing GCase levels and activity. However, some data have begun to question its activity as a chaperone against certain GCase variants. Then, a number of articles appeared pointing to other mechanisms of action of ambroxol, which may also contribute to the improvement of patients' condition. This paper summarizes the biological mechanisms of action of ambroxol in Gaucher disease and GBA1 mutation-associated Parkinson disease, focused on its activity as a chaperone, modulator of ERAD pathways, inducer of autophagy, and pain reliever in cellular and animal models as well as in patients. The effects of these activities on the reduction of disease markers and symptoms in patients are also discussed. Consideration of all the properties of ambroxol can help in the appropriate choice of therapy and the determination of the effective drug dose.
Collapse
Affiliation(s)
- Zuzanna Cyske
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Lidia Gaffke
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Estera Rintz
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Karolina Wiśniewska
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Karolina Pierzynowska
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland.
| |
Collapse
|
15
|
den Hollander B, Le HL, Swart EL, Bikker H, Hollak CEM, Brands MM. Clinical and preclinical insights into high-dose ambroxol therapy for Gaucher disease type 2 and 3: A comprehensive systematic review. Mol Genet Metab 2024; 143:108556. [PMID: 39116528 DOI: 10.1016/j.ymgme.2024.108556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/19/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024]
Abstract
RATIONALE Gaucher disease (GD), an autosomal recessive lysosomal storage disease, results from GBA1 variants causing glucocerebrosidase (GCase) deficiency. While enzyme replacement therapy (ERT) helps with systemic symptoms, neurological complications in GD2 and GD3 persist due to the blood-brain-barrier (BBB) limiting ERT efficacy. Ambroxol, a BBB-permeable chaperone, enhances GCase activity. Our review explores high-dose ambroxol's therapeutic potential, both preclinical and clinical, in GD2 and GD3. METHODS PubMed was searched for studies published before March 2023, including clinical, animal, and in vitro studies focusing on the effect of high-dose ambroxol in GD2 and GD3. A narrative synthesis was performed. RESULTS Nine in vitro, three animal, and eight clinical studies were included, demonstrating varied responses to ambroxol across diverse outcome measures. In vitro and animal studies demonstrated reduced endoplasmatic reticulum stress due to the relocation of GCase from the ER to the lysosomes. In vitro cell lines exhibited varying degrees of increased GCase activity. Clinical trials observed reduced lyso-GL1 levels in plasma (41-89%) and cerebrospinal fluid (CSF) (26-97%), alongside increased GCase activity in GD3 patients. Ambroxol exhibited varying effects on neurological outcomes and development. No severe adverse events were reported. CONCLUSION High-dose ambroxol shows promise in managing neurological manifestations in GD3, albeit with uncertainties resulting from genetic heterogeneity and variable response. Further clinical trials, are essential for elucidating dosage-response relationships and refining treatment outcomes and strategies for neuronopathic GD.
Collapse
Affiliation(s)
- Bibiche den Hollander
- Amsterdam UMC location University of Amsterdam, Department of Pediatrics, Emma Children's Hospital, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC, Emma Center for Personalized Medicine, Amsterdam, the Netherlands; United for Metabolic Diseases, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of Metabolism, Amsterdam, the Netherlands
| | - Hoang Lan Le
- Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of Metabolism, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Clinical Pharmacology and Pharmacy, Meibergdreef 9, Amsterdam, the Netherlands
| | - Eleonora L Swart
- Amsterdam UMC location University of Amsterdam, Department of Clinical Pharmacology and Pharmacy, Meibergdreef 9, Amsterdam, the Netherlands
| | - Hennie Bikker
- Amsterdam UMC location University of Amsterdam, Department of Human Genetics, Meibergdreef 9, Amsterdam, the Netherlands
| | - Carla E M Hollak
- Amsterdam UMC location University of Amsterdam, Department of Endocrinology and Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - Marion M Brands
- Amsterdam UMC location University of Amsterdam, Department of Pediatrics, Emma Children's Hospital, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC, Emma Center for Personalized Medicine, Amsterdam, the Netherlands; United for Metabolic Diseases, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of Metabolism, Amsterdam, the Netherlands; Amsterdam Reproduction & Development Research Institute, Amsterdam, the Netherlands.
| |
Collapse
|
16
|
Rubilar JC, Outeiro TF, Klein AD. The lysosomal β-glucocerebrosidase strikes mitochondria: implications for Parkinson's therapeutics. Brain 2024; 147:2610-2620. [PMID: 38437875 DOI: 10.1093/brain/awae070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/31/2024] [Accepted: 02/14/2024] [Indexed: 03/06/2024] Open
Abstract
Parkinson's disease is a neurodegenerative disorder primarily known for typical motor features that arise due to the loss of dopaminergic neurons in the substantia nigra. However, the precise molecular aetiology of the disease is still unclear. Several cellular pathways have been linked to Parkinson's disease, including the autophagy-lysosome pathway, α-synuclein aggregation and mitochondrial function. Interestingly, the mechanistic link between GBA1, the gene that encodes for lysosomal β-glucocerebrosidase (GCase), and Parkinson's disease lies in the interplay between GCase functions in the lysosome and mitochondria. GCase mutations alter mitochondria-lysosome contact sites. In the lysosome, reduced GCase activity leads to glycosphingolipid build-up, disrupting lysosomal function and autophagy, thereby triggering α-synuclein accumulation. Additionally, α-synuclein aggregates reduce GCase activity, creating a self-perpetuating cycle of lysosomal dysfunction and α-synuclein accumulation. GCase can also be imported into the mitochondria, where it promotes the integrity and function of mitochondrial complex I. Thus, GCase mutations that impair its normal function increase oxidative stress in mitochondria, the compartment where dopamine is oxidized. In turn, the accumulation of oxidized dopamine adducts further impairs GCase activity, creating a second cycle of GCase dysfunction. The oxidative state triggered by GCase dysfunction can also induce mitochondrial DNA damage which, in turn, can cause dopaminergic cell death. In this review, we highlight the pivotal role of GCase in Parkinson's disease pathogenesis and discuss promising examples of GCase-based therapeutics, such as gene and enzyme replacement therapies, small molecule chaperones and substrate reduction therapies, among others, as potential therapeutic interventions.
Collapse
Affiliation(s)
- Juan Carlos Rubilar
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7780272, Chile
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073, Göttingen, Germany
- Max Planck Institute for Natural Sciences, 37073, Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
- Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 37075, Göttingen, Germany
| | - Andrés D Klein
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7780272, Chile
| |
Collapse
|
17
|
Ullah I, Wang X, Li H. Novel and experimental therapeutics for the management of motor and non-motor Parkinsonian symptoms. Neurol Sci 2024; 45:2979-2995. [PMID: 38388896 DOI: 10.1007/s10072-023-07278-7] [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: 10/25/2023] [Accepted: 12/14/2023] [Indexed: 02/24/2024]
Abstract
BACKGROUND : Both motor and non-motor symptoms of Parkinson's disease (PD) have a substantial detrimental influence on the patient's quality of life. The most effective treatment remains oral levodopa. All currently known treatments just address the symptoms; they do not completely reverse the condition. METHODOLOGY In order to find literature on the creation of novel treatment agents and their efficacy for PD patients, we searched PubMed, Google Scholar, and other online libraries. RESULTS According to the most recent study on Parkinson's disease (PD), a great deal of work has been done in both the clinical and laboratory domains, and some current scientists have even been successful in developing novel therapies for PD patients. CONCLUSION The quality of life for PD patients has increased as a result of recent research, and numerous innovative medications are being developed for PD therapy. In the near future, we will see positive outcomes regarding PD treatment.
Collapse
Affiliation(s)
- Inam Ullah
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Xin Wang
- School of Pharmacy, Lanzhou University, Lanzhou, China.
| | - Hongyu Li
- School of Life Sciences, Lanzhou University, Lanzhou, China.
| |
Collapse
|
18
|
Skrahin A, Horowitz M, Istaiti M, Skrahina V, Lukas J, Yahalom G, Cohen ME, Revel-Vilk S, Goker-Alpan O, Becker-Cohen M, Hassin-Baer S, Svenningsson P, Rolfs A, Zimran A. GBA1-Associated Parkinson's Disease Is a Distinct Entity. Int J Mol Sci 2024; 25:7102. [PMID: 39000225 PMCID: PMC11241486 DOI: 10.3390/ijms25137102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
GBA1-associated Parkinson's disease (GBA1-PD) is increasingly recognized as a distinct entity within the spectrum of parkinsonian disorders. This review explores the unique pathophysiological features, clinical progression, and genetic underpinnings that differentiate GBA1-PD from idiopathic Parkinson's disease (iPD). GBA1-PD typically presents with earlier onset and more rapid progression, with a poor response to standard PD medications. It is marked by pronounced cognitive impairment and a higher burden of non-motor symptoms compared to iPD. Additionally, patients with GBA1-PD often exhibit a broader distribution of Lewy bodies within the brain, accentuating neurodegenerative processes. The pathogenesis of GBA1-PD is closely associated with mutations in the GBA1 gene, which encodes the lysosomal enzyme beta-glucocerebrosidase (GCase). In this review, we discuss two mechanisms by which GBA1 mutations contribute to disease development: 'haploinsufficiency,' where a single functional gene copy fails to produce a sufficient amount of GCase, and 'gain of function,' where the mutated GCase acquires harmful properties that directly impact cellular mechanisms for alpha-synuclein degradation, leading to alpha-synuclein aggregation and neuronal cell damage. Continued research is advancing our understanding of how these mechanisms contribute to the development and progression of GBA1-PD, with the 'gain of function' mechanism appearing to be the most plausible. This review also explores the implications of GBA1 mutations for therapeutic strategies, highlighting the need for early diagnosis and targeted interventions. Currently, small molecular chaperones have shown the most promising clinical results compared to other agents. This synthesis of clinical, pathological, and molecular aspects underscores the assertion that GBA1-PD is a distinct clinical and pathobiological PD phenotype, necessitating specific management and research approaches to better understand and treat this debilitating condition.
