1
|
Mark JR, Tansey MG. Immune cell metabolic dysfunction in Parkinson's disease. Mol Neurodegener 2025; 20:36. [PMID: 40128809 PMCID: PMC11934562 DOI: 10.1186/s13024-025-00827-y] [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: 12/12/2024] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
Parkinson's disease (PD) is a multi-system disorder characterized histopathologically by degeneration of dopaminergic neurons in the substantia nigra pars compacta. While the etiology of PD remains multifactorial and complex, growing evidence suggests that cellular metabolic dysfunction is a critical driver of neuronal death. Defects in cellular metabolism related to energy production, oxidative stress, metabolic organelle health, and protein homeostasis have been reported in both neurons and immune cells in PD. We propose that these factors act synergistically in immune cells to drive aberrant inflammation in both the CNS and the periphery in PD, contributing to a hostile inflammatory environment which renders certain subsets of neurons vulnerable to degeneration. This review highlights the overlap between established neuronal metabolic deficits in PD with emerging findings in central and peripheral immune cells. By discussing the rapidly expanding literature on immunometabolic dysfunction in PD, we aim to draw attention to potential biomarkers and facilitate future development of immunomodulatory strategies to prevent or delay the progression of PD.
Collapse
Affiliation(s)
- Julian R Mark
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Malú Gámez Tansey
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
- Department of Neurology and Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, 32608, USA.
| |
Collapse
|
2
|
Zhao H, Lv Y, Xu J, Song X, Wang Q, Zhai X, Ma X, Qiu J, Cui L, Sun Y. The activation of microglia by the complement system in neurodegenerative diseases. Ageing Res Rev 2025; 104:102636. [PMID: 39647582 DOI: 10.1016/j.arr.2024.102636] [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: 11/22/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
Neurodegenerative diseases (NDDs) are a group of neurological disorders characterized by the progressive loss of neuronal structure and function, leading to cognitive and behavioral impairments. Despite significant research advancements, there is currently no definitive cure for NDDs. With global aging on the rise, the burden of these diseases is becoming increasingly severe, highlighting the urgency of understanding their pathogenesis and developing effective therapeutic strategies. Microglia, specialized macrophages in the central nervous system, play a dual role in maintaining neural homeostasis. They are involved in clearing cellular debris and apoptotic cells, but in their activated state, they release inflammatory factors that contribute significantly to neuroinflammation. The complement system (CS), a critical component of the innate immune system, assists in clearing damaged cells and proteins. However, excessive or uncontrolled activation of the CS can lead to chronic neuroinflammation, exacerbating neuronal damage. This review aims to explore the roles of microglia and the CS in the progression of NDDs, with a specific focus on the mechanisms through which the CS activates microglia by modulating mitochondrial function. Understanding these interactions may provide insights into potential therapeutic targets for mitigating neuroinflammation and slowing neurodegeneration.
Collapse
Affiliation(s)
- He Zhao
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China
| | - Yayun Lv
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China
| | - Jiasen Xu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China
| | - Xiaoyu Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China
| | - Qi Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China
| | - Xiaoyu Zhai
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China
| | - Xiaohui Ma
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China
| | - Jingjing Qiu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China.
| | - Limei Cui
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China.
| | - Yan Sun
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong 264000, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong 264000, China.
| |
Collapse
|
3
|
Demmings MD, da Silva Chagas L, Traetta ME, Rodrigues RS, Acutain MF, Barykin E, Datusalia AK, German-Castelan L, Mattera VS, Mazengenya P, Skoug C, Umemori H. (Re)building the nervous system: A review of neuron-glia interactions from development to disease. J Neurochem 2025; 169:e16258. [PMID: 39680483 DOI: 10.1111/jnc.16258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 12/18/2024]
Abstract
Neuron-glia interactions are fundamental to the development and function of the nervous system. During development, glia, including astrocytes, microglia, and oligodendrocytes, influence neuronal differentiation and migration, synapse formation and refinement, and myelination. In the mature brain, glia are crucial for maintaining neural homeostasis, modulating synaptic activity, and supporting metabolic functions. Neurons, inherently vulnerable to various stressors, rely on glia for protection and repair. However, glia, in their reactive state, can also promote neuronal damage, which contributes to neurodegenerative and neuropsychiatric diseases. Understanding the dual role of glia-as both protectors and potential aggressors-sheds light on their complex contributions to disease etiology and pathology. By appropriately modulating glial activity, it may be possible to mitigate neurodegeneration and restore neuronal function. In this review, which originated from the International Society for Neurochemistry (ISN) Advanced School in 2019 held in Montreal, Canada, we first describe the critical importance of glia in the development and maintenance of a healthy nervous system as well as their contributions to neuronal damage and neurological disorders. We then discuss potential strategies to modulate glial activity during disease to protect and promote a properly functioning nervous system. We propose that targeting glial cells presents a promising therapeutic avenue for rebuilding the nervous system.
Collapse
Affiliation(s)
- Matthew D Demmings
- Neuroscience Program, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Luana da Silva Chagas
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Marianela E Traetta
- Instituto de Biología Celular y Neurociencia (IBCN), Facultad de Medicina, Conicet, Buenos Aires, Argentina
| | - Rui S Rodrigues
- University of Bordeaux, INSERM, Neurocentre Magendie U1215, Bordeaux, France
| | - Maria Florencia Acutain
- Instituto de Biología Celular y Neurociencia (IBCN), Facultad de Medicina, Conicet, Buenos Aires, Argentina
| | - Evgeny Barykin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER Raebareli), Raebareli, UP, India
| | - Liliana German-Castelan
- Neuroscience Program, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Vanesa S Mattera
- Instituto de Química y Fisicoquímica Biológica (IQUIFIB-FFyB-UBA), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pedzisai Mazengenya
- Center of Medical and bio-Allied Health Sciences Research, College of Medicine, Ajman University, Ajman, United Arab Emirates
| | - Cecilia Skoug
- Department of Neuroscience, Physiology & Pharmacology, Centre for Cardiovascular and Metabolic Neuroscience, University College London, London, UK
| | - Hisashi Umemori
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Kweon SH, Ryu HG, Kwon SH, Park H, Lee S, Kim NS, Ma SX, Jee HJ, Kim S, Ko HS. Gba1 E326K renders motor and non-motor symptoms with pathological α-synuclein, tau and glial activation. Brain 2024; 147:4072-4083. [PMID: 38976650 PMCID: PMC11629696 DOI: 10.1093/brain/awae222] [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: 09/20/2023] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 07/10/2024] Open
Abstract
Mutations in the GBA1 gene are common genetic risk factors for Parkinson's disease, disrupting enzymatic activity and causing lysosomal dysfunction, leading to elevated α-synuclein levels. Although the role of GBA1 in synucleinopathy is well established, recent research underscores neuroinflammation as a significant pathogenic mechanism in GBA1 deficiency. This study investigates neuroinflammation in Gba1 E326K knock-in mice, a model associated with increased risk of Parkinson's disease and dementia. At 9 and 24 months, we assessed GBA1 protein and activity, α-synuclein pathology, neurodegeneration, motor deficits and gliosis in the ventral midbrain and hippocampus using immunohistochemistry, western blot and glucocerebrosidase assays. Additionally, primary microglia from wild-type and Gba1E326K/E326K mice were treated with α-synuclein preformed fibrils to study microglia activation, pro-inflammatory cytokines, reactive astrocyte formation and neuronal death through quantitative PCR, western blot and immunocytochemistry analyses. We also evaluated the effects of gut inoculation of α-synuclein preformed fibrils in Gba1 E326K mice at 7 months and striatal inoculation at 10 months after injection, assessing motor/non-motor symptoms, α-synuclein pathology, neuroinflammation, gliosis and neurodegeneration via behavioural tests, immunohistochemistry and western blot assays. At 24 months, Gba1 E326K knock-in mice showed reduced glucocerebrosidase enzymatic activity and glucosylceramide build-up in the ventral midbrain and hippocampus. Increased pro-inflammatory cytokines and reactive astrocytes were observed in microglia and astrocytes from Gba1 E326K mice treated with pathological α-synuclein preformed fibrils. Gut inoculation of α-synuclein preformed fibrils increased Lewy body accumulation in the hippocampal dentate gyrus, with heightened microglia and astrocyte activation and worsened non-motor symptoms. Intrastriatal injection of α-synuclein preformed fibrils induced motor deficits, reactive glial protein accumulation and tauopathy in the prefrontal cortex and hippocampus of Gba1 E326K mice. GBA1 deficiency attributable to the Gba1 E326K mutation exacerbates neuroinflammation and promotes pathogenic α-synuclein transmission, intensifying disease pathology in Parkinson's disease models. This study enhances our understanding of how the Gba1 E326K mutation contributes to neuroinflammation and the spread of pathogenic α-synuclein in the brain, suggesting new therapeutic strategies for Parkinson's disease and related synucleinopathies.
Collapse
Affiliation(s)
- Sin Ho Kweon
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hye Guk Ryu
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Seung-Hwan Kwon
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hyeonwoo Park
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Saebom Lee
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Nam-Shik Kim
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shi-Xun Ma
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hee-Jung Jee
- Department of Information and Statistics, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Sangjune Kim
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Han Seok Ko
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
5
|
Duffy HB, Byrnes C, Zhu H, Tuymetova G, Lee YT, Platt FM, Proia RL. Deletion of Gba in neurons, but not microglia, causes neurodegeneration in a Gaucher mouse model. JCI Insight 2024; 9:e179126. [PMID: 39312723 PMCID: PMC11601582 DOI: 10.1172/jci.insight.179126] [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: 01/10/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024] Open
Abstract
Gaucher disease, the most prevalent lysosomal storage disease, is caused by homozygous mutations at the GBA gene, which is responsible for encoding the enzyme glucocerebrosidase. Neuronopathic Gaucher disease is associated with microgliosis, astrogliosis, and neurodegeneration. However, the role that microglia, astrocytes, and neurons play in the disease remains to be determined. In the current study, we developed inducible, cell-type-specific Gba-KO mice to better understand the individual impacts of Gba deficiencies on microglia and neurons. Gba was conditionally knocked out either exclusively in microglia or neurons or throughout the body. These mouse models were developed using a tamoxifen-inducible Cre system, with tamoxifen administration commencing at weaning. Microglia-specific Gba-KO mice showed no signs of disease. However, the neuron-specific Gba KO resulted in a shortened lifespan, severe weight loss, and ataxia. These mice also had significant neurodegeneration, microgliosis, and astrogliosis accompanied by the accumulation of glucosylceramide and glucosylsphingosine, recapitulating Gaucher disease-like symptoms. These surprising findings reveal that, unlike the neuron-specific Gba deficiency, microglia-specific Gba deficiency alone does not induce disease. The neuronal Gaucher disease mouse model, with a median survival of 16 weeks, may be useful for future studies of pathogenesis and the evaluation of therapies.
Collapse
Affiliation(s)
- Hannah B.D. Duffy
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Colleen Byrnes
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Hongling Zhu
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Galina Tuymetova
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Y. Terry Lee
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Frances M. Platt
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Richard L. Proia
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
6
|
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
|
7
|
Feng T, Zheng H, Zhang Z, Fan P, Yang X. Mechanism and therapeutic targets of the involvement of a novel lysosomal proton channel TMEM175 in Parkinson's disease. Ageing Res Rev 2024; 100:102373. [PMID: 38960046 DOI: 10.1016/j.arr.2024.102373] [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: 02/11/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024]
Abstract
Parkinson's disease (PD), recognized as the second most prevalent neurodegenerative disease in the aging population, presents a significant challenge due to the current lack of effective treatment methods to mitigate its progression. Many pathogenesis of PD are related to lysosomal dysfunction. Moreover, extensive genetic studies have shown a significant correlation between the lysosomal membrane protein TMEM175 and the risk of developing PD. Building on this discovery, TMEM175 has been identified as a novel potassium ion channel. Intriguingly, further investigations have found that potassium ion channels gradually close and transform into hydrion "excretion" channels in the microenvironment of lysosomes. This finding was further substantiated by studies on TMEM175 knockout mice, which exhibited pronounced motor dysfunction in pole climbing and suspension tests, alongside a notable reduction in dopamine neurons within the substantia nigra compacta. Despite these advancements, the current research landscape is not without its controversies. In light of this, the present review endeavors to methodically examine and consolidate a vast array of recent literature on TMEM175. This comprehensive analysis spans from the foundational research on the structure and function of TMEM175 to expansive population genetics studies and mechanism research utilizing cellular and animal models.A thorough understanding of the structure and function of TMEM175, coupled with insights into the intricate mechanisms underpinning lysosomal dysfunction in PD dopaminergic neurons, is imperative. Such knowledge is crucial for pinpointing precise intervention targets, thereby paving the way for novel therapeutic strategies that could potentially alter the neurodegenerative trajectory of PD.
Collapse
Affiliation(s)
- Tingting Feng
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, China; Xinjiang Key Laboratory of Nervous System Disease Research, Urumqi 830063,China; Xinjiang Clinical Research Center for Nervous System Diseases, Urumqi 830063, China; Xinjiang Medical University, Urumqi 830017, China
| | | | - Zhan Zhang
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, China; Xinjiang Key Laboratory of Nervous System Disease Research, Urumqi 830063,China; Xinjiang Clinical Research Center for Nervous System Diseases, Urumqi 830063, China
| | - Peidong Fan
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, China; Xinjiang Key Laboratory of Nervous System Disease Research, Urumqi 830063,China; Xinjiang Clinical Research Center for Nervous System Diseases, Urumqi 830063, China
| | - Xinling Yang
- Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, China; Xinjiang Key Laboratory of Nervous System Disease Research, Urumqi 830063,China; Xinjiang Clinical Research Center for Nervous System Diseases, Urumqi 830063, China; Xinjiang Medical University, Urumqi 830017, China.
| |
Collapse
|
8
|
Hertz E, Chen Y, Sidransky E. Gaucher disease provides a unique window into Parkinson disease pathogenesis. Nat Rev Neurol 2024; 20:526-540. [PMID: 39107435 DOI: 10.1038/s41582-024-00999-z] [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] [Accepted: 07/08/2024] [Indexed: 09/04/2024]
Abstract
An exciting development in the field of neurodegeneration is the association between the rare monogenic disorder Gaucher disease and the common complex disorder Parkinson disease (PD). Gaucher disease is a lysosomal storage disorder resulting from an inherited deficiency of the enzyme glucocerebrosidase, encoded by GBA1, which hydrolyses the glycosphingolipids glucosylceramide and glucosylsphingosine. The observation of parkinsonism in a rare subgroup of individuals with Gaucher disease first directed attention to the role of glucocerebrosidase deficiency in the pathogenesis of PD. PD occurs more frequently in people heterozygous for Gaucher GBA1 mutations, and 3-25% of people with Parkinson disease carry a GBA1 variant. However, only a small percentage of individuals with GBA1 variants develop parkinsonism, suggesting that the penetrance is low. Despite over a decade of intense research in this field, including clinical and radiological evaluations, genetic studies and investigations using model systems, the mechanism underlying GBA1-PD is still being pursued. Insights from this association have emphasized the role of lysosomal pathways in parkinsonism. Furthermore, different therapeutic strategies considered or developed for Gaucher disease can now inform drug development for PD.
Collapse
Affiliation(s)
- Ellen Hertz
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yu Chen
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ellen Sidransky
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
9
|
Kweon SH, Ryu HG, Park H, Lee S, Kim N, Kwon SH, Ma SX, Kim S, Ko HS. Linking Gba1 E326K mutation to microglia activation and mild age-dependent dopaminergic Neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.14.557673. [PMID: 37745332 PMCID: PMC10515932 DOI: 10.1101/2023.09.14.557673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Mutations in the GBA1 gene have been identified as a prevalent genetic risk factor for Parkinson's disease (PD). GBA1 mutations impair enzymatic activity, leading to lysosomal dysfunction and elevated levels of α-synuclein (α-syn). While most research has primarily focused on GBA1's role in promoting synucleinopathy, emerging evidence suggests that neuroinflammation may be a key pathogenic alteration caused by GBA1 deficiency. To examine the molecular mechanism underlying GBA1 deficiency-mediated neuroinflammation, we generated Gba1 E326K knock-in (KI) mice using the CRISPR/Cas9 technology, which is linked to an increased risk of PD and dementia with Lewy bodies (DLB). In the ventral midbrain and hippocampus of 24-month-old Gba1 E326K KI mice, we found a moderate decline in GBA1 enzymatic activity, a buildup of glucosylceramide, and an increase in microglia density. Furthermore, we observed increased levels of pro-inflammatory cytokines and formation of reactive astrocytes in primary microglia and astrocytes, respectively, cultured from Gba1 E326K KI mice following treatment with pathologic α-syn preformed fibrils (PFF). Additionally, the gut inoculation of α-syn PFF in Gba1 E326K KI mice significantly enhanced the accumulation of Lewy bodies in the dentate gyrus of the hippocampus, accompanied by aggravated neuroinflammation and exacerbated non-motor symptoms. This research significantly enhances our understanding of the Gba1 E326K mutation's involvement in neuroinflammation and the cell-to-cell transmission of pathogenic α-syn in the brain, thereby opening new therapeutic avenues.
Collapse
|
10
|
Dobert JP, Bub S, Mächtel R, Januliene D, Steger L, Regensburger M, Wilfling S, Chen J, Dejung M, Plötz S, Hehr U, Moeller A, Arnold P, Zunke F. Activation and Purification of ß-Glucocerebrosidase by Exploiting its Transporter LIMP-2 - Implications for Novel Treatment Strategies in Gaucher's and Parkinson's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401641. [PMID: 38666485 PMCID: PMC11220700 DOI: 10.1002/advs.202401641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/15/2024] [Indexed: 07/04/2024]
Abstract
Genetic variants of GBA1 can cause the lysosomal storage disorder Gaucher disease and are among the highest genetic risk factors for Parkinson's disease (PD). GBA1 encodes the lysosomal enzyme beta-glucocerebrosidase (GCase), which orchestrates the degradation of glucosylceramide (GluCer) in the lysosome. Recent studies have shown that GluCer accelerates α-synuclein aggregation, exposing GCase deficiency as a major risk factor in PD pathology and as a promising target for treatment. This study investigates the interaction of GCase and three disease-associated variants (p.E326K, p.N370S, p.L444P) with their transporter, the lysosomal integral membrane protein 2 (LIMP-2). Overexpression of LIMP-2 in HEK 293T cells boosts lysosomal abundance of wt, E326K, and N370S GCase and increases/rescues enzymatic activity of the wt and E326K variant. Using a novel purification approach, co-purification of untagged wt, E326K, and N370S GCase in complex with His-tagged LIMP-2 from cell supernatant of HEK 293F cells is achieved, confirming functional binding and trafficking for these variants. Furthermore, a single helix in the LIMP-2 ectodomain is exploited to design a lysosome-targeted peptide that enhances lysosomal GCase activity in PD patient-derived and control fibroblasts. These findings reveal LIMP-2 as an allosteric activator of GCase, suggesting a possible therapeutic potential of targeting this interaction.
Collapse
Affiliation(s)
- Jan Philipp Dobert
- Department of Molecular NeurologyUniversity Hospital ErlangenFriedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)91054ErlangenGermany
| | - Simon Bub
- Department of Molecular NeurologyUniversity Hospital ErlangenFriedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)91054ErlangenGermany
| | - Rebecca Mächtel
- Department of Molecular NeurologyUniversity Hospital ErlangenFriedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)91054ErlangenGermany
| | - Dovile Januliene
- Department of Structural BiologyOsnabrueck University49076OsnabrueckGermany
| | - Lisa Steger
- Department of Molecular NeurologyUniversity Hospital ErlangenFriedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)91054ErlangenGermany
| | - Martin Regensburger
- Department of Molecular NeurologyUniversity Hospital ErlangenFriedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)91054ErlangenGermany
- Deutsches Zentrum Immuntherapie (DZI)Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)91054ErlangenGermany
| | | | - Jia‐Xuan Chen
- Institute of Molecular Biology (IMB)55128MainzGermany
| | - Mario Dejung
- Institute of Molecular Biology (IMB)55128MainzGermany
| | - Sonja Plötz
- Department of Molecular NeurologyUniversity Hospital ErlangenFriedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)91054ErlangenGermany
| | - Ute Hehr
- Center for Human Genetics Regensburg93059RegensburgGermany
| | - Arne Moeller
- Department of Structural BiologyOsnabrueck University49076OsnabrueckGermany
| | - Philipp Arnold
- Institute of AnatomyFunctional and Clinical AnatomyFriedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)91054ErlangenGermany
| | - Friederike Zunke
- Department of Molecular NeurologyUniversity Hospital ErlangenFriedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)91054ErlangenGermany
| |
Collapse
|
11
|
Siddiqui T, Bhatt LK. Emerging autophagic endo-lysosomal targets in the management of Parkinson's disease. Rev Neurol (Paris) 2024; 180:477-485. [PMID: 37586941 DOI: 10.1016/j.neurol.2023.07.007] [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: 11/15/2022] [Revised: 06/26/2023] [Accepted: 07/04/2023] [Indexed: 08/18/2023]
Abstract
Synucleopathies, specifically Parkinson's disease, are still incurable and available therapeutic options are scarce and symptomatic. The autophagy-lysosomal-endosomal system is an indigenous mechanism to manage the proteome. Excess/misfolded protein accumulation activates this system, which degrades the undesired proteins via lysosomes. Cells also eliminate these proteins by releasing them into the extracellular space via exosomes. However, the sutophagy-lysosomal-endosomal system becomes unfunctional in Parkinson's disease and there is accumulation and spread of pathogenic alpha-synuclein. Neuronal degeneration results Owing to pathogenic alpha-synuclein. Thus, the autophagy-lysosomal-endosomal system could be a promising target for neuroprotection. In the present review, we discuss the autophagy-lysosomal-endosomal system as an emerging target for the management of Parkinson's disease. Modulation of these targets associated with the autophagy-lysosomal-endosomal system can aid in clearing pathogenic alpha-synuclein and prevent the degeneration of neurons.
Collapse
Affiliation(s)
- T Siddiqui
- Department of Pharmacology, SVKM's Doctor Bhanuben-Nanavati College of Pharmacy, Vile Parle (West), Mumbai, India
| | - L K Bhatt
- Department of Pharmacology, SVKM's Doctor Bhanuben-Nanavati College of Pharmacy, Vile Parle (West), Mumbai, India.
| |
Collapse
|
12
|
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
|
13
|
Wang R, Sun H, Cao Y, Zhang Z, Chen Y, Wang X, Liu L, Wu J, Xu H, Wu D, Mu C, Hao Z, Qin S, Ren H, Han J, Fang M, Wang G. Glucosylceramide accumulation in microglia triggers STING-dependent neuroinflammation and neurodegeneration in mice. Sci Signal 2024; 17:eadk8249. [PMID: 38530880 DOI: 10.1126/scisignal.adk8249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 03/06/2024] [Indexed: 03/28/2024]
Abstract
Mutations in the gene encoding the lysosomal enzyme glucocerebrosidase (GCase) are responsible for Gaucher disease (GD) and are considered the strongest genetic risk factor for Parkinson's disease (PD) and Lewy body dementia (LBD). GCase deficiency leads to extensive accumulation of glucosylceramides (GCs) in cells and contributes to the neuropathology of GD, PD, and LBD by triggering chronic neuroinflammation. Here, we investigated the mechanisms by which GC accumulation induces neuroinflammation. We found that GC accumulation within microglia induced by pharmacological inhibition of GCase triggered STING-dependent inflammation, which contributed to neuronal loss both in vitro and in vivo. GC accumulation in microglia induced mitochondrial DNA (mtDNA) leakage to the cytosol to trigger STING-dependent inflammation. Rapamycin, a compound that promotes lysosomal activity, improved mitochondrial function, thereby decreasing STING signaling. Furthermore, lysosomal damage caused by GC accumulation led to defects in the degradation of activated STING, further exacerbating inflammation mediated by microglia. Thus, limiting STING activity may be a strategy to suppress neuroinflammation caused by GCase deficiency.