Collapse
Affiliation(s)
- Aliaksandr Skrahin
- Rare Disease Consulting RCV GmbH, Leibnizstrasse 58, 10629 Berlin, Germany
| | - Mia Horowitz
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, 6997801 Ramat Aviv, Israel
| | - Majdolen Istaiti
- Gaucher Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Agyany Pharma Ltd., 9695614 Jerusalem, Israel
| | | | - Jan Lukas
- Translational Neurodegeneration Section Albrecht Kossel, Department of Neurology, University Medical Center Rostock, 18147 Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
| | - Gilad Yahalom
- Department of Neurology and Movement Disorders Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| | - Mikhal E. Cohen
- Department of Neurology and Movement Disorders Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| | - Shoshana Revel-Vilk
- Gaucher Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| | - Ozlem Goker-Alpan
- Lysosomal and Rare Disorders Research and Treatment Center, Fairfax, VA 22030, USA
| | | | - Sharon Hassin-Baer
- Movement Disorders Institute, Department of Neurology, Chaim Sheba Medical Center, 5262101 Tel-Hashomer, Israel
- Department of Neurology and Neurosurgery, Faculty of Medical and Health Sciences, Tel Aviv University, 6997801 Tel-Aviv, Israel
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
- Department of Basal and Clinical Neuroscience, King’s College London, London SE5 9RT, UK
| | - Arndt Rolfs
- Rare Disease Consulting RCV GmbH, Leibnizstrasse 58, 10629 Berlin, Germany
- Agyany Pharma Ltd., 9695614 Jerusalem, Israel
- Medical Faculty, University of Rostock, 18055 Rostock, Germany
| | - Ari Zimran
- Gaucher Unit, Shaare Zedek Medical Center, 9103102 Jerusalem, Israel
- Agyany Pharma Ltd., 9695614 Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102 Jerusalem, Israel
| |
Collapse
|
19
|
Siemeling O, Slingerland S, van der Zee S, van Laar T. Study protocol of the GRoningen early-PD Ambroxol treatment (GREAT) trial: a randomized, double-blind, placebo-controlled, single center trial with ambroxol in Parkinson patients with a GBA mutation. BMC Neurol 2024; 24:146. [PMID: 38693511 PMCID: PMC11061939 DOI: 10.1186/s12883-024-03629-9] [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: 02/06/2024] [Accepted: 04/08/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND To date, no disease modifying therapies are available for Parkinson's disease (PD). Since PD is the second most prevalent neurodegenerative disorder, there is a high demand for such therapies. Both environmental and genetic risk factors play an important role in the etiology and progression of PD. The most common genetic risk factor for PD is a mutation in the GBA1(GBA)-gene, encoding the lysosomal enzyme glucocerebrosidase (GCase). The mucolytic ambroxol is a repurposed drug, which has shown the property to upregulate GCase activity in-vitro and in-vivo. Ambroxol therefore has the potency to become a disease modifying therapy in PD, which was the reason to design this randomized controlled trial with ambroxol in PD patients. METHODS This trial is a single-center, double-blind, randomized, placebo-controlled study, including 80 PD patients with a GBA mutation, receiving either ambroxol 1800 mg/day or placebo for 48 weeks. The primary outcome measure is the Unified Parkinson's Disease Rating Scale motor subscore (part III) of the Movement Disorder Society (MDS-UPDRSIII) in the practically defined off-state at 60 weeks (after a 12-week washout period). Secondary outcomes include a 3,4-dihydroxy-6-18F-fluoro-I-phenylalanine ([18F]FDOPA) PET-scan of the brain, Magnetic Resonance Imaging (with resting state f-MRI and Diffusion Tensor Imaging), GCase activity, both intra- and extracellularly, sphingolipid profiles in plasma, Montreal Cognitive Assessment (MoCA), quality of life (QoL) measured by the Parkinson's Disease Questionnaire (PDQ-39) and the Non-Motor Symptom Scale (NMSS) questionnaire. DISCUSSION Ambroxol up to 1200 mg/day has shown effects on human cerebrospinal fluid endpoints, which supports at least passage of the blood-brain-barrier. The dose titration in this trial up to 1800 mg/day will reveal if this dose level is safe and also effective in modifying the course of the disease. TRIAL REGISTRATION NCT05830396. Registration date: March 20, 2023.
Collapse
Affiliation(s)
- O Siemeling
- Department of Neurology, University Medical Center Groningen, Groningen, The Netherlands.
- Parkinson Expertise Center Groningen, Groningen, The Netherlands.
| | - S Slingerland
- Department of Neurology, University Medical Center Groningen, Groningen, The Netherlands
- Parkinson Expertise Center Groningen, Groningen, The Netherlands
| | - S van der Zee
- Department of Neurology, University Medical Center Groningen, Groningen, The Netherlands
- Parkinson Expertise Center Groningen, Groningen, The Netherlands
| | - T van Laar
- Department of Neurology, University Medical Center Groningen, Groningen, The Netherlands
- Parkinson Expertise Center Groningen, Groningen, The Netherlands
| |
Collapse
|
20
|
Hull A, Atilano ML, Gergi L, Kinghorn KJ. Lysosomal storage, impaired autophagy and innate immunity in Gaucher and Parkinson's diseases: insights for drug discovery. Philos Trans R Soc Lond B Biol Sci 2024; 379:20220381. [PMID: 38368939 PMCID: PMC10874704 DOI: 10.1098/rstb.2022.0381] [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: 03/08/2023] [Accepted: 11/08/2023] [Indexed: 02/20/2024] Open
Abstract
Impairment of autophagic-lysosomal pathways is increasingly being implicated in Parkinson's disease (PD). GBA1 mutations cause the lysosomal storage disorder Gaucher disease (GD) and are the commonest known genetic risk factor for PD. GBA1 mutations have been shown to cause autophagic-lysosomal impairment. Defective autophagic degradation of unwanted cellular constituents is associated with several pathologies, including loss of normal protein homeostasis, particularly of α-synuclein, and innate immune dysfunction. The latter is observed both peripherally and centrally in PD and GD. Here, we will discuss the mechanistic links between autophagy and immune dysregulation, and the possible role of these pathologies in communication between the gut and brain in these disorders. Recent work in a fly model of neuronopathic GD (nGD) revealed intestinal autophagic defects leading to gastrointestinal dysfunction and immune activation. Rapamycin treatment partially reversed the autophagic block and reduced immune activity, in association with increased survival and improved locomotor performance. Alterations in the gut microbiome are a critical driver of neuroinflammation, and studies have revealed that eradication of the microbiome in nGD fly and mouse models of PD ameliorate brain inflammation. Following these observations, lysosomal-autophagic pathways, innate immune signalling and microbiome dysbiosis are discussed as potential therapeutic targets in PD and GD. This article is part of a discussion meeting issue 'Understanding the endo-lysosomal network in neurodegeneration'.
Collapse
Affiliation(s)
- Alexander Hull
- Department of Genetics, Evolution & Environment, Institute of Healthy Ageing, Darwin Building, Gower Street, London WC1E 6BT, UK
| | - Magda L Atilano
- Department of Genetics, Evolution & Environment, Institute of Healthy Ageing, Darwin Building, Gower Street, London WC1E 6BT, UK
| | - Laith Gergi
- Department of Genetics, Evolution & Environment, Institute of Healthy Ageing, Darwin Building, Gower Street, London WC1E 6BT, UK
| | - Kerri J Kinghorn
- Department of Genetics, Evolution & Environment, Institute of Healthy Ageing, Darwin Building, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
21
|
Schulze MSED, Scholz D, Jnoff E, Hall A, Melin J, Sands ZA, Rodriguez E, Andre VM. Identification of ß-Glucocerebrosidase Activators for Glucosylceramide hydrolysis. ChemMedChem 2024; 19:e202300548. [PMID: 38381042 DOI: 10.1002/cmdc.202300548] [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: 10/10/2023] [Revised: 01/31/2024] [Indexed: 02/22/2024]
Abstract
Several novel chemical series were identified that modulate glucocerebrosidase (GCase). Compounds from these series are active on glucosylceramide, unlike other known GCase modulators. We obtained GCase crystal structures with two compounds that have distinct chemotypes. Positive allosteric modulators bind to a site on GCase and induce conformational changes, but also induce an equilibrium state between monomer and dimer.
Collapse
Affiliation(s)
| | - Diana Scholz
- UCB, Avenue de l'Industrie, Braine l'Alleud, 1420, Belgium
| | - Eric Jnoff
- UCB, Avenue de l'Industrie, Braine l'Alleud, 1420, Belgium
| | - Adrian Hall
- UCB, Avenue de l'Industrie, Braine l'Alleud, 1420, Belgium
| | - Jonathan Melin
- Present address: Grünenthal GmbH, 52099, Aachen, Germany
| | - Zara A Sands
- Present address: Eli Lilly, San Diego, CA92121, USA
| | | | | |
Collapse
|
22
|
Brazdis RM, von Zimmermann C, Lenz B, Kornhuber J, Mühle C. Peripheral Upregulation of Parkinson's Disease-Associated Genes Encoding α-Synuclein, β-Glucocerebrosidase, and Ceramide Glucosyltransferase in Major Depression. Int J Mol Sci 2024; 25:3219. [PMID: 38542193 PMCID: PMC10970259 DOI: 10.3390/ijms25063219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 04/04/2024] Open
Abstract
Due to the high comorbidity of Parkinson's disease (PD) with major depressive disorder (MDD) and the involvement of sphingolipids in both conditions, we investigated the peripheral expression levels of three primarily PD-associated genes: α-synuclein (SNCA), lysosomal enzyme β-glucocerebrosidase (GBA1), and UDP-glucose ceramide glucosyltransferase (UGCG) in a sex-balanced MDD cohort. Normalized gene expression was determined by quantitative PCR in patients suffering from MDD (unmedicated n = 63, medicated n = 66) and controls (remitted MDD n = 39, healthy subjects n = 61). We observed that expression levels of SNCA (p = 0.036), GBA1 (p = 0.014), and UGCG (p = 0.0002) were higher in currently depressed patients compared to controls and remitted patients, and expression of GBA1 and UGCG decreased in medicated patients during three weeks of therapy. Additionally, in subgroups, expression was positively correlated with the severity of depression and anxiety. Furthermore, we identified correlations between the gene expression levels and PD-related laboratory parameters. Our findings suggest that SNCA, GBA1, and UGCG analysis could be instrumental in the search for biomarkers of MDD and in understanding the overlapping pathological mechanisms underlying neuro-psychiatric diseases.