Collapse
Affiliation(s)
- Rui Wang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210096, China
- Center of Translational Medicine, First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215400, China
| | - Hongyang Sun
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yifan Cao
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhixiong Zhang
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yajing Chen
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Xiying Wang
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai 200000, China
| | - Lele Liu
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jin Wu
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Hao Xu
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Dan Wu
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chenchen Mu
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zongbing Hao
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Song Qin
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai 200000, China
| | - Haigang Ren
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
- Jiangsu Provincial Medical Innovation Center of Trauma Medicine, Institute of Trauma Medicine, Suzhou, Jiangsu 215123, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Junhai Han
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210096, China
| | - Ming Fang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210096, China
| | - Guanghui Wang
- Center of Translational Medicine, First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215400, China
- Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
14
|
Gregorio I, Russo L, Torretta E, Barbacini P, Contarini G, Pacinelli G, Bizzotto D, Moriggi M, Braghetta P, Papaleo F, Gelfi C, Moro E, Cescon M. GBA1 inactivation in oligodendrocytes affects myelination and induces neurodegenerative hallmarks and lipid dyshomeostasis in mice. Mol Neurodegener 2024; 19:22. [PMID: 38454456 PMCID: PMC10921719 DOI: 10.1186/s13024-024-00713-z] [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: 06/28/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Mutations in the β-glucocerebrosidase (GBA1) gene do cause the lysosomal storage Gaucher disease (GD) and are among the most frequent genetic risk factors for Parkinson's disease (PD). So far, studies on both neuronopathic GD and PD primarily focused on neuronal manifestations, besides the evaluation of microglial and astrocyte implication. White matter alterations were described in the central nervous system of paediatric type 1 GD patients and were suggested to sustain or even play a role in the PD process, although the contribution of oligodendrocytes has been so far scarcely investigated. METHODS We exploited a system to study the induction of central myelination in vitro, consisting of Oli-neu cells treated with dibutyryl-cAMP, in order to evaluate the expression levels and function of β-glucocerebrosidase during oligodendrocyte differentiation. Conduritol-B-epoxide, a β-glucocerebrosidase irreversible inhibitor was used to dissect the impact of β-glucocerebrosidase inactivation in the process of myelination, lysosomal degradation and α-synuclein accumulation in vitro. Moreover, to study the role of β-glucocerebrosidase in the white matter in vivo, we developed a novel mouse transgenic line in which β-glucocerebrosidase function is abolished in myelinating glia, by crossing the Cnp1-cre mouse line with a line bearing loxP sequences flanking Gba1 exons 9-11, encoding for β-glucocerebrosidase catalytic domain. Immunofluorescence, western blot and lipidomic analyses were performed in brain samples from wild-type and knockout animals in order to assess the impact of genetic inactivation of β-glucocerebrosidase on myelination and on the onset of early neurodegenerative hallmarks, together with differentiation analysis in primary oligodendrocyte cultures. RESULTS Here we show that β-glucocerebrosidase inactivation in oligodendrocytes induces lysosomal dysfunction and inhibits myelination in vitro. Moreover, oligodendrocyte-specific β-glucocerebrosidase loss-of-function was sufficient to induce in vivo demyelination and early neurodegenerative hallmarks, including axonal degeneration, α-synuclein accumulation and astrogliosis, together with brain lipid dyshomeostasis and functional impairment. CONCLUSIONS Our study sheds light on the contribution of oligodendrocytes in GBA1-related diseases and supports the need for better characterizing oligodendrocytes as actors playing a role in neurodegenerative diseases, also pointing at them as potential novel targets to set a brake to disease progression.
Collapse
Affiliation(s)
- Ilaria Gregorio
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Loris Russo
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Enrica Torretta
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, Milan, 20161, Italy
| | - Pietro Barbacini
- Department of Biomedical Sciences for Health, University of Milan, 20133, Milan, Italy
| | - Gabriella Contarini
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano Di Tecnologia, 16163, Genova, Italy
- Department of Biomedical and Technological Sciences, University of Catania, 95125, Catania, Italy
| | - Giada Pacinelli
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano Di Tecnologia, 16163, Genova, Italy
- Padova Neuroscience Center (PNC), University of Padova, 35131, Padua, Italy
| | - Dario Bizzotto
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Manuela Moriggi
- Department of Biomedical Sciences for Health, University of Milan, 20133, Milan, Italy
| | - Paola Braghetta
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano Di Tecnologia, 16163, Genova, Italy
| | - Cecilia Gelfi
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, Milan, 20161, Italy
- Department of Biomedical Sciences for Health, University of Milan, 20133, Milan, Italy
| | - Enrico Moro
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy.
| |
Collapse
|
15
|
Vernet Machado Bressan Wilke M, Iop GD, Faqueti L, Lemos da Silva LA, Kubaski F, Poswar FO, Michelin-Tirelli K, Randon D, Borelli WV, Giugliani R, Schwartz IVD. A Brazilian Rare-Disease Center's Experience with Glucosylsphingosine (lyso-Gb1) in Patients with Gaucher Disease: Exploring a Novel Correlation with IgG Levels in Plasma and a Biomarker Measurement in CSF. Int J Mol Sci 2024; 25:2870. [PMID: 38474117 PMCID: PMC10931658 DOI: 10.3390/ijms25052870] [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: 01/22/2024] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Gaucher disease (GD, OMIM 230800) is one of the most common lysosomal disorders, being caused by the deficient activity of the enzyme acid β-glucocerebrosidase (Gcase). Three clinical forms of Gaucher's disease (GD) are classified based on neurological involvement. Type 1 (GD1) is non-neuronopathic, while types 2 (GD2) and 3 (GD3) are neuronopathic forms. Gcase catalyzes the conversion of glucosylceramide (GlcCer) into ceramide and glucose. As GlcCer accumulates in lysosomal macrophages, it undergoes deacylation to become glycosylsphingosine (lyso-Gb1), which has shown to be a useful and reliable biomarker for the diagnosis and monitoring of treated and untreated patients with GD. Multiple myeloma (MM) is one of the leading causes of cancer-related death among patients with GD and monoclonal gammopathy of undetermined significance (MGUS) is a non-neoplastic condition that can be a telltale sign of a B clonal proliferation caused by the chronic activation of B cells. This study aimed to quantify Lyso-Gb1 levels in dried blood spots (DBS) and cerebrospinal fluid (CSF) as biomarkers for Gaucher disease (GD) and discuss the association of this biomarker with other clinical parameters. This is a mixed-methods study incorporating both cross-sectional and longitudinal elements within a cohort design with a convenience-sampling strategy. Data collection took place from January 2012 to March 2023. Lyso-Gb1 extraction from DBS involved the use of a methanol-acetonitrile-water mixture, followed by incubation and centrifugation. Analysis was performed using UPLC-MS/MS with MassLynx software version 4.2 and the control group for the DBS measurements included general newborns. CSF Lyso-Gb1 was extracted using ethyl acetate, analyzed by UPLC-MS/MS with a calibration curve, and expressed in pmol/L. Lysosomal activity in CSF was assessed by measuring chitotriosidase (Cht), and other lysosomal enzyme activities were assessed as previously described in the literature. Patients with metachromatic leukodystrophy (MLD) were used as controls. Thirty-two treated patients (twenty-nine GD1 and three GD3, all on ERT except for one GD type on SRT with eliglustat) and three untreated patients (one GD1, one GD2, and one GD3) were included. When analyzing only the treated GD1 group, a significant correlation was found between lyso-Gb1 and age (rho = -0.447, p = 0.001), ChT, and IgG levels (rho = 0.73, p < 0.001; and rho = 0.36, p = 0.03, respectively). Five GD1 patients (three females, mean age 40 years) also had their CSF collected and analyzed. The average measurement of lyso-Gb1 in CSF was 94 pmol/L (range: 57.1-157.9 pmol/L) versus <6.2 pmol/L in the control group (MLD). This is the first time, to the best of our knowledge, that lyso-Gb1 has been associated with IgG levels. While this finding reflects a risk for MGUS or MM and not only chronic plasma B-cell activation, it still requires further studies. Moreover, the analysis of CSF lyso-Gb1 levels in GD1 patients was demonstrated to be significantly higher than the control group. This raises the hypothesis that CSF lyso-Gb1 may serve as a valuable indicator for neurological involvement in GD, providing insights into the potential implications for neurological manifestations in GD, including GD1. The correlation between lyso-Gb1 and ChT levels in treated GD1 patients further underscores the interconnectedness of lysosomal markers and their relevance in monitoring.
Collapse
Affiliation(s)
| | - Gabrielle Dineck Iop
- Biodiscovery Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035903, Brazil; (G.D.I.); (L.F.); (L.A.L.d.S.)
| | - Larissa Faqueti
- Biodiscovery Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035903, Brazil; (G.D.I.); (L.F.); (L.A.L.d.S.)
| | - Layzon Antonio Lemos da Silva
- Biodiscovery Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035903, Brazil; (G.D.I.); (L.F.); (L.A.L.d.S.)
| | - Francyne Kubaski
- Biochemical Genetics Laboratory, Greenwood Genetics Center, Greenwood, SC 29646, USA;
| | - Fabiano O. Poswar
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035003, Brazil;
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035003, Brazil
| | - Kristiane Michelin-Tirelli
- LEIM-Biochemical Genetics Laboratory, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035003, Brazil;
| | - Dévora Randon
- BRAIN Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035003, Brazil
| | | | - Roberto Giugliani
- Postgraduate Program in Medical Sciences, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre 90035003, Brazil; (M.V.M.B.W.); (R.G.)
- Biodiscovery Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035903, Brazil; (G.D.I.); (L.F.); (L.A.L.d.S.)
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035003, Brazil;
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035003, Brazil
- Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035003, Brazil
- Dasa Genômica, São Paulo 04078013, Brazil
- Casa dos Raros, Porto Alegre 90035003, Brazil
| | - Ida Vanessa D. Schwartz
- Postgraduate Program in Medical Sciences, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre 90035003, Brazil; (M.V.M.B.W.); (R.G.)
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035003, Brazil;
- BRAIN Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035003, Brazil
- Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035003, Brazil
- Instituto Nacional de Doenças Raras—InRaras, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035003, Brazil
| |
Collapse
|
16
|
Kou L, Chi X, Sun Y, Yin S, Wu J, Zou W, Wang Y, Jin Z, Huang J, Xiong N, Xia Y, Wang T. Circadian regulation of microglia function: Potential targets for treatment of Parkinson's Disease. Ageing Res Rev 2024; 95:102232. [PMID: 38364915 DOI: 10.1016/j.arr.2024.102232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/11/2024] [Accepted: 02/11/2024] [Indexed: 02/18/2024]
Abstract
Circadian rhythms are involved in the regulation of many aspects of the body, including cell function, physical activity and disease. Circadian disturbance often predates the typical symptoms of neurodegenerative diseases and is not only a non-motor symptom, but also one of the causes of their occurrence and progression. Glial cells possess circadian clocks that regulate their function to maintain brain development and homeostasis. Emerging evidence suggests that the microglial circadian clock is involved in the regulation of many physiological processes, such as cytokine release, phagocytosis, and nutritional and metabolic support, and that disruption of the microglia clock may affect multiple aspects of Parkinson's disease, especially neuroinflammation and α-synuclein processes. Herein, we review recent advances in the circadian control of microglia function in health and disease, and discuss novel pharmacological interventions for microglial clocks in neurodegenerative disorders.
Collapse
Affiliation(s)
- Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaosa Chi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yadi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiawei Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenkai Zou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yiming Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zongjie Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
17
|
Cooper O, Hallett P, Isacson O. Upstream lipid and metabolic systems are potential causes of Alzheimer's disease, Parkinson's disease and dementias. FEBS J 2024; 291:632-645. [PMID: 36165619 PMCID: PMC10040476 DOI: 10.1111/febs.16638] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/02/2022] [Accepted: 09/26/2022] [Indexed: 11/28/2022]
Abstract
Brain health requires circuits, cells and molecular pathways to adapt when challenged and to promptly reset once the challenge has resolved. Neurodegeneration occurs when adaptability becomes confined, causing challenges to overwhelm neural circuitry. Studies of rare and common neurodegenerative diseases suggest that the accumulation of lipids can compromise circuit adaptability. Using microglia as an example, we review data that suggest increased lipid concentrations cause dysfunctional inflammatory responses to immune challenges, leading to Alzheimer's disease, Parkinson's disease and dementia. We highlight current approaches to treat lipid metabolic and clearance pathways and identify knowledge gaps towards restoring adaptive homeostasis in individuals who are at-risk of losing cognition.