Collapse
Affiliation(s)
- Razvan-Marius Brazdis
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (R.-M.B.); (B.L.); (J.K.)
| | - Claudia von Zimmermann
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (R.-M.B.); (B.L.); (J.K.)
| | - Bernd Lenz
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (R.-M.B.); (B.L.); (J.K.)
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health (CIMH), Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (R.-M.B.); (B.L.); (J.K.)
| | - Christiane Mühle
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (R.-M.B.); (B.L.); (J.K.)
| |
Collapse
|
23
|
Mohamed FE, Al-Jasmi F. Exploring the efficacy and safety of Ambroxol in Gaucher disease: an overview of clinical studies. Front Pharmacol 2024; 15:1335058. [PMID: 38414738 PMCID: PMC10896849 DOI: 10.3389/fphar.2024.1335058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/17/2024] [Indexed: 02/29/2024] Open
Abstract
Gaucher disease (GD) is mainly caused by glucocerebrosidase (GCase) enzyme deficiency due to genetic variations in the GBA1 gene leading to the toxic accumulation of sphingolipids in various organs, which causes symptoms such as anemia, thrombocytopenia, hepatosplenomegaly, and neurological manifestations. GD is clinically classified into the non-neuronopathic type 1, and the acute and chronic neuronopathic forms, types 2 and 3, respectively. In addition to the current approved GD medications, the repurposing of Ambroxol (ABX) has emerged as a prospective enzyme enhancement therapy option showing its potential to enhance mutated GCase activity and reduce glucosylceramide accumulation in GD-affected tissues of different GBA1 genotypes. The variability in response to ABX varies across different variants, highlighting the diversity in patients' therapeutic outcomes. Its oral availability and safety profile make it an attractive option, particularly for patients with neurological manifestations. Clinical trials are essential to explore further ABX's potential as a therapeutic medication for GD to encourage pharmaceutical companies' investment in its development. This review highlights the potential of ABX as a pharmacological chaperone therapy for GD and stresses the importance of addressing response variability in clinical studies to improve the management of this rare and complex disorder.
Collapse
Affiliation(s)
- Feda E. Mohamed
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Abu Dhabi, United Arab Emirates
| | - Fatma Al-Jasmi
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Abu Dhabi, United Arab Emirates
- Department of Pediatrics, Tawam Hospital, Al Ain, United Arab Emirates
| |
Collapse
|
24
|
Wyse RK, Isaacs T, Barker RA, Cookson MR, Dawson TM, Devos D, Dexter DT, Duffen J, Federoff H, Fiske B, Foltynie T, Fox S, Greenamyre JT, Kieburtz K, Kordower JH, Krainc D, Matthews H, Moore DJ, Mursaleen L, Schwarzschild MA, Stott SR, Sulzer D, Svenningsson P, Tanner CM, Carroll C, Simon DK, Brundin P. Twelve Years of Drug Prioritization to Help Accelerate Disease Modification Trials in Parkinson's Disease: The International Linked Clinical Trials Initiative. JOURNAL OF PARKINSON'S DISEASE 2024; 14:657-666. [PMID: 38578902 PMCID: PMC11191436 DOI: 10.3233/jpd-230363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/10/2024] [Indexed: 04/07/2024]
Abstract
In 2011, the UK medical research charity Cure Parkinson's set up the international Linked Clinical Trials (iLCT) committee to help expedite the clinical testing of potentially disease modifying therapies for Parkinson's disease (PD). The first committee meeting was held at the Van Andel Institute in Grand Rapids, Michigan in 2012. This group of PD experts has subsequently met annually to assess and prioritize agents that may slow the progression of this neurodegenerative condition, using a systematic approach based on preclinical, epidemiological and, where possible, clinical data. Over the last 12 years, 171 unique agents have been evaluated by the iLCT committee, and there have been 21 completed clinical studies and 20 ongoing trials associated with the initiative. In this review, we briefly outline the iLCT process as well as the clinical development and outcomes of some of the top prioritized agents. We also discuss a few of the lessons that have been learnt, and we conclude with a perspective on what the next decade may bring, including the introduction of multi-arm, multi-stage clinical trial platforms and the possibility of combination therapies for PD.
Collapse
Affiliation(s)
| | | | - Roger A. Barker
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Ted M. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David Devos
- Department of Medical Pharmacology and Neurology, University of Lille, CHU Lille, Lille Neurosciences and Cognition Inserm UMR-S-U1172, Lille, France
| | | | | | - Howard Federoff
- Henry and Susan Samueli College of Health Sciences, University of California, Irvine CA, USA
| | - Brian Fiske
- Research Programs, The Michael J. Fox Foundation for Parkinson’s Research, New York, NY, USA
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Susan Fox
- Edmond J. Safra Program in Parkinson’s Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, University Health Network, Division of Neurology, University of Toronto, Toronto, ON, Canada
| | - J. Timothy Greenamyre
- Department of Neurology, Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Karl Kieburtz
- Department of Neurology Center for Health and Technology, University of Rochester, Rochester, NY, USA
| | - Jeffrey H. Kordower
- ASU-Banner Neurodegenerative Disease Research Center and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | | | | | | | | - David Sulzer
- Department of Neurology, Columbia University, New York, NY, USA
| | | | - Caroline M. Tanner
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Camille Carroll
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - David K. Simon
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Patrik Brundin
- Neuroscience and Rare Diseases, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| |
Collapse
|
25
|
Tolea MI, Ezzeddine R, Camacho S, Galvin JE. Emerging drugs for dementia with Lewy Bodies: a review of Phase II & III trials. Expert Opin Emerg Drugs 2023; 28:167-180. [PMID: 37531299 DOI: 10.1080/14728214.2023.2244425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/04/2023]
Abstract
INTRODUCTION Despite faster cognitive decline and greater negative impact on patients and family caregivers, drug development efforts in Dementia with Lewy Bodies (DLB) fall behind those for Alzheimer's Disease (AD). Current off-label drug DLB treatment options are limited to symptomatic agents developed to address cognitive deficits in AD, motor deficits in Parkinson's Disease, or behavioral symptoms in psychiatric disease. Aided by recent improvements in DLB diagnosis, a new focus on the development of disease-modifying agents (DMA) is emerging. AREAS COVERED Driven by evidence supporting different pathological mechanisms in DLB and PDD, this review assesses the evidence on symptomatic drug treatments and describes current efforts in DMA development in DLB. Specifically, our goals were to: (1) review evidence supporting the use of symptomatic drug treatments in DLB; (2) review the current DMA pipeline in DLB with a focus on Phase II and III clinical trials; and (3) identify potential issues with the development of DMA in DLB. Included in this review were completed and ongoing drug clinical trials in DLB registered on ClinicalTrials.gov (no time limits set for the search) or disseminated at the 2023 international conference on Clinical Trials in AD. Drug clinical trials registered in non-US clinical trial registries were not included. EXPERT OPINION Adoption of current symptomatic drug treatments used off-label in DLB relied on efficacy of benefits in other disorders rather than evidence from randomized controlled clinical trials. Symptoms remain difficult to manage. Several DMA drugs are currently being evaluated as either repurposing candidates or novel small molecules. Continued improvement in methodological aspects including development of DLB-specific outcome measures and biomarkers is needed to move the field of DMA drug development forward.
Collapse
Affiliation(s)
- Magdalena I Tolea
- Comprehensive Center for Brain Health, Lewy Body Dementia Research Center of Excellence, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Reem Ezzeddine
- Comprehensive Center for Brain Health, Lewy Body Dementia Research Center of Excellence, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Simone Camacho
- Comprehensive Center for Brain Health, Lewy Body Dementia Research Center of Excellence, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - James E Galvin
- Comprehensive Center for Brain Health, Lewy Body Dementia Research Center of Excellence, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
26
|
Colucci F, Avenali M, De Micco R, Fusar Poli M, Cerri S, Stanziano M, Bacila A, Cuconato G, Franco V, Franciotta D, Ghezzi C, Gastaldi M, Elia AE, Romito L, Devigili G, Leta V, Garavaglia B, Golfrè Andreasi N, Cazzaniga F, Reale C, Galandra C, Germani G, Mitrotti P, Ongari G, Palmieri I, Picascia M, Pichiecchio A, Verri M, Esposito F, Cirillo M, Di Nardo F, Aloisio S, Siciliano M, Prioni S, Amami P, Piacentini S, Bruzzone MG, Grisoli M, Moda F, Eleopra R, Tessitore A, Valente EM, Cilia R. Ambroxol as a disease-modifying treatment to reduce the risk of cognitive impairment in GBA-associated Parkinson's disease: a multicentre, randomised, double-blind, placebo-controlled, phase II trial. The AMBITIOUS study protocol. BMJ Neurol Open 2023; 5:e000535. [PMID: 38027469 PMCID: PMC10679992 DOI: 10.1136/bmjno-2023-000535] [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: 09/12/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Background Heterozygous mutations in the GBA gene, encoding the lysosomal enzyme β-glucocerebrosidase (GCase), are the most frequent genetic risk factor for Parkinson's disease (PD). GBA-related PD (GBA-PD) patients have higher risk of dementia and reduced survival than non-carriers. Preclinical studies and one open-label trial in humans demonstrated that the chaperone ambroxol (ABX) increases GCase levels and modulates α-synuclein levels in the blood and cerebrospinal fluid (CSF). Methods and analysis In this multicentre, double-blind, placebo-controlled, phase II clinical trial, we randomise patients with GBA-PD in a 1:1 ratio to either oral ABX 1.2 g/day or placebo. The duration of treatment is 52 weeks. Each participant is assessed at baseline and weeks 12, 26, 38, 52 and 78. Changes in the Montreal Cognitive Assessment score and the frequency of mild cognitive impairment and dementia between baseline and weeks 52 are the primary outcome measures. Secondary outcome measures include changes in validated scales/questionnaires assessing motor and non-motor symptoms. Neuroimaging features and CSF neurodegeneration markers are used as surrogate markers of disease progression. GCase activity, ABX and α-synuclein levels are also analysed in blood and CSF. A repeated-measures analysis of variance will be used for elaborating results. The primary analysis will be by intention to treat. Ethics and dissemination The study and protocols have been approved by the ethics committee of centres. The study is conducted according to good clinical practice and the Declaration of Helsinki. The trial findings will be published in peer-reviewed journals and presented at conferences. Trial registration numbers NCT05287503, EudraCT 2021-004565-13.
Collapse
Affiliation(s)
- Fabiana Colucci
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Micol Avenali
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- IRCCS Mondino Foundation, Pavia, Italy
| | - Rosita De Micco
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Marco Fusar Poli
- Neuropsychology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | | | - Mario Stanziano
- Neuroradiology Unit, Foundation IRCCS Carlo Besta Neurological Institute, Milano, Italy
| | | | - Giada Cuconato
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Valentina Franco
- IRCCS Mondino Foundation, Pavia, Italy
- Division of Clinical and Experimental Pharmacology, Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | | | | | | | - Antonio Emanuele Elia
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Luigi Romito
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Grazia Devigili
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Valentina Leta
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
- Parkinson's Centre of Excellence, King's College London, London, UK
| | - Barbara Garavaglia
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Nico Golfrè Andreasi
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Federico Cazzaniga
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Chiara Reale
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | | | | | | | | | | | | | - Anna Pichiecchio
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- IRCCS Mondino Foundation, Pavia, Italy
| | - Mattia Verri
- Neuroradiology Unit, Foundation IRCCS Carlo Besta Neurological Institute, Milano, Italy
| | - Fabrizio Esposito
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mario Cirillo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Federica Di Nardo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Simone Aloisio
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mattia Siciliano
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- Department of Psychology, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Sara Prioni
- Neuropsychology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Paolo Amami
- Neuropsychology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Sylvie Piacentini
- Neuropsychology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Maria Grazia Bruzzone
- Neuroradiology Unit, Foundation IRCCS Carlo Besta Neurological Institute, Milano, Italy
| | - Marina Grisoli
- Neuroradiology Unit, Foundation IRCCS Carlo Besta Neurological Institute, Milano, Italy
| | - Fabio Moda
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Roberto Eleopra
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Alessandro Tessitore
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Enza Maria Valente
- IRCCS Mondino Foundation, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Roberto Cilia
- Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| |
Collapse
|
27
|
Kim MS, Lee J, Kang D, Kim H, Kang SY. Association Between Ambroxol at the Usual Dose and the Risk of Parkinson's Disease in Chronic Lung Disease. J Clin Neurol 2023; 19:621-623. [PMID: 37901900 PMCID: PMC10622721 DOI: 10.3988/jcn.2023.0172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/01/2023] [Indexed: 10/31/2023] Open
Affiliation(s)
- Min Seung Kim
- Department of Neurology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Korea
| | - Jungkuk Lee
- Data Science Team, Hanmi Pharm. Co., Ltd, Seoul, Korea
| | - Dongwoo Kang
- Data Science Team, Hanmi Pharm. Co., Ltd, Seoul, Korea
| | - Hasung Kim
- Data Science Team, Hanmi Pharm. Co., Ltd, Seoul, Korea
| | - Suk Yun Kang
- Department of Neurology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Korea.
| |
Collapse
|
28
|
Skylar-Scott IA, Sha SJ. Lewy Body Dementia: An Overview of Promising Therapeutics. Curr Neurol Neurosci Rep 2023; 23:581-592. [PMID: 37572228 DOI: 10.1007/s11910-023-01292-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2023] [Indexed: 08/14/2023]
Abstract
PURPOSE OF REVIEW Lewy body dementia (LBD) encompasses dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD). This article will emphasize potential disease-modifying therapies as well as investigative symptomatic treatments for non-motor symptoms including cognitive impairment and psychosis that can present a tremendous burden to patients with LBD and their caregivers. RECENT FINDINGS We review 11 prospective disease-modifying therapies (DMT) including four with phase 2 data (neflamapimod, nilotinib, bosutinib, and E2027); four with some limited data in symptomatic populations including phase 1, open-label, registry, or cohort data (vodabatinib, ambroxol, clenbuterol, and terazosin); and three with phase 1 data in healthy populations (Anle138b, fosgonimeton, and CT1812). We also appraise four symptomatic therapies for cognitive impairment, but due to safety and efficacy concerns, only NYX-458 remains under active investigation. Of symptomatic therapies for psychosis recently investigated, pimavanserin shows promise in LBD, but studies of nelotanserin have been suspended. Although the discovery of novel symptomatic and disease-modifying therapeutics remains a significant challenge, recently published and upcoming trials signify promising strides toward that aim.
Collapse
Affiliation(s)
- Irina A Skylar-Scott
- Memory Disorders Division, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 213 Quarry Road, Palo Alto, CA, 94305, USA.
| | - Sharon J Sha
- Memory Disorders Division, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 213 Quarry Road, Palo Alto, CA, 94305, USA
| |
Collapse
|
29
|
Huh YE, Usnich T, Scherzer CR, Klein C, Chung SJ. GBA1 Variants and Parkinson's Disease: Paving the Way for Targeted Therapy. J Mov Disord 2023; 16:261-278. [PMID: 37302978 PMCID: PMC10548077 DOI: 10.14802/jmd.23023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/28/2023] [Accepted: 06/09/2023] [Indexed: 06/13/2023] Open
Abstract
Glucosylceramidase beta 1 (GBA1) variants have attracted enormous attention as the most promising and important genetic candidates for precision medicine in Parkinson's disease (PD). A substantial correlation between GBA1 genotypes and PD phenotypes could inform the prediction of disease progression and promote the development of a preventive intervention for individuals at a higher risk of a worse disease prognosis. Moreover, the GBA1-regulated pathway provides new perspectives on the pathogenesis of PD, such as dysregulated sphingolipid metabolism, impaired protein quality control, and disrupted endoplasmic reticulum-Golgi trafficking. These perspectives have led to the development of novel disease-modifying therapies for PD targeting the GBA1-regulated pathway by repositioning treatment strategies for Gaucher's disease. This review summarizes the current hypotheses on a mechanistic link between GBA1 variants and PD and possible therapeutic options for modulating GBA1-regulated pathways in PD patients.
Collapse
Affiliation(s)
- Young Eun Huh
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Tatiana Usnich
- Institute of Neurogenetics, University of Lübeck and University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - Clemens R. Scherzer
- Advanced Center for Parkinson’s Disease Research, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, USA
- Precision Neurology Program, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, USA
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck and University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - Sun Ju Chung
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
30
|
Chwiszczuk LJ, Breitve MH, Kirsebom BEB, Selnes P, Fløvig JC, Knapskog AB, Skogseth RE, Hubbers J, Holst-Larsen E, Rongve A. The ANeED study - ambroxol in new and early dementia with Lewy bodies (DLB): protocol for a phase IIa multicentre, randomised, double-blinded and placebo-controlled trial. Front Aging Neurosci 2023; 15:1163184. [PMID: 37304077 PMCID: PMC10250712 DOI: 10.3389/fnagi.2023.1163184] [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: 02/10/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Background Currently, there are no disease-modifying pharmacological treatment options for dementia with Lewy bodies (DLB). The hallmark of DLB is pathological alpha-synuclein (aS) deposition. There are growing amounts of data suggesting that reduced aS clearance is caused by failure in endolysosomal and authophagic pathways, as well as and glucocerebrosidase (GCase) dysfunction and mutations in the GCase gene (GBA). The population's studies demonstrated that the incidence of GBA mutations is higher among Parkinson's disease (PD) patients, and carriers of such mutations have a higher risk of developing PD. The incidence of GBA mutations is even higher in DLB and a genome-wide association study (GWAS) confirmed the correlation between GBA mutations and DLB. In vivo experiments have shown that ambroxol (ABX) may increase GCase activity and GCase levels and therefore enhance aS autophagy-lysosome degradation pathways. Moreover, there is an emerging hypothesis that ABX may have an effect as a DLB modifying drug. The aims of the study "Ambroxol in new and early Dementia with Lewy Bodies (ANeED) are to investigate the tolerability, safety and effects of ABX in patients with DLB. Methods This is a multicentre, phase IIa, double-blinded, randomised and placebo-controlled clinical trial, using a parallel arm design for 18 months' follow-up. The allocation ratio is 1:1 (treatment:placebo). Discussion The ANeED study is an ongoing clinical drug trial with ABX. The unique, but not fully understood mechanism of ABX on the enhancement of lysosomal aS clearance may be promising as a possible modifying treatment in DLB. Trial Registration The clinical trial is registered in the international trials register - clinicaltrials.com (NCT0458825) and nationally at the Current Research Information System in Norway (CRISTIN 2235504).