Collapse
Affiliation(s)
- Oliver Cooper
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| | - Penny Hallett
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| | - Ole Isacson
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| |
Collapse
|
18
|
Elangovan A, Dahiya B, Kirola L, Iyer M, Jeeth P, Maharaj S, Kumari N, Lakhanpal V, Michel TM, Rao KRSS, Cho SG, Yadav MK, Gopalakrishnan AV, Kadhirvel S, Kumar NS, Vellingiri B. Does gut brain axis has an impact on Parkinson's disease (PD)? Ageing Res Rev 2024; 94:102171. [PMID: 38141735 DOI: 10.1016/j.arr.2023.102171] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/04/2023] [Accepted: 12/13/2023] [Indexed: 12/25/2023]
Abstract
Parkinson's Disease (PD) is becoming a growing global concern by being the second most prevalent disease next to Alzheimer's Disease (AD). Henceforth new exploration is needed in search of new aspects towards the disease mechanism and origin. Evidence from recent studies has clearly stated the role of Gut Microbiota (GM) in the maintenance of the brain and as a root cause of various diseases and disorders including other neurological conditions. In the case of PD, with an unknown etiology, the GM is said to have a larger impact on the disease pathophysiology. Although GM and its metabolites are crucial for maintaining the normal physiology of the host, it is an undeniable fact that there is an influence of GM in the pathophysiology of PD. As such the Enteroendocrine Cells (EECs) in the epithelium of the intestine are one of the significant regulators of the gut-brain axis and act as a communication mediator between the gut and the brain. The communication is established via the molecules of neuroendocrine which are said to have a crucial part in neurological diseases such as AD, PD, and other psychiatry-related disorders. This review is focused on understanding the proper role of GM and EECs in PD. Here, we also focus on some of the metabolites and compounds that can interact with the PD genes causing various dysfunctions in the cell and facilitating the disease conditions using bioinformatical tools. Various mechanisms concerning EECs and PD, their identification, the latest studies, and available current therapies have also been discussed.
Collapse
Affiliation(s)
- Ajay Elangovan
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Bhawna Dahiya
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Laxmi Kirola
- Department of Biotechnology, School of Health Sciences and Technology (SoHST), UPES University, Dehradun, Uttarakhand 248007, India
| | - Mahalaxmi Iyer
- Department of Microbiology, Central University of Punjab, Bathinda 151401, Punjab, India; Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to be University), Coimbatore 641021, Tamil Nadu, India
| | - Priyanka Jeeth
- Department of Computational Sciences, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Sakshi Maharaj
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Nikki Kumari
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Vikas Lakhanpal
- Department of Neurology, All India Institute of Medical Sciences, Bathinda 151005, Punjab, India
| | - Tanja Maria Michel
- Research Unit of Psychiatry, Dept. of Psychiatry Odense, Clinical Institute, University of Southern Denmark, J.B. Winslowsvej 20, Indg. 220B, Odense, Denmark
| | - K R S Sambasiva Rao
- Mangalayatan University - Jabalpur, Jabalpur - 481662, Madhya Pradesh, India
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Mukesh Kumar Yadav
- Department of Microbiology, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632 014, India
| | - Saraboji Kadhirvel
- Department of Computational Sciences, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Nachimuthu Senthil Kumar
- Department of Biotechnology, Mizoram University (A Central University), Aizawl, 796 004 Mizoram, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda 151401, Punjab, India.
| |
Collapse
|
19
|
Xue J, Tao K, Wang W, Wang X. What Can Inflammation Tell Us about Therapeutic Strategies for Parkinson's Disease? Int J Mol Sci 2024; 25:1641. [PMID: 38338925 PMCID: PMC10855787 DOI: 10.3390/ijms25031641] [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: 01/01/2024] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder with a complicated etiology and pathogenesis. α-Synuclein aggregation, dopaminergic (DA) neuron loss, mitochondrial injury, oxidative stress, and inflammation are involved in the process of PD. Neuroinflammation has been recognized as a key element in the initiation and progression of PD. In this review, we summarize the inflammatory response and pathogenic mechanisms of PD. Additionally, we describe the potential anti-inflammatory therapies, including nod-like receptor pyrin domain containing protein 3 (NLRP3) inflammasome inhibition, nuclear factor κB (NF-κB) inhibition, microglia inhibition, astrocyte inhibition, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibition, the peroxisome proliferator-activated receptor γ (PPARγ) agonist, targeting the mitogen-activated protein kinase (MAPK) pathway, targeting the adenosine monophosphate-activated protein kinase (AMPK)-dependent pathway, targeting α-synuclein, targeting miRNA, acupuncture, and exercise. The review focuses on inflammation and will help in designing new prevention strategies for PD.
Collapse
Affiliation(s)
- Jinsong Xue
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China; (K.T.); (W.W.)
| | | | | | - Xiaofei Wang
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China; (K.T.); (W.W.)
| |
Collapse
|
20
|
Sato S, Matsumoto SI, Kosugi Y. Quantitation and characterization of glucosylsphingosine in cerebrospinal fluid (CSF), plasma, and brain of monkey model with Gaucher disease. Drug Metab Pharmacokinet 2023; 53:100530. [PMID: 37924723 DOI: 10.1016/j.dmpk.2023.100530] [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: 02/10/2023] [Revised: 08/18/2023] [Accepted: 09/12/2023] [Indexed: 11/06/2023]
Abstract
Treatment with conduritol-β-epoxide (CBE) in preclinical species is expected to be a powerful approach to generate animal models of Gaucher disease (GD) and Parkinson's disease associated with heterozygous mutations in Glucocerebrosidase (GBA-PD). However, it is not fully elucidated how quantitatively the change in glucosylsphingosine (GlcSph) levels in cerebrospinal fluid (CSF) correlates with that in the brain, which is expected to be clinically informative. Herein, we aimed to investigate the correlation with successfully quantified GlcSph in monkey CSF by developing highly sensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS) methods. The GlcSph in normal monkey CSF was 0.635 ± 0.177 pg/mL at baseline and increased by CBE treatment at 3 mg/kg daily for five days up to a moderate level, comparable to that in GD patients. The balance between GlcSph and galactosylsphingosine (GalSph) in the CSF matched that in the brain rather than plasma. In addition, GlcSph in the CSF was increased, accompanied by that in the brain at a dose of 3 mg/kg daily. These results indicate that GlcSph in the CSF is worth evaluating for concentration changes in the brain. Thus, this model can be useful for evaluating GBA-related diseases such as GD and GBA-PD.
Collapse
Affiliation(s)
- Sho Sato
- Drug Metabolism & Pharmacokinetics Research Laboratories, Preclinical & Translational Sciences, Research, Takeda Pharmaceutical Company Limited, Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan.
| | - Shin-Ichi Matsumoto
- Drug Metabolism & Pharmacokinetics Research Laboratories, Preclinical & Translational Sciences, Research, Takeda Pharmaceutical Company Limited, Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Yohei Kosugi
- Drug Metabolism & Pharmacokinetics Research Laboratories, Preclinical & Translational Sciences, Research, Takeda Pharmaceutical Company Limited, Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| |
Collapse
|
21
|
Connolly KJ, Margaria J, Di Biase E, Cooper O, Hallett PJ, Isacson O. Loss of Lipid Carrier ApoE Exacerbates Brain Glial and Inflammatory Responses after Lysosomal GBA1 Inhibition. Cells 2023; 12:2564. [PMID: 37947642 PMCID: PMC10647680 DOI: 10.3390/cells12212564] [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: 08/04/2023] [Revised: 09/26/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Tightly regulated and highly adaptive lipid metabolic and transport pathways are critical to maintaining brain cellular lipid homeostasis and responding to lipid and inflammatory stress to preserve brain function and health. Deficits in the lipid handling genes APOE and GBA1 are the most significant genetic risk factors for Lewy body dementia and related dementia syndromes. Parkinson's disease patients who carry both APOE4 and GBA1 variants have accelerated cognitive decline compared to single variant carriers. To investigate functional interactions between brain ApoE and GBA1, in vivo GBA1 inhibition was tested in WT versus ApoE-deficient mice. The experiments demonstrated glycolipid stress caused by GBA1 inhibition in WT mice induced ApoE expression in several brain regions associated with movement and dementia disorders. The absence of ApoE in ApoE-KO mice amplified complement C1q elevations, reactive microgliosis and astrocytosis after glycolipid stress. Mechanistically, GBA1 inhibition triggered increases in cell surface and intracellular lipid transporters ABCA1 and NPC1, respectively. Interestingly, the absence of NPC1 in mice also triggered elevations of brain ApoE levels. These new data show that brain ApoE, GBA1 and NPC1 functions are interconnected in vivo, and that the removal or reduction of ApoE would likely be detrimental to brain function. These results provide important insights into brain ApoE adaptive responses to increased lipid loads.
Collapse
Affiliation(s)
| | | | | | | | - Penelope J. Hallett
- Departments of Psychiatry and Neurology Harvard Medical School, Neuroregeneration Institute, McLean Hospital, Belmont, MA 02478, USA
| | - Ole Isacson
- Departments of Psychiatry and Neurology Harvard Medical School, Neuroregeneration Institute, McLean Hospital, Belmont, MA 02478, USA
| |
Collapse
|
22
|
Piovesana E, Magrin C, Ciccaldo M, Sola M, Bellotto M, Molinari M, Papin S, Paganetti P. Tau accumulation in degradative organelles is associated to lysosomal stress. Sci Rep 2023; 13:18024. [PMID: 37865674 PMCID: PMC10590387 DOI: 10.1038/s41598-023-44979-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/14/2023] [Indexed: 10/23/2023] Open
Abstract
Neurodegenerative disorders are characterized by the brain deposition of insoluble amyloidogenic proteins, such as α-synuclein or Tau, and the concomitant deterioration of cell functions such as the autophagy-lysosomal pathway (ALP). The ALP is involved in the degradation of intracellular macromolecules including protein aggregates. ALP dysfunction due to inherited defects in lysosomal or non-lysosomal proteins causes a group of diseases called lysosomal storage disorders (LSD) because of abnormal accumulation of lysosomal degradation substrates. Supporting the contribution of ALP defects in neurodegenerative diseases, deposition of amyloidogenic proteins occurs in LSD. Moreover, heterozygous mutations of several ALP genes represent risk factors for Parkinson's disease. The reciprocal contribution of α-synuclein accumulation and lysosomal dysfunction have been extensively studied. However, whether this adverse crosstalk also embraces Tau pathology needs more investigation. Here, we show in human primary fibroblasts that Tau seeds isolated from the brain of Alzheimer's disease induce Tau accumulation in acidic degradative organelles and lysosomal stress. Furthermore, inhibition of glucocerebrosidase, a lysosomal enzyme mutated in Gaucher's disease and a main risk for Parkinson's disease, causes lysosomal dysfunction in primary fibroblasts and contributes to the accumulation of Tau. Considering the presence of Tau lesions in Parkinson's disease as well as in multiple neurodegenerative disorders including Alzheimer's disease, our data call for further studies on strategies to alleviate ALP dysfunction as new therapeutic opportunity for neurodegenerative diseases and LSD.
Collapse
Affiliation(s)
- Ester Piovesana
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- PhD Program in Neurosciences, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Claudia Magrin
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- PhD Program in Neurosciences, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Matteo Ciccaldo
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Martina Sola
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- PhD Program in Neurosciences, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | | | - Maurizio Molinari
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera italiana, Bellinzona, Switzerland
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Stéphanie Papin
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Paolo Paganetti
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
- PhD Program in Neurosciences, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland.
- Neurocentro della Svizzera Italiana, Ente Ospedaliero Cantonale, Lugano, Switzerland.
| |
Collapse
|
23
|
Thomas R, Connolly KJ, Brekk OR, Hinrich AJ, Hastings ML, Isacson O, Hallett PJ. Viral-like TLR3 induction of cytokine networks and α-synuclein are reduced by complement C3 blockade in mouse brain. Sci Rep 2023; 13:15164. [PMID: 37704739 PMCID: PMC10499893 DOI: 10.1038/s41598-023-41240-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/23/2023] [Indexed: 09/15/2023] Open
Abstract
Inflammatory processes and mechanisms are of central importance in neurodegenerative diseases. In the brain, α-synucleinopathies such as Parkinson's disease (PD) and Lewy body dementia (LBD) show immune cytokine network activation and increased toll like receptor 3 (TLR3) levels for viral double-stranded RNA (dsRNA). Brain inflammatory reactions caused by TLR3 activation are also relevant to understand pathogenic cascades by viral SARS-CoV-2 infection causing post- COVID-19 brain-related syndromes. In the current study, following regional brain TLR3 activation induced by dsRNA in mice, an acute complement C3 response was seen at 2 days. A C3 splice-switching antisense oligonucleotide (ASO) that promotes the splicing of a non-productive C3 mRNA, prevented downstream cytokines, such as IL-6, and α-synuclein changes. This report is the first demonstration that α-synuclein increases occur downstream of complement C3 activation. Relevant to brain dysfunction, post-COVID-19 syndromes and pathological changes leading to PD and LBD, viral dsRNA TLR3 activation in the presence of C3 complement blockade further revealed significant interactions between complement systems, inflammatory cytokine networks and α-synuclein changes.