Collapse
Affiliation(s)
- Luiza Jadwiga Chwiszczuk
- Department of Old Age Related Medicine, Haugesund Hospital, Helse Fonna Trust, Haugesund, Norway
- Department of Research and Innovation, Haugesund Hospital, Helse Fonna Trust, Haugesund, Norway
| | - Monica Haraldseid Breitve
- Department of Research and Innovation, Haugesund Hospital, Helse Fonna Trust, Haugesund, Norway
- Department of Clinical Neuropsychology, Haugesund Hospital, Helse Fonna Trust, Haugesund, Norway
| | | | - Per Selnes
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| | | | | | - Ragnhild E. Skogseth
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
| | - Jessica Hubbers
- Department of Research and Innovation, Haugesund Hospital, Helse Fonna Trust, Haugesund, Norway
| | | | - Arvid Rongve
- Department of Research and Innovation, Haugesund Hospital, Helse Fonna Trust, Haugesund, Norway
- Sant Olavs Hospital, Trondheim, Norway
- Institute of Clinical Medicine, University in Bergen, Bergen, Norway
| |
Collapse
|
31
|
Menozzi E, Toffoli M, Schapira AHV. Targeting the GBA1 pathway to slow Parkinson disease: Insights into clinical aspects, pathogenic mechanisms and new therapeutic avenues. Pharmacol Ther 2023; 246:108419. [PMID: 37080432 DOI: 10.1016/j.pharmthera.2023.108419] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/31/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
The GBA1 gene encodes the lysosomal enzyme glucocerebrosidase (GCase), which is involved in sphingolipid metabolism. Biallelic variants in GBA1 cause Gaucher disease (GD), a lysosomal storage disorder characterised by loss of GCase activity and aberrant intracellular accumulation of GCase substrates. Carriers of GBA1 variants have an increased risk of developing Parkinson disease (PD), with odds ratio ranging from 2.2 to 30 according to variant severity. GBA1 variants which do not cause GD in homozygosis can also increase PD risk. Patients with PD carrying GBA1 variants show a more rapidly progressive phenotype compared to non-carriers, emphasising the need for disease modifying treatments targeting the GBA1 pathway. Several mechanisms secondary to GCase dysfunction are potentially responsible for the pathological changes leading to PD. Misfolded GCase proteins induce endoplasmic reticulum stress and subsequent unfolded protein response and impair the autophagy-lysosomal pathway. This results in α-synuclein accumulation and spread, and promotes neurodegenerative changes. Preclinical evidence also shows that products of GCase activity can promote accumulation of α-synuclein, however there is no convincing evidence of substrate accumulation in GBA1-PD brains. Altered lipid homeostasis secondary to loss of GCase activity could also contribute to PD pathology. Treatments that target the GBA1 pathway could reverse these pathological processes and halt/slow the progression of PD. These range from augmentation of GCase activity via GBA1 gene therapy, restoration of normal intracellular GCase trafficking via molecular chaperones, and substrate reduction therapy. This review discusses the pathways associated with GBA1-PD and related novel GBA1-targeted interventions for PD treatment.
Collapse
Affiliation(s)
- Elisa Menozzi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States of America
| | - Marco Toffoli
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States of America
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States of America.
| |
Collapse
|
32
|
Qiao J, Wang C, Chen Y, Yu S, Liu Y, Yu S, Jiang L, Jin C, Wang X, Zhang P, Zhao D, Wang J, Liu M. Herbal/Natural Compounds Resist Hallmarks of Brain Aging: From Molecular Mechanisms to Therapeutic Strategies. Antioxidants (Basel) 2023; 12:antiox12040920. [PMID: 37107295 PMCID: PMC10136184 DOI: 10.3390/antiox12040920] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Aging is a complex process of impaired physiological integrity and function, and is associated with increased risk of cardiovascular disease, diabetes, neurodegeneration, and cancer. The cellular environment of the aging brain exhibits perturbed bioenergetics, impaired adaptive neuroplasticity and flexibility, abnormal neuronal network activity, dysregulated neuronal Ca2+ homeostasis, accumulation of oxidatively modified molecules and organelles, and clear signs of inflammation. These changes make the aging brain susceptible to age-related diseases, such as Alzheimer's and Parkinson's diseases. In recent years, unprecedented advances have been made in the study of aging, especially the effects of herbal/natural compounds on evolutionarily conserved genetic pathways and biological processes. Here, we provide a comprehensive review of the aging process and age-related diseases, and we discuss the molecular mechanisms underlying the therapeutic properties of herbal/natural compounds against the hallmarks of brain aging.
Collapse
Affiliation(s)
- Juhui Qiao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Chenxi Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yu Chen
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Shuang Yu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Ying Liu
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Shiting Yu
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Leilei Jiang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Chenrong Jin
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Xinran Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Peiguang Zhang
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun 130033, China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jiawen Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
- Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Meichen Liu
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|
33
|
Patzwaldt K, Berezhnoy G, Ionescu T, Schramm L, Wang Y, Owczorz M, Calderón E, Poli S, Serna Higuita LM, Gonzalez-Menendez I, Quintanilla-Martinez L, Herfert K, Pichler B, Trautwein C, Castaneda-Vega S. Repurposing the mucolytic agent ambroxol for treatment of sub-acute and chronic ischaemic stroke. Brain Commun 2023; 5:fcad099. [PMID: 37065090 PMCID: PMC10090797 DOI: 10.1093/braincomms/fcad099] [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: 06/20/2022] [Revised: 01/31/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Ambroxol is a well-known mucolytic expectorant, which has gained much attention in amyotrophic lateral sclerosis, Parkinson's and Gaucher's disease. A specific focus has been placed on ambroxol's glucocerebrosidase-stimulating activity, on grounds that the point mutation of the gba1 gene, which codes for this enzyme, is a risk factor for developing Parkinson's disease. However, ambroxol has been attributed other characteristics, such as the potent inhibition of sodium channels, modification of calcium homeostasis, anti-inflammatory effects and modifications of oxygen radical scavengers. We hypothesized that ambroxol could have a direct impact on neuronal rescue if administered directly after ischaemic stroke induction. We longitudinally evaluated 53 rats using magnetic resonance imaging to examine stroke volume, oedema, white matter integrity, resting state functional MRI and behaviour for 1 month after ischemic stroke onset. For closer mechanistic insights, we evaluated tissue metabolomics of different brain regions in a subgroup of animals using ex vivo nuclear magnetic resonance spectroscopy. Ambroxol-treated animals presented reduced stroke volumes, reduced cytotoxic oedema, reduced white matter degeneration, reduced necrosis, improved behavioural outcomes and complex changes in functional brain connectivity. Nuclear magnetic resonance spectroscopy tissue metabolomic data at 24 h post-stroke proposes several metabolites that are capable of minimizing post-ischaemic damage and that presented prominent shifts during ambroxol treatment in comparison to controls. Taking everything together, we propose that ambroxol catalyzes recovery in energy metabolism, cellular homeostasis, membrane repair mechanisms and redox balance. One week of ambroxol administration following stroke onset reduced ischaemic stroke severity and improved functional outcome in the subacute phase followed by reduced necrosis in the chronic stroke phase.
Collapse
Affiliation(s)
- Kristin Patzwaldt
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen 72076, Germany
| | - Georgy Berezhnoy
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen 72076, Germany
| | - Tudor Ionescu
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen 72076, Germany
| | - Linda Schramm
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen 72076, Germany
| | - Yi Wang
- Hertie Institute for Clinical Brain Research, Department for Neurology, University Hospital Tuebingen, Tuebingen 72076, Germany
| | - Miriam Owczorz
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen 72076, Germany
| | - Eduardo Calderón
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Tuebingen 72076, Germany
| | - Sven Poli
- Hertie Institute for Clinical Brain Research, Department for Neurology, University Hospital Tuebingen, Tuebingen 72076, Germany
| | - Lina M Serna Higuita
- Institute for Clinical Epidemiology and Applied Biostatistics, University Hospital Tuebingen, Tuebingen 72076, Germany
| | - Irene Gonzalez-Menendez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, Eberhard Karls University, Tuebingen 72076, Germany
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, Tuebingen 72076, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, Eberhard Karls University, Tuebingen 72076, Germany
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, Tuebingen 72076, Germany
| | - Kristina Herfert
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen 72076, Germany
| | - Bernd Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen 72076, Germany
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, Tuebingen 72076, Germany
| | - Christoph Trautwein
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen 72076, Germany
| | - Salvador Castaneda-Vega
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen 72076, Germany
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Tuebingen 72076, Germany
| |
Collapse
|
34
|
Grigor’eva EV, Kopytova AE, Yarkova ES, Pavlova SV, Sorogina DA, Malakhova AA, Malankhanova TB, Baydakova GV, Zakharova EY, Medvedev SP, Pchelina SN, Zakian SM. Biochemical Characteristics of iPSC-Derived Dopaminergic Neurons from N370S GBA Variant Carriers with and without Parkinson's Disease. Int J Mol Sci 2023; 24:ijms24054437. [PMID: 36901867 PMCID: PMC10002967 DOI: 10.3390/ijms24054437] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/15/2023] [Accepted: 02/18/2023] [Indexed: 03/12/2023] Open
Abstract
GBA variants increase the risk of Parkinson's disease (PD) by 10 times. The GBA gene encodes the lysosomal enzyme glucocerebrosidase (GCase). The p.N370S substitution causes a violation of the enzyme conformation, which affects its stability in the cell. We studied the biochemical characteristics of dopaminergic (DA) neurons generated from induced pluripotent stem cells (iPSCs) from a PD patient with the GBA p.N370S mutation (GBA-PD), an asymptomatic GBA p.N370S carrier (GBA-carrier), and two healthy donors (control). Using liquid chromatography with tandem mass spectrometry (LC-MS/MS), we measured the activity of six lysosomal enzymes (GCase, galactocerebrosidase (GALC), alpha-glucosidase (GAA), alpha-galactosidase (GLA), sphingomyelinase (ASM), and alpha-iduronidase (IDUA)) in iPSC-derived DA neurons from the GBA-PD and GBA-carrier. DA neurons from the GBA mutation carrier demonstrated decreased GCase activity compared to the control. The decrease was not associated with any changes in GBA expression levels in DA neurons. GCase activity was more markedly decreased in the DA neurons of GBA-PD patient compared to the GBA-carrier. The amount of GCase protein was decreased only in GBA-PD neurons. Additionally, alterations in the activity of the other lysosomal enzymes (GLA and IDUA) were found in GBA-PD neurons compared to GBA-carrier and control neurons. Further study of the molecular differences between the GBA-PD and the GBA-carrier is essential to investigate whether genetic factors or external conditions are the causes of the penetrance of the p.N370S GBA variant.