Collapse
Affiliation(s)
- Ria Thomas
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA
| | - Kyle J Connolly
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA
| | - Oeystein R Brekk
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA
| | - Anthony J Hinrich
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Michelle L Hastings
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Ole Isacson
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA.
| | - Penelope J Hallett
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA.
| |
Collapse
|
24
|
Spanos F, Deleidi M. Glycolipids in Parkinson's disease: beyond neuronal function. FEBS Open Bio 2023; 13:1558-1579. [PMID: 37219461 PMCID: PMC10476577 DOI: 10.1002/2211-5463.13651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/10/2023] [Accepted: 05/22/2023] [Indexed: 05/24/2023] Open
Abstract
Glycolipid balance is key to normal body function, and its alteration can lead to a variety of diseases involving multiple organs and tissues. Glycolipid disturbances are also involved in Parkinson's disease (PD) pathogenesis and aging. Increasing evidence suggests that glycolipids affect cellular functions beyond the brain, including the peripheral immune system, intestinal barrier, and immunity. Hence, the interplay between aging, genetic predisposition, and environmental exposures could initiate systemic and local glycolipid changes that lead to inflammatory reactions and neuronal dysfunction. In this review, we discuss recent advances in the link between glycolipid metabolism and immune function and how these metabolic changes can exacerbate immunological contributions to neurodegenerative diseases, with a focus on PD. Further understanding of the cellular and molecular mechanisms that control glycolipid pathways and their impact on both peripheral tissues and the brain will help unravel how glycolipids shape immune and nervous system communication and the development of novel drugs to prevent PD and promote healthy aging.
Collapse
Affiliation(s)
- Fokion Spanos
- Institut Imagine, INSERM UMR1163Paris Cité UniversityFrance
- Aligning Science Across Parkinson's (ASAP) Collaborative Research NetworkChevy ChaseMDUSA
| | - Michela Deleidi
- Institut Imagine, INSERM UMR1163Paris Cité UniversityFrance
- Aligning Science Across Parkinson's (ASAP) Collaborative Research NetworkChevy ChaseMDUSA
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain ResearchUniversity of TübingenGermany
| |
Collapse
|
25
|
Chatterjee D, Krainc D. Mechanisms of Glucocerebrosidase Dysfunction in Parkinson's Disease. J Mol Biol 2023; 435:168023. [PMID: 36828270 PMCID: PMC10247409 DOI: 10.1016/j.jmb.2023.168023] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023]
Abstract
Beta-glucocerebrosidase is a lysosomal hydrolase, encoded by GBA1 that represents the most common risk gene associated with Parkinson's disease (PD) and Lewy Body Dementia. Glucocerebrosidase dysfunction has been also observed in the absence of GBA1 mutations across different genetic and sporadic forms of PD and related disorders, suggesting a broader role of glucocerebrosidase in neurodegeneration. In this review, we highlight recent advances in mechanistic characterization of glucocerebrosidase function as the foundation for development of novel therapeutics targeting glucocerebrosidase in PD and related disorders.
Collapse
Affiliation(s)
- Diptaman Chatterjee
- Ken and Ruth Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA. https://twitter.com/NeilChatterBox
| | - Dimitri Krainc
- Ken and Ruth Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA; Simpson Querrey Center for Neurogenetics, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
26
|
Koeglsperger T, Rumpf SL, Schließer P, Struebing FL, Brendel M, Levin J, Trenkwalder C, Höglinger GU, Herms J. Neuropathology of incidental Lewy body & prodromal Parkinson's disease. Mol Neurodegener 2023; 18:32. [PMID: 37173733 PMCID: PMC10182593 DOI: 10.1186/s13024-023-00622-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurodegenerative disorder associated with a loss of dopaminergic (DA) neurons. Despite symptomatic therapies, there is currently no disease-modifying treatment to halt neuronal loss in PD. A major hurdle for developing and testing such curative therapies results from the fact that most DA neurons are already lost at the time of the clinical diagnosis, rendering them inaccessible to therapy. Understanding the early pathological changes that precede Lewy body pathology (LBP) and cell loss in PD will likely support the identification of novel diagnostic and therapeutic strategies and help to differentiate LBP-dependent and -independent alterations. Several previous studies identified such specific molecular and cellular changes that occur prior to the appearance of Lewy bodies (LBs) in DA neurons, but a concise map of such early disease events is currently missing. METHODS Here, we conducted a literature review to identify and discuss the results of previous studies that investigated cases with incidental Lewy body disease (iLBD), a presumed pathological precursor of PD. RESULTS Collectively, our review demonstrates numerous cellular and molecular neuropathological changes occurring prior to the appearance of LBs in DA neurons. CONCLUSIONS Our review provides the reader with a summary of early pathological events in PD that may support the identification of novel therapeutic and diagnostic targets and aid to the development of disease-modifying strategies in PD.
Collapse
Affiliation(s)
- Thomas Koeglsperger
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany.
- Department of Translational Brain Research, DZNE-German Center for Neurodegenerative Diseases, 81377, Munich, Germany.
| | - Svenja-Lotta Rumpf
- Department of Translational Brain Research, DZNE-German Center for Neurodegenerative Diseases, 81377, Munich, Germany
| | - Patricia Schließer
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Felix L Struebing
- Department of Translational Brain Research, DZNE-German Center for Neurodegenerative Diseases, 81377, Munich, Germany
- Centre for Neuropathology and Prion Research, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Translational Brain Research, DZNE-German Center for Neurodegenerative Diseases, 81377, Munich, Germany
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
| | - Johannes Levin
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
- Clinical Study Unit, DZNE - German Center for Neurodegenerative Diseases, 81377, Munich, Germany
| | - Claudia Trenkwalder
- Paracelsus-Elena Klinik, Kassel, Germany
- Department of Neurosurgery, University Medical Center Goettingen, Goettingen, Germany
| | - Günter U Höglinger
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Neurology, Medizinische Hochschule Hannover (MHH), Hannover, Germany
| | - Jochen Herms
- Department of Translational Brain Research, DZNE-German Center for Neurodegenerative Diseases, 81377, Munich, Germany
- Centre for Neuropathology and Prion Research, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
| |
Collapse
|
27
|
Yahya V, Di Fonzo A, Monfrini E. Genetic Evidence for Endolysosomal Dysfunction in Parkinson’s Disease: A Critical Overview. Int J Mol Sci 2023; 24:ijms24076338. [PMID: 37047309 PMCID: PMC10094484 DOI: 10.3390/ijms24076338] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/23/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder in the aging population, and no disease-modifying therapy has been approved to date. The pathogenesis of PD has been related to many dysfunctional cellular mechanisms, however, most of its monogenic forms are caused by pathogenic variants in genes involved in endolysosomal function (LRRK2, VPS35, VPS13C, and ATP13A2) and synaptic vesicle trafficking (SNCA, RAB39B, SYNJ1, and DNAJC6). Moreover, an extensive search for PD risk variants revealed strong risk variants in several lysosomal genes (e.g., GBA1, SMPD1, TMEM175, and SCARB2) highlighting the key role of lysosomal dysfunction in PD pathogenesis. Furthermore, large genetic studies revealed that PD status is associated with the overall “lysosomal genetic burden”, namely the cumulative effect of strong and weak risk variants affecting lysosomal genes. In this context, understanding the complex mechanisms of impaired vesicular trafficking and dysfunctional endolysosomes in dopaminergic neurons of PD patients is a fundamental step to identifying precise therapeutic targets and developing effective drugs to modify the neurodegenerative process in PD.
Collapse
Affiliation(s)
- Vidal Yahya
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, 20122 Milan, Italy;
| | - Alessio Di Fonzo
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, 20122 Milan, Italy;
| | - Edoardo Monfrini
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, 20122 Milan, Italy;
- Correspondence:
| |
Collapse
|
28
|
Bogetofte H, Ryan BJ, Jensen P, Schmidt SI, Vergoossen DLE, Barnkob MB, Kiani LN, Chughtai U, Heon-Roberts R, Caiazza MC, McGuinness W, Márquez-Gómez R, Vowles J, Bunn FS, Brandes J, Kilfeather P, Connor JP, Fernandes HJR, Caffrey TM, Meyer M, Cowley SA, Larsen MR, Wade-Martins R. Post-translational proteomics platform identifies neurite outgrowth impairments in Parkinson's disease GBA-N370S dopamine neurons. Cell Rep 2023; 42:112180. [PMID: 36870058 DOI: 10.1016/j.celrep.2023.112180] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/04/2022] [Accepted: 02/13/2023] [Indexed: 03/05/2023] Open
Abstract
Variants at the GBA locus, encoding glucocerebrosidase, are the strongest common genetic risk factor for Parkinson's disease (PD). To understand GBA-related disease mechanisms, we use a multi-part-enrichment proteomics and post-translational modification (PTM) workflow, identifying large numbers of dysregulated proteins and PTMs in heterozygous GBA-N370S PD patient induced pluripotent stem cell (iPSC) dopamine neurons. Alterations in glycosylation status show disturbances in the autophagy-lysosomal pathway, which concur with upstream perturbations in mammalian target of rapamycin (mTOR) activation in GBA-PD neurons. Several native and modified proteins encoded by PD-associated genes are dysregulated in GBA-PD neurons. Integrated pathway analysis reveals impaired neuritogenesis in GBA-PD neurons and identify tau as a key pathway mediator. Functional assays confirm neurite outgrowth deficits and identify impaired mitochondrial movement in GBA-PD neurons. Furthermore, pharmacological rescue of glucocerebrosidase activity in GBA-PD neurons improves the neurite outgrowth deficit. Overall, this study demonstrates the potential of PTMomics to elucidate neurodegeneration-associated pathways and potential drug targets in complex disease models.
Collapse
Affiliation(s)
- Helle Bogetofte
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK; Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark; Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Brent J Ryan
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK.
| | - Pia Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Sissel I Schmidt
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK; Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| | - Dana L E Vergoossen
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Mike B Barnkob
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Department of Clinical Immunology, Odense University Hospital, University of Southern Denmark, 5000 Odense C, Denmark
| | - Lisa N Kiani
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Uroosa Chughtai
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Rachel Heon-Roberts
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Maria Claudia Caiazza
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - William McGuinness
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Ricardo Márquez-Gómez
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Jane Vowles
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Fiona S Bunn
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Janine Brandes
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Peter Kilfeather
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Jack P Connor
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Hugo J R Fernandes
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Tara M Caffrey
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark; Department of Neurology, Odense University Hospital, 5000 Odense C, Denmark
| | - Sally A Cowley
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK.
| |
Collapse
|
29
|
Senkevich K, Rudakou U, Gan-Or Z. Genetic mechanism vs genetic subtypes: The example of GBA. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:155-170. [PMID: 36803808 DOI: 10.1016/b978-0-323-85555-6.00016-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Genetic variants in GBA, encoding the lysosomal enzyme glucocerebrosidase (GCase), are common risk factors for Parkinson's disease (PD). Genotype-phenotype studies have demonstrated that different types of GBA variants have differential effects on the phenotype. Variants could be classified as mild or severe depending on the type of Gaucher disease they cause in the biallelic state. It was shown that severe GBA variants, as compared to mild variants, are associated with higher risk of PD, earlier age at onset, and faster progression of motor and nonmotor symptoms. The observed difference in phenotype might be caused by a diversity of cellular mechanisms related to the particular variants. The lysosomal function of GCase is thought to play a significant role in the development of GBA-associated PD, and other mechanisms such as endoplasmic reticulum retention, mitochondrial dysfunction, and neuroinflammation have also been suggested. Moreover, genetic modifiers such as LRRK2, TMEM175, SNCA, and CTSB can either affect GCase activity or modulate risk and age at onset of GBA-associated PD. To achieve ideal outcomes with precision medicine, therapies will have to be tailored to individuals with specific variants, potentially in combination with known modifiers.
Collapse
Affiliation(s)
- Konstantin Senkevich
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Uladzislau Rudakou
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada; Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Ziv Gan-Or
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada; Department of Human Genetics, McGill University, Montréal, QC, Canada.
| |
Collapse
|
30
|
Smith LJ, Bolsinger MM, Chau KY, Gegg ME, Schapira AHV. The GBA variant E326K is associated with alpha-synuclein aggregation and lipid droplet accumulation in human cell lines. Hum Mol Genet 2023; 32:773-789. [PMID: 36130205 PMCID: PMC9941838 DOI: 10.1093/hmg/ddac233] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/25/2022] [Accepted: 09/09/2022] [Indexed: 11/14/2022] Open
Abstract
Sequence variants or mutations in the GBA gene are numerically the most important risk factor for Parkinson disease (PD). The GBA gene encodes for the lysosomal hydrolase enzyme, glucocerebrosidase (GCase). GBA mutations often reduce GCase activity and lead to the impairment of the autophagy-lysosomal pathway, which is important in the turnover of alpha-synuclein, accumulation of which is a key pathological hallmark of PD. Although the E326K variant is one of the most common GBA variants associated with PD, there is limited understanding of its biochemical effects. We have characterized homozygous and heterozygous E326K variants in human fibroblasts. We found that E326K variants did not cause a significant loss of GCase protein or activity, endoplasmic reticulum (ER) retention or ER stress, in contrast to the L444P GBA mutation. This was confirmed in human dopaminergic SH-SY5Y neuroblastoma cell lines overexpressing GCase with either E326K or L444P protein. Despite no loss of the GCase activity, a significant increase in insoluble alpha-synuclein aggregates in E326K and L444P mutants was observed. Notably, SH-SY5Y overexpressing E326K demonstrated a significant increase in the lipid droplet number under basal conditions, which was exacerbated following treatment with the fatty acid oleic acid. Similarly, a significant increase in lipid droplet formation following lipid loading was observed in heterozygous and homozygous E326K fibroblasts. In conclusion, the work presented here demonstrates that the E326K mutation behaves differently to the common loss of function GBA mutations; however, lipid dyshomeostasis and alpha-synuclein pathology are still evident.
Collapse
Affiliation(s)
- Laura J Smith
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, Royal Free Campus, London NW3 2PF, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Magdalena M Bolsinger
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, Royal Free Campus, London NW3 2PF, UK
- Division of Medicine, Friedrich-Alexander University Erlangen-Nurnberg, Schloßplatz 4, 91054 Erlangen, Germany
| | - Kai-Yin Chau
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, Royal Free Campus, London NW3 2PF, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Matthew E Gegg
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, Royal Free Campus, London NW3 2PF, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, Royal Free Campus, London NW3 2PF, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
31
|
Roecker AJ, Schirripa KM, Loughran HM, Tong L, Liang T, Fillgrove KL, Kuo Y, Bleasby K, Collier H, Altman MD, Ford MC, Drolet RE, Cosden M, Jinn S, Hatcher NG, Yao L, Kandebo M, Vardigan JD, Flick RB, Liu X, Minnick C, Price LA, Watt ML, Lemaire W, Burlein C, Adam GC, Austin LA, Marcus JN, Smith SM, Fraley ME. Pyrazole Ureas as Low Dose, CNS Penetrant Glucosylceramide Synthase Inhibitors for the Treatment of Parkinson's Disease. ACS Med Chem Lett 2023; 14:146-155. [PMID: 36793422 PMCID: PMC9923837 DOI: 10.1021/acsmedchemlett.2c00441] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
Parkinson's disease is the second most prevalent progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra. Loss-of-function mutations in GBA, the gene that encodes for the lysosomal enzyme glucosylcerebrosidase, are a major genetic risk factor for the development of Parkinson's disease potentially through the accumulation of glucosylceramide and glucosylsphingosine in the CNS. A therapeutic strategy to reduce glycosphingolipid accumulation in the CNS would entail inhibition of the enzyme responsible for their synthesis, glucosylceramide synthase (GCS). Herein, we report the optimization of a bicyclic pyrazole amide GCS inhibitor discovered through HTS to low dose, oral, CNS penetrant, bicyclic pyrazole urea GCSi's with in vivo activity in mouse models and ex vivo activity in iPSC neuronal models of synucleinopathy and lysosomal dysfunction. This was accomplished through the judicious use of parallel medicinal chemistry, direct-to-biology screening, physics-based rationalization of transporter profiles, pharmacophore modeling, and use a novel metric: volume ligand efficiency.
Collapse
Affiliation(s)
- Anthony J. Roecker
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Kathy M. Schirripa
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - H. Marie Loughran
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Ling Tong
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Tao Liang
- Discovery
Process Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Kerry L. Fillgrove
- ADME
& Discovery Toxicology, Merck &
Co., Inc., West Point, Pennsylvania 19486, United States
| | - Yuhsin Kuo
- ADME
& Discovery Toxicology, Merck &
Co., Inc., West Point, Pennsylvania 19486, United States
| | - Kelly Bleasby
- ADME
Transporters, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Hannah Collier
- ADME
Transporters, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Michael D. Altman
- Computational
and Structural Chemistry, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Melissa C. Ford
- Computational
and Structural Chemistry, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - Robert E. Drolet
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Mali Cosden
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Sarah Jinn
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Nathan G. Hatcher
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Lihang Yao
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Monika Kandebo
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Joshua D. Vardigan
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Rosemarie B. Flick
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Xiaomei Liu
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Christina Minnick
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Laura A. Price
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Marla L. Watt
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Wei Lemaire
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Christine Burlein
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Gregory C. Adam
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Lauren A. Austin
- Discovery
Pharmaceutical Sciences, Merck & Co.,
Inc., West Point, Pennsylvania 19486, United States
| | - Jacob N. Marcus
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Sean M. Smith
- Discovery
Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Mark E. Fraley
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
32
|
Mahoney-Crane CL, Viswanathan M, Russell D, Curtiss RAC, Freire J, Bobba SS, Coyle SD, Kandebo M, Yao L, Wan BL, Hatcher NG, Smith SM, Marcus JN, Volpicelli-Daley LA. Neuronopathic GBA1L444P Mutation Accelerates Glucosylsphingosine Levels and Formation of Hippocampal Alpha-Synuclein Inclusions. J Neurosci 2023; 43:501-521. [PMID: 36639889 PMCID: PMC9864632 DOI: 10.1523/jneurosci.0680-22.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 09/09/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
The most common genetic risk factor for Parkinson's disease (PD) is heterozygous mutations GBA1, which encodes for the lysosomal enzyme, glucocerebrosidase. Reduced glucocerebrosidase activity associates with an accumulation of abnormal α-synuclein (α-syn) called Lewy pathology, which characterizes PD. PD patients heterozygous for the neuronotypic GBA1L444P mutation (GBA1+/L444P) have a 5.6-fold increased risk of cognitive impairments. In this study, we used GBA1+/L444P mice of either sex to determine its effects on lipid metabolism, expression of synaptic proteins, behavior, and α-syn inclusion formation. At 3 months of age, GBA1+/L444P mice demonstrated impaired contextual fear conditioning, and increased motor activity. Hippocampal levels of vGLUT1 were selectively reduced in GBA1+/L444P mice. We show, using mass spectrometry, that GBA1L444P expression increased levels of glucosylsphingosine, but not glucosylceramide, in the brains and serum of GBA1+/L444P mice. Templated induction of α-syn pathology in mice showed an increase in α-syn inclusion formation in the hippocampus of GBA1+/L444P mice compared with GBA1+/+ mice, but not in the cortex, or substantia nigra pars compacta. Pathologic α-syn reduced SNc dopamine neurons by 50% in both GBA1+/+ and GBA1+/L444P mice. Treatment with a GlcCer synthase inhibitor did not affect abundance of α-syn inclusions in the hippocampus or rescue dopamine neuron loss. Overall, these data suggest the importance of evaluating the contribution of elevated glucosylsphingosine to PD phenotypes. Further, our data suggest that expression of neuronotypic GBA1L444P may cause defects in the hippocampus, which may be a mechanism by which cognitive decline is more prevalent in individuals with GBA1-PD.SIGNIFICANCE STATEMENT Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are both pathologically characterized by abnormal α-synuclein (α-syn). Mutant GBA1 is a risk factor for both PD and DLB. Our data show the expression of neuronotypic GBA1L444P impairs behaviors related to hippocampal function, reduces expression of a hippocampal excitatory synaptic protein, and that the hippocampus is more susceptible to α-syn inclusion formation. Further, our data strengthen support for the importance of evaluating the contribution of glucosylsphingosine to PD phenotypes. These outcomes suggest potential mechanisms by which GBA1L444P contributes to the cognitive symptoms clinically observed in PD and DLB. Our findings also highlight the importance of glucosylsphingosine as a relevant biomarker for future therapeutics.
Collapse
Affiliation(s)
- Casey L Mahoney-Crane
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Megha Viswanathan
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Dreson Russell
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Rachel A C Curtiss
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Jennifer Freire
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Sai Sumedha Bobba
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Sean D Coyle
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Monika Kandebo
- Neuroscience Discovery, Merck & Company, Inc, West Point, Pennsylvania 19486
| | - Lihang Yao
- Neuroscience Discovery, Merck & Company, Inc, West Point, Pennsylvania 19486
| | - Bang-Lin Wan
- Neuroscience Discovery, Merck & Company, Inc, West Point, Pennsylvania 19486
| | - Nathan G Hatcher
- Neuroscience Discovery, Merck & Company, Inc, West Point, Pennsylvania 19486
| | - Sean M Smith
- Neuroscience Discovery, Merck & Company, Inc, West Point, Pennsylvania 19486
| | - Jacob N Marcus
- Neuroscience Discovery, Merck & Company, Inc, West Point, Pennsylvania 19486
| | - Laura A Volpicelli-Daley
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
33
|
Sex-Specific Microglial Responses to Glucocerebrosidase Inhibition: Relevance to GBA1-Linked Parkinson's Disease. Cells 2023; 12:cells12030343. [PMID: 36766684 PMCID: PMC9913749 DOI: 10.3390/cells12030343] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Microglia are heterogenous cells characterized by distinct populations each contributing to specific biological processes in the nervous system, including neuroprotection. To elucidate the impact of sex-specific microglia heterogenicity to the susceptibility of neuronal stress, we video-recorded with time-lapse microscopy the changes in shape and motility occurring in primary cells derived from mice of both sexes in response to pro-inflammatory or neurotoxic stimulations. With this morpho-functional analysis, we documented distinct microglia subpopulations eliciting sex-specific responses to stimulation: male microglia tended to have a more pro-inflammatory phenotype, while female microglia showed increased sensitivity to conduritol-B-epoxide (CBE), a small molecule inhibitor of glucocerebrosidase, the enzyme encoded by the GBA1 gene, mutations of which are the major risk factor for Parkinson's Disease (PD). Interestingly, glucocerebrosidase inhibition particularly impaired the ability of female microglia to enhance the Nrf2-dependent detoxification pathway in neurons, attenuating the sex differences observed in this neuroprotective function. This finding is consistent with the clinical impact of GBA1 mutations, in which the 1.5-2-fold reduced risk of developing idiopathic PD observed in female individuals is lost in the GBA1 carrier population, thus suggesting a sex-specific role for microglia in the etiopathogenesis of PD-GBA1.
Collapse
|
34
|
Mechanisms of Autoimmune Cell in DA Neuron Apoptosis of Parkinson's Disease: Recent Advancement. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7965433. [PMID: 36567855 PMCID: PMC9771667 DOI: 10.1155/2022/7965433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder that manifests as motor and nonmotor symptoms due to the selective loss of midbrain DArgic (DA) neurons. More and more studies have shown that pathological reactions initiated by autoimmune cells play an essential role in the progression of PD. Autoimmune cells exist in the brain parenchyma, cerebrospinal fluid, and meninges; they are considered inducers of neuroinflammation and regulate the immune in the human brain in PD. For example, T cells can recognize α-synuclein presented by antigen-presenting cells to promote neuroinflammation. In addition, B cells will accelerate the apoptosis of DA neurons in the case of PD-related gene mutations. Activation of microglia and damage of DA neurons even form the self-degeneration cycle to deteriorate PD. Numerous autoimmune cells have been considered regulators of apoptosis, α-synuclein misfolding and aggregation, mitochondrial dysfunction, autophagy, and neuroinflammation of DA neurons in PD. The evidence is mounting that autoimmune cells promote DA neuron apoptosis. In this review, we discuss the current knowledge regarding the regulation and function of B cell, T cell, and microglia as well as NK cell in PD pathogenesis, focusing on DA neuron apoptosis to understand the disease better and propose potential target identification for the treatment in the early stages of PD. However, there are still some limitations in our work, for example, the specific mechanism of PD progression caused by autoimmune cells in mitochondrial dysfunction, ferroptosis, and autophagy has not been clarified in detail, which needs to be summarized in further work.