Collapse
Affiliation(s)
- Elena V. Grigor’eva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Alena E. Kopytova
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Center «Kurchatov Institute», Gatchina 188300, Russia
- Department of Molecular Genetic and Nanobiological Technologies, Scientific and Research Centre, Pavlov First Saint-Petersburg State Medical University, Saint-Petersburg 197022, Russia
| | - Elena S. Yarkova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Sophia V. Pavlova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Diana A. Sorogina
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Anastasia A. Malakhova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Tuyana B. Malankhanova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | | | | | - Sergey P. Medvedev
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Sofia N. Pchelina
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Center «Kurchatov Institute», Gatchina 188300, Russia
- Department of Molecular Genetic and Nanobiological Technologies, Scientific and Research Centre, Pavlov First Saint-Petersburg State Medical University, Saint-Petersburg 197022, Russia
| | - Suren M. Zakian
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russia
- Correspondence:
| |
Collapse
|
35
|
Haider A, Elghazawy NH, Dawoud A, Gebhard C, Wichmann T, Sippl W, Hoener M, Arenas E, Liang SH. Translational molecular imaging and drug development in Parkinson's disease. Mol Neurodegener 2023; 18:11. [PMID: 36759912 PMCID: PMC9912681 DOI: 10.1186/s13024-023-00600-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that primarily affects elderly people and constitutes a major source of disability worldwide. Notably, the neuropathological hallmarks of PD include nigrostriatal loss and the formation of intracellular inclusion bodies containing misfolded α-synuclein protein aggregates. Cardinal motor symptoms, which include tremor, rigidity and bradykinesia, can effectively be managed with dopaminergic therapy for years following symptom onset. Nonetheless, patients ultimately develop symptoms that no longer fully respond to dopaminergic treatment. Attempts to discover disease-modifying agents have increasingly been supported by translational molecular imaging concepts, targeting the most prominent pathological hallmark of PD, α-synuclein accumulation, as well as other molecular pathways that contribute to the pathophysiology of PD. Indeed, molecular imaging modalities such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT) can be leveraged to study parkinsonism not only in animal models but also in living patients. For instance, mitochondrial dysfunction can be assessed with probes that target the mitochondrial complex I (MC-I), while nigrostriatal degeneration is typically evaluated with probes designed to non-invasively quantify dopaminergic nerve loss. In addition to dopaminergic imaging, serotonin transporter and N-methyl-D-aspartate (NMDA) receptor probes are increasingly used as research tools to better understand the complexity of neurotransmitter dysregulation in PD. Non-invasive quantification of neuroinflammatory processes is mainly conducted by targeting the translocator protein 18 kDa (TSPO) on activated microglia using established imaging agents. Despite the overwhelming involvement of the brain and brainstem, the pathophysiology of PD is not restricted to the central nervous system (CNS). In fact, PD also affects various peripheral organs such as the heart and gastrointestinal tract - primarily via autonomic dysfunction. As such, research into peripheral biomarkers has taken advantage of cardiac autonomic denervation in PD, allowing the differential diagnosis between PD and multiple system atrophy with probes that visualize sympathetic nerve terminals in the myocardium. Further, α-synuclein has recently gained attention as a potential peripheral biomarker in PD. This review discusses breakthrough discoveries that have led to the contemporary molecular concepts of PD pathophysiology and how they can be harnessed to develop effective imaging probes and therapeutic agents. Further, we will shed light on potential future trends, thereby focusing on potential novel diagnostic tracers and disease-modifying therapeutic interventions.
Collapse
Affiliation(s)
- Achi Haider
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Department of Radiology and Imaging Sciences, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| | - Nehal H Elghazawy
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
| | - Alyaa Dawoud
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Thomas Wichmann
- Department of Neurology/School of Medicine, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Wolfgang Sippl
- Institute of Pharmacy, Department of Medicinal Chemistry, Martin-Luther-University Halle-Wittenberg, W.-Langenbeck-Str. 4, 06120, Halle, Germany
| | - Marius Hoener
- Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Ernest Arenas
- Karolinska Institutet, MBB, Molecular Neurobiology, Stockholm, Sweden
| | - Steven H Liang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Department of Radiology and Imaging Sciences, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| |
Collapse
|
36
|
Smith AR, Richards DM, Lunnon K, Schapira AHV, Migdalska-Richards A. DNA Methylation of α-Synuclein Intron 1 Is Significantly Decreased in the Frontal Cortex of Parkinson's Individuals with GBA1 Mutations. Int J Mol Sci 2023; 24:ijms24032687. [PMID: 36769009 PMCID: PMC9917152 DOI: 10.3390/ijms24032687] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
Parkinson's disease (PD) is a common movement disorder, estimated to affect 4% of individuals by the age of 80. Mutations in the glucocerebrosidase 1 (GBA1) gene represent the most common genetic risk factor for PD, with at least 7-10% of non-Ashkenazi PD individuals carrying a GBA1 mutation (PD-GBA1). Although similar to idiopathic PD, the clinical presentation of PD-GBA1 includes a slightly younger age of onset, a higher incidence of neuropsychiatric symptoms, and a tendency to earlier, more prevalent and more significant cognitive impairment. The pathophysiological mechanisms underlying PD-GBA1 are incompletely understood, but, as in idiopathic PD, α-synuclein accumulation is thought to play a key role. It has been hypothesized that this overexpression of α-synuclein is caused by epigenetic modifications. In this paper, we analyze DNA methylation levels at 17 CpG sites located within intron 1 and the promoter of the α-synuclein (SNCA) gene in three different brain regions (frontal cortex, putamen and substantia nigra) in idiopathic PD, PD-GBA1 and elderly non-PD controls. In all three brain regions we find a tendency towards a decrease in DNA methylation within an eight CpG region of intron 1 in both idiopathic PD and PD-GBA1. The trend towards a reduction in DNA methylation was more pronounced in PD-GBA1, with a significant decrease in the frontal cortex. This suggests that PD-GBA1 and idiopathic PD have distinct epigenetic profiles, and highlights the importance of separating idiopathic PD and PD-GBA1 cases. This work also provides initial evidence that different genetic subtypes might exist within PD, each characterized by its own pathological mechanism. This may have important implications for how PD is diagnosed and treated.
Collapse
Affiliation(s)
- Adam R. Smith
- Faculty of Health and Life Sciences, University of Exeter, Exeter EX2 5DW, UK
| | - David M. Richards
- Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
- Department of Physics and Astronomy, University of Exeter, Exeter EX4 4QD, UK
| | - Katie Lunnon
- Faculty of Health and Life Sciences, University of Exeter, Exeter EX2 5DW, UK
| | - Anthony H. V. Schapira
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London NW3 2PF, UK
| | - Anna Migdalska-Richards
- Faculty of Health and Life Sciences, University of Exeter, Exeter EX2 5DW, UK
- Correspondence:
| |
Collapse
|
37
|
Pérez-Arancibia R, Cisternas-Olmedo M, Sepúlveda D, Troncoso-Escudero P, Vidal RL. Small molecules to perform big roles: The search for Parkinson's and Huntington's disease therapeutics. Front Neurosci 2023; 16:1084493. [PMID: 36699535 PMCID: PMC9868863 DOI: 10.3389/fnins.2022.1084493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
Neurological motor disorders (NMDs) such as Parkinson's disease and Huntington's disease are characterized by the accumulation and aggregation of misfolded proteins that trigger cell death of specific neuronal populations in the central nervous system. Differential neuronal loss initiates the impaired motor control and cognitive function in the affected patients. Although major advances have been carried out to understand the molecular basis of these diseases, to date there are no treatments that can prevent, cure, or significantly delay the progression of the disease. In this context, strategies such as gene editing, cellular therapy, among others, have gained attention as they effectively reduce the load of toxic protein aggregates in different models of neurodegeneration. Nevertheless, these strategies are expensive and difficult to deliver into the patients' nervous system. Thus, small molecules and natural products that reduce protein aggregation levels are highly sought after. Numerous drug discovery efforts have analyzed large libraries of synthetic compounds for the treatment of different NMDs, with a few candidates reaching clinical trials. Moreover, the recognition of new druggable targets for NMDs has allowed the discovery of new small molecules that have demonstrated their efficacy in pre-clinical studies. It is also important to recognize the contribution of natural products to the discovery of new candidates that can prevent or cure NMDs. Additionally, the repurposing of drugs for the treatment of NMDs has gained huge attention as they have already been through clinical trials confirming their safety in humans, which can accelerate the development of new treatment. In this review, we will focus on the new advances in the discovery of small molecules for the treatment of Parkinson's and Huntington's disease. We will begin by discussing the available pharmacological treatments to modulate the progression of neurodegeneration and to alleviate the motor symptoms in these diseases. Then, we will analyze those small molecules that have reached or are currently under clinical trials, including natural products and repurposed drugs.
Collapse
Affiliation(s)
- Rodrigo Pérez-Arancibia
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Departamento de Ciencias Básicas, Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| | - Marisol Cisternas-Olmedo
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Denisse Sepúlveda
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Paulina Troncoso-Escudero
- Molecular Diagnostic and Biomarkers Laboratory, Department of Pathology, Faculty of Medicine Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Rene L. Vidal
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| |
Collapse
|
38
|
Zhao X, Lin Y, Liou B, Fu W, Jian J, Fannin V, Zhang W, Setchell KDR, Grabowski GA, Sun Y, Liu CJ. PGRN deficiency exacerbates, whereas a brain penetrant PGRN derivative protects, GBA1 mutation-associated pathologies and diseases. Proc Natl Acad Sci U S A 2023; 120:e2210442120. [PMID: 36574647 PMCID: PMC9910439 DOI: 10.1073/pnas.2210442120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/08/2022] [Indexed: 12/28/2022] Open
Abstract
Mutations in GBA1, encoding glucocerebrosidase (GCase), cause Gaucher disease (GD) and are also genetic risks in developing Parkinson's disease (PD). Currently, the approved therapies are only effective for directly treating visceral symptoms, but not for primary neuronopathic involvement in GD (nGD). Progranulin (PGRN), encoded by GRN, is a novel modifier of GCase, but the impact of PGRN in GBA1 mutation-associated pathologies in vivo remains unknown. Herein, Grn-/- mice crossed into Gba9v/9v mice, a Gba1 mutant line homozygous for the Gba1 D409V mutation, generating Grn-/-Gba9v/9v (PG9V) mice. PG9V mice exhibited neurobehavioral deficits, early onset, and more severe GD phenotypes compared to Grn-/- and Gba9v/9v mice. Moreover, PG9V mice also displayed PD-like phenotype. Mechanistic analysis revealed that PGRN deficiency caused severe neuroinflammation with microgliosis and astrogliosis, along with impaired autophagy associated with the Gba1 mutation. A PGRN-derived peptide, termed ND7, ameliorated the disease phenotype in GD patient fibroblasts ex vivo. Unexpectedly, ND7 penetrated the blood-brain barrier (BBB) and effectively ameliorated the nGD manifestations and PD pathology in Gba9v/null and PG9V mice. Collectively, this study not only provides the first line of in vivo but also ex vivo evidence demonstrating the crucial role of PGRN in GBA1/Gba1 mutation-related pathologies, as well as a clinically relevant mouse model for mechanistic and potential therapeutics studies for nGD and PD. Importantly, a BBB penetrant PGRN-derived biologic was developed that may provide treatment for rare lysosomal storage diseases and common neurodegenerative disorders, particularly nGD and PD.