Collapse
|
35
|
Vallés AS, Barrantes FJ. The synaptic lipidome in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:184033. [PMID: 35964712 DOI: 10.1016/j.bbamem.2022.184033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 06/15/2023]
Abstract
Adequate homeostasis of lipid, protein and carbohydrate metabolism is essential for cells to perform highly specific tasks in our organism, and the brain, with its uniquely high energetic requirements, posesses singular characteristics. Some of these are related to its extraordinary dotation of synapses, the specialized subcelluar structures where signal transmission between neurons occurs in the central nervous system. The post-synaptic compartment of excitatory synapses, the dendritic spine, harbors key molecules involved in neurotransmission tightly packed within a minute volume of a few femtoliters. The spine is further compartmentalized into nanodomains that facilitate the execution of temporo-spatially separate functions in the synapse. Lipids play important roles in this structural and functional compartmentalization and in mechanisms that impact on synaptic transmission. This review analyzes the structural and dynamic processes involving lipids at the synapse, highlighting the importance of their homeostatic balance for the physiology of this complex and highly specialized structure, and underscoring the pathologies associated with disbalances of lipid metabolism, particularly in the perinatal and late adulthood periods of life. Although small variations of the lipid profile in the brain take place throughout the adult lifespan, the pathophysiological consequences are clinically manifested mostly during late adulthood. Disturbances in lipid homeostasis in the perinatal period leads to alterations during nervous system development, while in late adulthood they favor the occurrence of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ana Sofia Vallés
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (UNS-CONICET), 8000 Bahía Blanca, Argentina.
| | - Francisco J Barrantes
- Laboratory of Molecular Neurobiology, Institute of Biomedical Research (BIOMED), UCA-CONICET, Av. Alicia Moreau de Justo 1600, Buenos Aires C1107AAZ, Argentina.
| |
Collapse
|
36
|
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
|
37
|
Imbriani P, Martella G, Bonsi P, Pisani A. Oxidative stress and synaptic dysfunction in rodent models of Parkinson's disease. Neurobiol Dis 2022; 173:105851. [PMID: 36007757 DOI: 10.1016/j.nbd.2022.105851] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 08/02/2022] [Accepted: 08/20/2022] [Indexed: 11/26/2022] Open
Abstract
Parkinson's disease (PD) is a multifactorial disorder involving a complex interplay between a variety of genetic and environmental factors. In this scenario, mitochondrial impairment and oxidative stress are widely accepted as crucial neuropathogenic mechanisms, as also evidenced by the identification of PD-associated genes that are directly involved in mitochondrial function. The concept of mitochondrial dysfunction is closely linked to that of synaptic dysfunction. Indeed, compelling evidence supports the role of mitochondria in synaptic transmission and plasticity, although many aspects have not yet been fully elucidated. Here, we will provide a brief overview of the most relevant evidence obtained in different neurotoxin-based and genetic rodent models of PD, focusing on mitochondrial impairment and synaptopathy, an early central event preceding overt nigrostriatal neurodegeneration. The identification of early deficits occurring in PD pathogenesis is crucial in view of the development of potential disease-modifying therapeutic strategies.
Collapse
Affiliation(s)
- Paola Imbriani
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; IRCCS Mondino Foundation, Pavia, Italy.
| |
Collapse
|
38
|
Arévalo NB, Lamaizon CM, Cavieres VA, Burgos PV, Álvarez AR, Yañez MJ, Zanlungo S. Neuronopathic Gaucher disease: Beyond lysosomal dysfunction. Front Mol Neurosci 2022; 15:934820. [PMID: 35992201 PMCID: PMC9381931 DOI: 10.3389/fnmol.2022.934820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022] Open
Abstract
Gaucher disease (GD) is an inherited disorder caused by recessive mutations in the GBA1 gene that encodes the lysosomal enzyme β-glucocerebrosidase (β-GC). β-GC hydrolyzes glucosylceramide (GluCer) into glucose and ceramide in the lysosome, and the loss of its activity leads to GluCer accumulation in different tissues. In severe cases, enzymatic deficiency triggers inflammation, organomegaly, bone disease, and neurodegeneration. Neuronopathic Gaucher disease (nGD) encompasses two different forms of the disease, characterized by chronic or acute damage to the central nervous system (CNS). The cellular and molecular studies that uncover the pathological mechanisms of nGD mainly focus on lysosomal dysfunction since the lysosome is the key organelle affected in GD. However, new studies show alterations in other organelles that contribute to nGD pathology. For instance, abnormal accumulation of GluCer in lysosomes due to the loss of β-GC activity leads to excessive calcium release from the endoplasmic reticulum (ER), activating the ER-associated degradation pathway and the unfolded protein response. Recent evidence indicates mitophagy is altered in nGD, resulting in the accumulation of dysfunctional mitochondria, a critical factor in disease progression. Additionally, nGD patients present alterations in mitochondrial morphology, membrane potential, ATP production, and increased reactive oxygen species (ROS) levels. Little is known about potential dysfunction in other organelles of the secretory pathway, such as the Golgi apparatus and exosomes. This review focuses on collecting evidence regarding organelle dysfunction beyond lysosomes in nGD. We briefly describe cellular and animal models and signaling pathways relevant to uncovering the pathological mechanisms and new therapeutic targets in GD.
Collapse
Affiliation(s)
- Nohela B. Arévalo
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Cell and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica, Santiago, Chile
| | - Cristian M. Lamaizon
- Department of Cell and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica, Santiago, Chile
| | - Viviana A. Cavieres
- Facultad de Medicina y Ciencia, Centro de Biología Celular y Biomedicina (CEBICEM), Universidad San Sebastián, Santiago, Chile
- Facultad de Ciencias Biológicas, Centro de Envejecimiento y Regeneración (CARE-UC), Pontificia Universidad Católica, Santiago, Chile
| | - Patricia V. Burgos
- Facultad de Medicina y Ciencia, Centro de Biología Celular y Biomedicina (CEBICEM), Universidad San Sebastián, Santiago, Chile
- Facultad de Ciencias Biológicas, Centro de Envejecimiento y Regeneración (CARE-UC), Pontificia Universidad Católica, Santiago, Chile
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago, Chile
| | - Alejandra R. Álvarez
- Department of Cell and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica, Santiago, Chile
- Facultad de Ciencias Biológicas, Centro de Envejecimiento y Regeneración (CARE-UC), Pontificia Universidad Católica, Santiago, Chile
| | - María J. Yañez
- Faculty of Medicine and Science, School of Medical Technology, Universidad San Sebastian, Concepción, Chile
- *Correspondence: María J. Yañez
| | - Silvana Zanlungo
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Silvana Zanlungo
| |
Collapse
|
39
|
Zhang F, Wu Z, Long F, Tan J, Gong N, Li X, Lin C. The Roles of ATP13A2 Gene Mutations Leading to Abnormal Aggregation of α-Synuclein in Parkinson’s Disease. Front Cell Neurosci 2022; 16:927682. [PMID: 35875356 PMCID: PMC9296842 DOI: 10.3389/fncel.2022.927682] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease. PARK9 (also known as ATP13A2) is recognized as one of the key genes that cause PD, and a mutation in this gene was first discovered in a rare case of PD in an adolescent. Lewy bodies (LBs) formed by abnormal aggregation of α-synuclein, which is encoded by the SNCA gene, are one of the pathological diagnostic criteria for PD. LBs are also recognized as one of the most important features of PD pathogenesis. In this article, we first summarize the types of mutations in the ATP13A2 gene and their effects on ATP13A2 mRNA and protein structure; then, we discuss lysosomal autophagy inhibition and the molecular mechanism of abnormal α-synuclein accumulation caused by decreased levels and dysfunction of the ATP13A2 protein in lysosomes. Finally, this article provides a new direction for future research on the pathogenesis and therapeutic targets for ATP13A2 gene-related PD from the perspective of ATP13A2 gene mutations and abnormal aggregation of α-synuclein.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhiwei Wu
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Fei Long
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jieqiong Tan
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Key Laboratory of Molecular Precision Medicine of Hunan Province, Center for Medical Genetics, Institute of Molecular Precision Medicine, Xiangya Hospital of Central South University, Changsha, China
| | - Ni Gong
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiaorong Li
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Changwei Lin
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- *Correspondence: Changwei Lin, orcid.org/0000-0003-1676-0912
| |
Collapse
|
40
|
Lysosomal functions and dysfunctions: Molecular and cellular mechanisms underlying Gaucher disease and its association with Parkinson disease. Adv Drug Deliv Rev 2022; 187:114402. [DOI: 10.1016/j.addr.2022.114402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 04/28/2022] [Accepted: 06/17/2022] [Indexed: 01/18/2023]
|
41
|
Polinski NK, Martinez TN, Ramboz S, Sasner M, Herberth M, Switzer R, Ahmad SO, Pelligrino LJ, Clark SW, Marcus JN, Smith SM, Dave KD, Frasier MA. The GBA1 D409V mutation exacerbates synuclein pathology to differing extents in two alpha-synuclein models. Dis Model Mech 2022; 15:dmm049192. [PMID: 35419585 PMCID: PMC9150115 DOI: 10.1242/dmm.049192] [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: 07/20/2021] [Accepted: 04/11/2022] [Indexed: 11/28/2022] Open
Abstract
Heterozygous mutations in the GBA1 gene - encoding lysosomal glucocerebrosidase (GCase) - are the most common genetic risk factors for Parkinson's disease (PD). Experimental evidence suggests a correlation between decreased GCase activity and accumulation of alpha-synuclein (aSyn). To enable a better understanding of the relationship between aSyn and GCase activity, we developed and characterized two mouse models that investigate aSyn pathology in the context of reduced GCase activity. The first model used constitutive overexpression of wild-type human aSyn in the context of the homozygous GCase activity-reducing D409V mutant form of GBA1. Although increased aSyn pathology and grip strength reductions were observed in this model, the nigrostriatal system remained largely intact. The second model involved injection of aSyn preformed fibrils (PFFs) into the striatum of the homozygous GBA1 D409V knock-in mouse model. The GBA1 D409V mutation did not exacerbate the pathology induced by aSyn PFF injection. This study sheds light on the relationship between aSyn and GCase in mouse models, highlighting the impact of model design on the ability to model a relationship between these proteins in PD-related pathology.
Collapse
Affiliation(s)
- Nicole K. Polinski
- The Michael J. Fox Foundation for Parkinson's Research, Grand Central Station PO Box 4777, New York, NY 10163, USA
| | - Terina N. Martinez
- The Michael J. Fox Foundation for Parkinson's Research, Grand Central Station PO Box 4777, New York, NY 10163, USA
| | - Sylvie Ramboz
- PsychoGenics, Inc, 215 College Road, Paramus, NJ 07652, USA
| | - Michael Sasner
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Mark Herberth
- Charles River Laboratories, 1407 George Road, Ashland, OH 44805, USA
| | - Robert Switzer
- NeuroScience Associates, 10915 Lake Ridge Drive, Knoxville, TN 37934, USA
| | - Syed O. Ahmad
- Saint Louis University, 3437 Caroline Street, St. Louis, MO 63104, USA
| | | | - Sean W. Clark
- Amicus Therapeutics, 1 Cedarbrook Dr, Cranbury, NJ 08512, USA
| | - Jacob N. Marcus
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Sean M. Smith
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Kuldip D. Dave
- The Michael J. Fox Foundation for Parkinson's Research, Grand Central Station PO Box 4777, New York, NY 10163, USA
| | - Mark A. Frasier
- The Michael J. Fox Foundation for Parkinson's Research, Grand Central Station PO Box 4777, New York, NY 10163, USA
| |
Collapse
|
42
|
Ghatti S, Yoon E, Lopez G, Ehrlich D, Horovitz SG. Imaging and genetics in Parkinson's disease: assessment of the GBA1 mutation. J Neurol 2022; 269:5347-5355. [PMID: 35604467 PMCID: PMC10402751 DOI: 10.1007/s00415-022-11181-0] [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: 11/23/2021] [Revised: 04/21/2022] [Accepted: 05/10/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Several genetic variants are associated with an increased risk for developing Parkinson's Disease (PD) and limited genotype/phenotype correlation. Specifically, mutations in GBA1, the gene coding for the lysosomal enzyme glucocerebrosidase, are associated with an earlier age of onset and faster disease progression. Given these phenotypic differences associated with GBA1 variants, we explored whether cortical thickness and other biomarkers of neurodegeneration differed in healthy controls and PD patients with and without GBA1 variants. METHODS To understand how different GBA1 variants influence PD phenotype early in the disease, we retrieved neuroimaging and biospecimen data from the Parkinson's Progression Markers Initiative database. Using FreeSurfer, we compared T1-weighted MRI images from healthy controls (N = 47) to PD patients with heterozygous N370S (N = 21), heterozygous E326K (N = 18) or heterozygous T369M (N = 8) variants, and GBA1 non-mutation carriers (N = 47). RESULTS Cortical thickness in PD patients differed from controls in the parietal cortex, with E365K, T369M variants, and GBA1 non-mutation carriers showing more cortical thinning than N370S variants. Patients with N370S variants had significantly higher serum neurofilament light levels among all groups. CONCLUSION Our results demonstrate significant cortical thinning in PD patients independent of genotype in superior parietal and postcentral regions when compared to the controls. They highlight the impact of GBA1 variants on cortical thickness in the parietal cortex. Finally, they suggest that recently diagnosed PD patients with N370S variants have a higher cortical thickness and increased active neurodegeneration when compared to PD patients without GBA1 mutations and PD patients with E326K or T369M variants.