Collapse
Affiliation(s)
- Xiangli Zhao
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY10003
| | - Yi Lin
- The Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH45229
| | - Benjamin Liou
- The Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH45229
| | - Wenyu Fu
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY10003
| | - Jinlong Jian
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY10003
| | - Venette Fannin
- The Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH45229
| | - Wujuan Zhang
- The Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH45229
| | - Kenneth D. R. Setchell
- The Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH45229
| | - Gregory A. Grabowski
- The Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH45229
| | - Ying Sun
- The Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH45229
| | - Chuan-ju Liu
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY10003
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY10016
| |
Collapse
|
39
|
Volta M. Lysosomal Pathogenesis of Parkinson's Disease: Insights From LRRK2 and GBA1 Rodent Models. Neurotherapeutics 2023; 20:127-139. [PMID: 36085537 PMCID: PMC10119359 DOI: 10.1007/s13311-022-01290-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2022] [Indexed: 01/18/2023] Open
Abstract
The discovery of mutations in LRRK2 and GBA1 that are linked to Parkinson's disease provided further evidence that autophagy and lysosome pathways are likely implicated in the pathogenic process. Their protein products are important regulators of lysosome function. LRRK2 has kinase-dependent effects on lysosome activity, autophagic efficacy and lysosomal Ca2+ signaling. Glucocerebrosidase (encoded by GBA1) is a hydrolytic enzyme contained in the lysosomes and contributes to the degradation of alpha-synuclein. PD-related mutations in LRRK2 and GBA1 slow the degradation of alpha-synuclein, thus directly implicating the dysfunction of the process in the neuropathology of Parkinson's disease. The development of genetic rodent models of LRRK2 and GBA1 provided hopes of obtaining reliable preclinical models in which to study pathogenic processes and perform drug validation studies. Here, I will review the extensive characterization of these models, their impact on understanding lysosome alterations in the course of Parkinson's disease and what novel insights have been obtained. In addition, I will discuss how these models fare with respect to the features of a "gold standard" animal models and what could be attempted in future studies to exploit LRRK2 and GBA1 rodent models in the fight against Parkinson's disease.
Collapse
Affiliation(s)
- Mattia Volta
- Institute for Biomedicine, Eurac Research - Affiliated Institute of the University of Lübeck, via Volta 21, Bolzano, 39100, Italy.
| |
Collapse
|
40
|
Mächtel R, Boros FA, Dobert JP, Arnold P, Zunke F. From Lysosomal Storage Disorders to Parkinson's Disease - Challenges and Opportunities. J Mol Biol 2022:167932. [PMID: 36572237 DOI: 10.1016/j.jmb.2022.167932] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Lysosomes are specialized organelles with an acidic pH that act as recycling hubs for intracellular and extracellular components. They harbour numerous different hydrolytic enzymes to degrade substrates like proteins, peptides, and glycolipids. Reduced catalytic activity of lysosomal enzymes can cause the accumulation of these substrates and loss of lysosomal integrity, resulting in lysosomal dysfunction and lysosomal storage disorders (LSDs). Post-mitotic cells, such as neurons, seem to be highly sensitive to damages induced by lysosomal dysfunction, thus LSDs often manifest with neurological symptoms. Interestingly, some LSDs and Parkinson's disease (PD) share common cellular pathomechanisms, suggesting convergence of aetiology of the two disease types. This is further underlined by genetic associations of several lysosomal genes involved in LSDs with PD. The increasing number of lysosome-associated genetic risk factors for PD makes it necessary to understand functions and interactions of lysosomal proteins/enzymes both in health and disease, thereby holding the potential to identify new therapeutic targets. In this review, we highlight genetic and mechanistic interactions between the complex lysosomal network, LSDs and PD, and elaborate on methodical challenges in lysosomal research.
Collapse
Affiliation(s)
- Rebecca Mächtel
- Department of Molecular Neurology, University Clinics Erlangen, Erlangen, Germany
| | | | - Jan Philipp Dobert
- Department of Molecular Neurology, University Clinics Erlangen, Erlangen, Germany
| | - Philipp Arnold
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Friederike Zunke
- Department of Molecular Neurology, University Clinics Erlangen, Erlangen, Germany.
| |
Collapse
|
41
|
O'Brien JT, Chouliaras L, Sultana J, Taylor JP, Ballard C. RENEWAL: REpurposing study to find NEW compounds with Activity for Lewy body dementia-an international Delphi consensus. Alzheimers Res Ther 2022; 14:169. [PMID: 36369100 PMCID: PMC9650797 DOI: 10.1186/s13195-022-01103-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022]
Abstract
Drug repositioning and repurposing has proved useful in identifying new treatments for many diseases, which can then rapidly be brought into clinical practice. Currently, there are few effective pharmacological treatments for Lewy body dementia (which includes both dementia with Lewy bodies and Parkinson's disease dementia) apart from cholinesterase inhibitors. We reviewed several promising compounds that might potentially be disease-modifying agents for Lewy body dementia and then undertook an International Delphi consensus study to prioritise compounds. We identified ambroxol as the top ranked agent for repurposing and identified a further six agents from the classes of tyrosine kinase inhibitors, GLP-1 receptor agonists, and angiotensin receptor blockers that were rated by the majority of our expert panel as justifying a clinical trial. It would now be timely to take forward all these compounds to Phase II or III clinical trials in Lewy body dementia.
Collapse
Affiliation(s)
- John T O'Brien
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK.
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK.
| | - Leonidas Chouliaras
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Janet Sultana
- College of Medicine and Health, University of Exeter, Exeter, UK
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle, UK
| | - Clive Ballard
- College of Medicine and Health, University of Exeter, Exeter, UK
| |
Collapse
|
42
|
Sahyadri M, Nadiga APR, Mehdi S, Mruthunjaya K, Nayak PG, Parihar VK, Manjula SN. Mitochondria-lysosome crosstalk in GBA1-associated Parkinson's disease. 3 Biotech 2022; 12:230. [PMID: 35992895 PMCID: PMC9388709 DOI: 10.1007/s13205-022-03261-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 07/17/2022] [Indexed: 11/26/2022] Open
Abstract
Organelle crosstalk is significant in regulating their respective functions and subsequent cell fate. Mitochondria and lysosomes are amongst the essential organelles in maintaining cellular homeostasis. Mitochondria-lysosome connections, which may develop dynamically in the human neurons, have been identified as sites of bidirectional communication. Aberrancies are often associated with neurodegenerative disorders like Parkinson's disease (PD), suggesting the physical and functional link between these two organelles. PD is often linked with genetic mutations of several mutations discovered in the familial forms of the disease; some are considered risk factors. Many of these genes are either associated with mitochondrial function or belong to endo-lysosomal pathways. The recent investigations have indicated that neurons with mutant glucosylceramidase beta (GBA1) exhibit extended mitochondria-lysosome connections in individuals with PD. This may be due to impaired control of the untethering protein, which aids in the hydrolysis of Rab7 GTP required for contact untethering. A GCase modulator may be used to augment the reduced GBA1 lysosomal enzyme activity in the neurons of PD patients. This review focuses on how GBA1 mutation in PD is interlinked with mitochondria-lysosome (ML) crosstalk, exploring the pathways governing these interactions and mechanistically comprehending the mitochondrial and lysosomal miscommunication in the pathophysiology of PD. This review is based on the limited literature available on the topic and hence may be subject to bias in its views. Our estimates may be conservative and limited due to the lack of studies under the said discipline due to its inherent complex nature. The current association of GBA1 to PD pathogenesis is based on the limited scope of study and further research is necessary to explore the risk factors further and identify the relationship with more detail.
Collapse
Affiliation(s)
- M. Sahyadri
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015 Karnataka India
| | - Abhishek P. R. Nadiga
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015 Karnataka India
| | - Seema Mehdi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015 Karnataka India
| | - K. Mruthunjaya
- Department of Pharmacognosy, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015 Karnataka India
| | - Pawan G. Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, 576104 Karnataka India
| | - Vipan K. Parihar
- Department of Pharmacology and Toxicology, NIPER-Hajipur, Bihar, 844102 India
| | - S. N. Manjula
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015 Karnataka India
| |
Collapse
|
43
|
Vieira SRL, Schapira AHV. Glucocerebrosidase mutations and Parkinson disease. J Neural Transm (Vienna) 2022; 129:1105-1117. [PMID: 35932311 PMCID: PMC9463283 DOI: 10.1007/s00702-022-02531-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/07/2022] [Indexed: 11/06/2022]
Abstract
The discovery of glucocerebrosidase (GBA1) mutations as the greatest numerical genetic risk factor for the development of Parkinson disease (PD) resulted in a paradigm shift within the research landscape. Efforts to elucidate the mechanisms behind GBA1-associated PD have highlighted shared pathways in idiopathic PD including the loss and gain-of-function hypotheses, endoplasmic reticulum stress, lipid metabolism, neuroinflammation, mitochondrial dysfunction and altered autophagy-lysosomal pathway responsible for degradation of aggregated and misfolded a-synuclein. GBA1-associated PD exhibits subtle differences in phenotype and disease progression compared to idiopathic counterparts notably an earlier age of onset, faster motor decline and greater frequency of non-motor symptoms (which also constitute a significant aspect of the prodromal phase of the disease). GBA1-targeted therapies have been developed and are being investigated in clinical trials. The most notable are Ambroxol, a small molecule chaperone, and Venglustat, a blood-brain-barrier-penetrant substrate reduction therapy agent. It is imperative that further studies clarify the aetiology of GBA1-associated PD, enabling the development of a greater abundance of targeted therapies in this new era of precision medicine.