Collapse
Affiliation(s)
- Sweta Ghatti
- National Institutes of Neurological Disease and Stroke, Bethesda, MD, USA.
| | - Esther Yoon
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Grisel Lopez
- National Human Genome Research Institutes, Bethesda, MD, USA
| | - Debra Ehrlich
- National Institutes of Neurological Disease and Stroke, Bethesda, MD, USA
| | - Silvina G Horovitz
- National Institutes of Neurological Disease and Stroke, Bethesda, MD, USA
| |
Collapse
|
43
|
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
|
44
|
GBA Variants and Parkinson Disease: Mechanisms and Treatments. Cells 2022; 11:cells11081261. [PMID: 35455941 PMCID: PMC9029385 DOI: 10.3390/cells11081261] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/01/2022] [Accepted: 04/05/2022] [Indexed: 01/01/2023] Open
Abstract
The GBA gene encodes for the lysosomal enzyme glucocerebrosidase (GCase), which maintains glycosphingolipid homeostasis. Approximately 5–15% of PD patients have mutations in the GBA gene, making it numerically the most important genetic risk factor for Parkinson disease (PD). Clinically, GBA-associated PD is identical to sporadic PD, aside from the earlier age at onset (AAO), more frequent cognitive impairment and more rapid progression. Mutations in GBA can be associated with loss- and gain-of-function mechanisms. A key hallmark of PD is the presence of intraneuronal proteinaceous inclusions named Lewy bodies, which are made up primarily of alpha-synuclein. Mutations in the GBA gene may lead to loss of GCase activity and lysosomal dysfunction, which may impair alpha-synuclein metabolism. Models of GCase deficiency demonstrate dysfunction of the autophagic-lysosomal pathway and subsequent accumulation of alpha-synuclein. This dysfunction can also lead to aberrant lipid metabolism, including the accumulation of glycosphingolipids, glucosylceramide and glucosylsphingosine. Certain mutations cause GCase to be misfolded and retained in the endoplasmic reticulum (ER), activating stress responses including the unfolded protein response (UPR), which may contribute to neurodegeneration. In addition to these mechanisms, a GCase deficiency has also been associated with mitochondrial dysfunction and neuroinflammation, which have been implicated in the pathogenesis of PD. This review discusses the pathways associated with GBA-PD and highlights potential treatments which may act to target GCase and prevent neurodegeneration.
Collapse
|
45
|
Li S, Kim HE. Implications of Sphingolipids on Aging and Age-Related Diseases. FRONTIERS IN AGING 2022; 2:797320. [PMID: 35822041 PMCID: PMC9261390 DOI: 10.3389/fragi.2021.797320] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/31/2021] [Indexed: 01/14/2023]
Abstract
Aging is a process leading to a progressive loss of physiological integrity and homeostasis, and a primary risk factor for many late-onset chronic diseases. The mechanisms underlying aging have long piqued the curiosity of scientists. However, the idea that aging is a biological process susceptible to genetic manipulation was not well established until the discovery that the inhibition of insulin/IGF-1 signaling extended the lifespan of C. elegans. Although aging is a complex multisystem process, López-Otín et al. described aging in reference to nine hallmarks of aging. These nine hallmarks include: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. Due to recent advances in lipidomic, investigation into the role of lipids in biological aging has intensified, particularly the role of sphingolipids (SL). SLs are a diverse group of lipids originating from the Endoplasmic Reticulum (ER) and can be modified to create a vastly diverse group of bioactive metabolites that regulate almost every major cellular process, including cell cycle regulation, senescence, proliferation, and apoptosis. Although SL biology reaches all nine hallmarks of aging, its contribution to each hallmark is disproportionate. In this review, we will discuss in detail the major contributions of SLs to the hallmarks of aging and age-related diseases while also summarizing the importance of their other minor but integral contributions.
Collapse
Affiliation(s)
- Shengxin Li
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, TX, United States
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hyun-Eui Kim
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, TX, United States
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
46
|
Bo RX, Li YY, Zhou TT, Chen NH, Yuan YH. The neuroinflammatory role of glucocerebrosidase in Parkinson's disease. Neuropharmacology 2022; 207:108964. [PMID: 35065083 DOI: 10.1016/j.neuropharm.2022.108964] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/10/2022] [Accepted: 01/15/2022] [Indexed: 10/19/2022]
Abstract
The lysosomal enzyme glucocerebrosidase (GCase), encoded by the GBA1 gene, is a membrane-associated protein catalyzing the cleavage of glucosylceramide (GlcCer) and glucosylsphingosine (GlcSph). Homologous GBA1 mutations cause Gaucher disease (GD) and heterologous mutations cause Parkinson's disease (PD). Importantly, heterologous GBA1 mutations are recognized as the second risk factor of PD. The pathological features of PD are Lewy neurites (LNs) and Lewy bodies (LBs) composed of pathological α-synuclein. Oxidative stress, inflammatory response, autophagic impairment, and α-synuclein accumulation play critical roles in PD pathogenic cascades, but the pathogenesis of PD has not yet been fully elucidated. What's more, PD treatment drugs can only relieve symptoms to a certain extent, but cannot alleviate neurodegenerative progression. Therefore, it's urgent to explore new targets that can alleviate the neurodegenerative process. Deficient GCase can cause lysosomal dysfunction, obstructing the metabolism of α-synuclein. Meanwhile, GCase dysfunction causes accumulation of its substrates, leading to lipid metabolism disorders. Subsequently, astrocytes and microglia are activated, releasing amounts of pro-inflammatory mediators and causing extensive neuroinflammation. All these cascades can induce neuron damage and death, eventually promoting PD pathology. This review aims to summarize these points and the potential of GCase as an original target to provide some ideas for elucidating the pathogenesis of PD.
Collapse
Affiliation(s)
- Ru-Xue Bo
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Yan-Yan Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Tian-Tian Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Yu-He Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
47
|
Abeliovich A, Hefti F, Sevigny J. Gene Therapy for Parkinson's Disease Associated with GBA1 Mutations. JOURNAL OF PARKINSON'S DISEASE 2022; 11:S183-S188. [PMID: 34151863 PMCID: PMC8543272 DOI: 10.3233/jpd-212739] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 11/24/2022]
Abstract
Human genetic studies as well as studies in animal models indicate that lysosomal dysfunction plays a key role in the pathogenesis of Parkinson's disease. Among the lysosomal genes involved, GBA1 has the largest impact on Parkinson's disease risk. Deficiency in the GBA1 encoded enzyme glucocerebrosidase (GCase) leads to the accumulation of the GCase glycolipid substrates glucosylceramide and glucosylsphingosine and ultimately results in toxicity and inflammation and negatively affect many clinical aspects of Parkinson's disease, including disease risk, the severity of presentation, age of onset, and likelihood of progression to dementia. These findings support the view that re-establishing normal levels of GCase enzyme activity may reduce the progression of Parkinson's disease in patients carrying GBA1 mutations. Studies in mouse models indicate that PR001, a AAV9 vector-based gene therapy designed to deliver a functional GBA1 gene to the brain, suggest that this therapeutic approach may slow or stop disease progression. PR001 is currently being evaluated in clinical trials with Parkinson's disease patients carrying GBA1 mutations.
Collapse
Affiliation(s)
- Asa Abeliovich
- Prevail Therapeutics, A Wholly-Owned Subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Franz Hefti
- Prevail Therapeutics, A Wholly-Owned Subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Jeffrey Sevigny
- Prevail Therapeutics, A Wholly-Owned Subsidiary of Eli Lilly and Company, New York, NY, USA
| |
Collapse
|
48
|
Al-Azzawi ZAM, Arfaie S, Gan-Or Z. GBA1 and The Immune System: A Potential Role in Parkinson's Disease? JOURNAL OF PARKINSON'S DISEASE 2022; 12:S53-S64. [PMID: 36057834 PMCID: PMC9535551 DOI: 10.3233/jpd-223423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
It is clear that the immune system and inflammation have a role in Parkinson's disease (PD), including sporadic PD and some genetic forms such as LRRK2-associated PD. One of the most important genes associated with PD is GBA1, as variants in this gene are found in 5-20% of PD patients in different populations worldwide. Biallelic variants in GBA1 may cause Gaucher disease, a lysosomal storage disorder with involvement of the immune system, and other lines of evidence link GBA1 to the immune system and inflammation. In this review, we discuss these different pieces of evidence and whether the interplay between GBA1 and the immune system may have a role in PD.
Collapse
Affiliation(s)
- Zaid A M Al-Azzawi
- Faculty of Medicine, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Saman Arfaie
- Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Ziv Gan-Or
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- The Neuro - Montreal Neurological Institute-Hospital, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| |
Collapse
|
49
|
Francelle L, Mazzulli JR. Neuroinflammation in aucher disease, neuronal ceroid lipofuscinosis, and commonalities with Parkinson’s disease. Brain Res 2022; 1780:147798. [PMID: 35063468 PMCID: PMC9126024 DOI: 10.1016/j.brainres.2022.147798] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 01/05/2022] [Accepted: 01/13/2022] [Indexed: 12/13/2022]
Abstract
Lysosomal storage diseases (LSDs) are rare genetic disorders caused by a disruption in cellular clearance, resulting in pathological storage of undegraded lysosomal substrates. Recent clinical and genetic studies have uncovered links between multiple LSDs and common neurodegenerative diseases such as Parkinson's disease (PD). Here, we review recent literature describing the role of glia cells and neuroinflammation in PD and LSDs, including Gaucher disease (GD) and neuronal ceroid lipofuscinosis (NCL), and highlight converging inflammation pathways that lead to neuron loss. Recent data indicates that lysosomal dysfunction and accumulation of storage materials can initiate the activation of glial cells, through interaction with cell surface or cytosolic pattern recognition receptors that detect pathogenic aggregates of cellular debris. Activated glia cells could act to protect neurons through the elimination of toxic protein or lipid aggregates early in the disease process. However prolonged glial activation that occurs over several decades in chronic-age related neurodegeneration could induce the inappropriate elimination of synapses, leading to neuron loss. These studies provide mechanistic insight into the relationship between lysosomal dysfunction and glial activation, and offer novel therapeutic pathways for the treatment of PD and LSDs focused on reducing neuroinflammation and mitigating cell loss.
Collapse
|
50
|
Petese A, Cesaroni V, Cerri S, Blandini F. Are Lysosomes Potential Therapeutic Targets for Parkinson's Disease? CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 21:642-655. [PMID: 34370650 DOI: 10.2174/1871527320666210809123630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/16/2021] [Accepted: 06/10/2021] [Indexed: 06/13/2023]
Abstract
Parkinson´s Disease (PD) is the second most common neurodegenerative disorder, affecting ~2-3% of the population over 65 years old. In addition to progressive degeneration of nigrostriatal neurons, the histopathological feature of PD is the accumulation of misfolded α-synuclein protein in abnormal cytoplasmatic inclusions, known as Lewy Bodies (LBs). Recently, Genome-Wide Association Studies (GWAS) have indicated a clear association of variants within several lysosomal genes with risk for PD. Newly evolving data have been shedding light on the relationship between lysosomal dysfunction and alpha-synuclein aggregation. Defects in lysosomal enzymes could lead to the insufficient clearance of neurotoxic protein materials, possibly leading to selective degeneration of dopaminergic neurons. Specific modulation of lysosomal pathways and their components could be considered a novel opportunity for therapeutic intervention for PD. The purpose of this review is to illustrate lysosomal biology and describe the role of lysosomal dysfunction in PD pathogenesis. Finally, the most promising novel therapeutic approaches designed to modulate lysosomal activity, as a potential disease-modifying treatment for PD will be highlighted.
Collapse
Affiliation(s)
- Alessandro Petese
- Cellular and Molecular Neurobiology Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Valentina Cesaroni
- Cellular and Molecular Neurobiology Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Silvia Cerri
- Cellular and Molecular Neurobiology Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Fabio Blandini
- Cellular and Molecular Neurobiology Unit, IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|