Collapse
Affiliation(s)
- Sophia R L Vieira
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, Rowland Hill St., London, NW3 2PF, UK
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, Rowland Hill St., London, NW3 2PF, UK.
| |
Collapse
|
44
|
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
|
45
|
Lang M, Pramstaller PP, Pichler I. Crosstalk of organelles in Parkinson's disease - MiT family transcription factors as central players in signaling pathways connecting mitochondria and lysosomes. Mol Neurodegener 2022; 17:50. [PMID: 35842725 PMCID: PMC9288732 DOI: 10.1186/s13024-022-00555-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 07/01/2022] [Indexed: 11/10/2022] Open
Abstract
Living organisms constantly need to adapt to their surrounding environment and have evolved sophisticated mechanisms to deal with stress. Mitochondria and lysosomes are central organelles in the response to energy and nutrient availability within a cell and act through interconnected mechanisms. However, when such processes become overwhelmed, it can lead to pathologies. Parkinson's disease (PD) is a common neurodegenerative disorder (NDD) characterized by proteinaceous intracellular inclusions and progressive loss of dopaminergic neurons, which causes motor and non-motor symptoms. Genetic and environmental factors may contribute to the disease etiology. Mitochondrial dysfunction has long been recognized as a hallmark of PD pathogenesis, and several aspects of mitochondrial biology are impaired in PD patients and models. In addition, defects of the autophagy-lysosomal pathway have extensively been observed in cell and animal models as well as PD patients' brains, where constitutive autophagy is indispensable for adaptation to stress and energy deficiency. Genetic and molecular studies have shown that the functions of mitochondria and lysosomal compartments are tightly linked and influence each other. Connections between these organelles are constituted among others by mitophagy, organellar dynamics and cellular signaling cascades, such as calcium (Ca2+) and mTOR (mammalian target of rapamycin) signaling and the activation of transcription factors. Members of the Microphthalmia-associated transcription factor family (MiT), including MITF, TFE3 and TFEB, play a central role in regulating cellular homeostasis in response to metabolic pressure and are considered master regulators of lysosomal biogenesis. As such, they are part of the interconnection between mitochondria and lysosome functions and therefore represent attractive targets for therapeutic approaches against NDD, including PD. The activation of MiT transcription factors through genetic and pharmacological approaches have shown encouraging results at ameliorating PD-related phenotypes in in vitro and in vivo models. In this review, we summarize the relationship between mitochondrial and autophagy-lysosomal functions in the context of PD etiology and focus on the role of the MiT pathway and its potential as pharmacological target against PD.
Collapse
Affiliation(s)
- Martin Lang
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy.
| | - Peter P Pramstaller
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy.,Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Irene Pichler
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| |
Collapse
|
46
|
GCase Enhancers: A Potential Therapeutic Option for Gaucher Disease and Other Neurological Disorders. Pharmaceuticals (Basel) 2022; 15:ph15070823. [PMID: 35890122 PMCID: PMC9325019 DOI: 10.3390/ph15070823] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 12/07/2022] Open
Abstract
Pharmaceutical chaperones (PCs) are small compounds able to bind and stabilize misfolded proteins, allowing them to recover their native folding and thus their biological activity. In particular, lysosomal storage disorders (LSDs), a class of metabolic disorders due to genetic mutations that result in misfolded lysosomal enzymes, can strongly benefit from the use of PCs able to facilitate their translocation to the lysosomes. This results in a recovery of their catalytic activity. No PC for the GCase enzyme (lysosomal acid-β-glucosidase, or glucocerebrosidase) has reached the market yet, despite the importance of this enzyme not only for Gaucher disease, the most common LSD, but also for neurological disorders, such as Parkinson’s disease. This review aims to describe the efforts made by the scientific community in the last 7 years (since 2015) in order to identify new PCs for the GCase enzyme, which have been mainly identified among glycomimetic-based compounds.
Collapse
|
47
|
Drobny A, Prieto Huarcaya S, Dobert J, Kluge A, Bunk J, Schlothauer T, Zunke F. The role of lysosomal cathepsins in neurodegeneration: Mechanistic insights, diagnostic potential and therapeutic approaches. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119243. [PMID: 35217144 DOI: 10.1016/j.bbamcr.2022.119243] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 12/12/2022]
Abstract
Lysosomes are ubiquitous organelles with a fundamental role in maintaining cellular homeostasis by mediating degradation and recycling processes. Cathepsins are the most abundant lysosomal hydrolyses and are responsible for the bulk degradation of various substrates. A correct autophagic function is essential for neuronal survival, as most neurons are post-mitotic and thus susceptible to accumulate cellular components. Increasing evidence suggests a crucial role of the lysosome in neurodegeneration as a key regulator of aggregation-prone and disease-associated proteins, such as α-synuclein, β-amyloid and huntingtin. Particularly, alterations in lysosomal cathepsins CTSD, CTSB and CTSL can contribute to the pathogenesis of neurodegenerative diseases as seen for neuronal ceroid lipofuscinosis, synucleinopathies (Parkinson's disease, Dementia with Lewy Body and Multiple System Atrophy) as well as Alzheimer's and Huntington's disease. In this review, we provide an overview of recent evidence implicating CTSD, CTSB and CTSL in neurodegeneration, with a special focus on the role of these enzymes in α-synuclein metabolism. In addition, we summarize the potential role of lysosomal cathepsins as clinical biomarkers in neurodegenerative diseases and discuss potential therapeutic approaches by targeting lysosomal function.
Collapse
Affiliation(s)
- Alice Drobny
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | | | - Jan Dobert
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Annika Kluge
- Department of Neurology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Josina Bunk
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | | | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
48
|
Azari A, Goodarzi A, Jafarkhani B, Eghbali M, Karimi Z, Hosseini Balef SS, Irannejad H. Novel molecular targets and mechanisms for neuroprotective modulation in neurodegenerative disorders. Cent Nerv Syst Agents Med Chem 2022; 22:88-107. [PMID: 35713146 DOI: 10.2174/1871524922666220616092132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/04/2022] [Accepted: 04/11/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Neuronal death underlies the symptoms of several human neurological disorders, including Alzheimer's, Parkinson's and Huntington's diseases, and amyotrophic lateral sclerosis that their precise pathophysiology have not yet been elucidated. According to various studies the prohibition is the best therapy with neuroprotective approaches which are advanced and safe methods. METHODS This review summarizes some of the already-known and newly emerged neuroprotective targets and strategies that their experimental effects have been reported. Accordingly, literature was studied from 2000 to 2021 and appropriate articles were searched in Google Scholar and Scopus with the keywords given in the Keywords section of the current review. RESULTS Lewy bodies are the histopathologic characteristics of neurodegenerative disorders and are protein-rich intracellular deposits in which Alpha-Synuclein is its major protein. Alpha-Synuclein's toxic potential provides a compelling rationale for therapeutic strategies aimed at decreasing its burden in neuronal cells through numerous pathways including ubiquitin-proteasome system and autophagy-lysosome Pathway, proteolytic breakdown via cathepsin D, kallikrein-6 (neurosin), calpain-1 or MMP9, heat shock proteins, and proteolysis targeting chimera which consists of a target protein ligand and an E3 ubiquitin ligase (E3) followed by target protein ubiquitination (PROTACs). Other targets that have been noticed recently are the mutant huntingtin, tau proteins and glycogen synthase kinase 3β that their accumulation proceeds extensive neuronal damage and up to the minute approach such as Proteolysis Targeting Chimera promotes its degradation in cells. As various studies demonstrated that Mendelian gene mutations can result into the neurodegenerative diseases, additional target that has gained much interest is epigenetics such as mutation, phosphodiesterase, RNA binding proteins and Nuclear respiratory factor 1. CONCLUSION The novel molecular targets and new strategies compiled and introduced here can be used by scientists to design and discover more efficient small molecule drugs against the neurodegenerative diseases. And also the genes in which their mutations can lead to the α-synuclein aggregation or accumulation are discussed and considered a valuable information of epigenetics in dementia.
Collapse
Affiliation(s)
- Aala Azari
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amin Goodarzi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Behrouz Jafarkhani
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Eghbali
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zohreh Karimi
- Department of Obstetrics & Gynecology, Imam Khomeini hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Sajad Hosseini Balef
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Hamid Irannejad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
49
|
Höglinger G, Schulte C, Jost WH, Storch A, Woitalla D, Krüger R, Falkenburger B, Brockmann K. GBA-associated PD: chances and obstacles for targeted treatment strategies. J Neural Transm (Vienna) 2022; 129:1219-1233. [PMID: 35639160 PMCID: PMC9463270 DOI: 10.1007/s00702-022-02511-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/01/2022] [Indexed: 11/08/2022]
Abstract
Given the clear role of GBA in the pathogenesis of Parkinson’s disease (PD) and its impact on phenotypical characteristics, this review provides an overview of the current knowledge of GBA-associated PD with a special focus on clinical trajectories and the underlying pathological mechanisms. Importantly, differences and characteristics based on mutation severity are recognized, and current as well as potential future treatment options are discussed. These findings will inform future strategies for patient stratification and cohort enrichment as well as suitable outcome measures when designing clinical trials.
Collapse
Affiliation(s)
- Günter Höglinger
- Department of Neurology, Hannover Medical School, 30625, Hannover, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Claudia Schulte
- Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.,German Center for Neurodegenerative Disease (DZNE), Tuebingen, Germany
| | | | - Alexander Storch
- Department of Neurology, Rostock University, Gehlsheimer Str. 20, 18147, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Gehlsheimer Str. 20, 18147, Rostock, Germany
| | - Dirk Woitalla
- Department of Neurology, St. Josef-Hospital, Katholische Kliniken Ruhrhalbinsel, Contilia Gruppe, Essen, Germany
| | - Rejko Krüger
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg.,Translational Neuroscience, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Parkinson Research Clinic, Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg
| | - Björn Falkenburger
- Department of Neurology, Faculty of Medicine, University Hospital Carl Gustav Carus and Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Kathrin Brockmann
- Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany. .,German Center for Neurodegenerative Disease (DZNE), Tuebingen, Germany.
| |
Collapse
|
50
|
Glucocerebrosidase-associated Parkinson disease: Pathogenic mechanisms and potential drug treatments. Neurobiol Dis 2022; 166:105663. [DOI: 10.1016/j.nbd.2022.105663] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/30/2022] [Accepted: 02/15/2022] [Indexed: 02/07/2023] Open
|