1
|
Gaweda-Walerych K, Aragona V, Lodato S, Sitek EJ, Narożańska E, Buratti E. Progranulin deficiency in the brain: the interplay between neuronal and non-neuronal cells. Transl Neurodegener 2025; 14:18. [PMID: 40234992 PMCID: PMC12001433 DOI: 10.1186/s40035-025-00475-8] [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: 09/20/2024] [Accepted: 02/21/2025] [Indexed: 04/17/2025] Open
Abstract
Heterozygous mutations in GRN gene lead to insufficient levels of the progranulin (PGRN) protein, resulting in frontotemporal dementia (FTD) with TAR DNA-binding protein 43 (TDP-43) inclusions, classified pathologically as frontotemporal lobar degeneration (FTLD-TDP). Homozygous GRN mutations are exceedingly rare and cause neuronal ceroid lipofuscinosis 11, a lysosomal storage disease with onset in young adulthood, or an FTD syndrome with late-onset manifestations. In this review, we highlight the broad spectrum of clinical phenotypes associated with PGRN deficiency, including primary progressive aphasia and behavioral variant of frontotemporal dementia. We explore these phenotypes alongside relevant rodent and in vitro human models, ranging from the induced pluripotent stem cell-derived neural progenitors, neurons, microglia, and astrocytes to genetically engineered heterotypic organoids containing both neurons and astrocytes. We summarize advantages and limitations of these models in recapitulating the main FTLD-GRN hallmarks, highlighting the role of non-cell-autonomous mechanisms in the formation of TDP-43 pathology, neuroinflammation, and neurodegeneration. Data obtained from patients' brain tissues and biofluids, in parallel with single-cell transcriptomics, demonstrate the complexity of interactions among the highly heterogeneous cellular clusters present in the brain, including neurons, astrocytes, microglia, oligodendroglia, endothelial cells, and pericytes. Emerging evidence has revealed that PGRN deficiency is associated with cell cluster-specific, often conserved, genetic and molecular phenotypes in the central nervous system. In this review, we focus on how these distinct cellular populations and their dysfunctional crosstalk contribute to neurodegeneration and neuroinflammation in FTD-GRN. Specifically, we characterize the phenotypes of lipid droplet-accumulating microglia and alterations of myelin lipid content resulting from lysosomal dysfunction caused by PGRN deficiency. Additionally, we consider how the deregulation of glia-neuron communication affects the exchange of organelles such as mitochondria, and the removal of excess toxic products such as protein aggregates, in PGRN-related neurodegeneration.
Collapse
Affiliation(s)
- Katarzyna Gaweda-Walerych
- Department of Neurogenetics and Functional Genomics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland.
| | - Vanessa Aragona
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Emilia J Sitek
- Division of Neurological and Psychiatric Nursing, Laboratory of Clinical Neuropsychology, Neurolinguistics, and Neuropsychotherapy, Faculty of Health Sciences, Medical University of Gdansk, 80-210, Gdansk, Poland.
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland.
| | - Ewa Narożańska
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland
| | - Emanuele Buratti
- Molecular Pathology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, 34149, Trieste, Italy
| |
Collapse
|
2
|
Xia M, Gao C, Shang J, Li D, Yang A, Zang W, Zhang J. A novel c.1468 G > A GRN mutation causes frontotemporal dementia in a Chinese Han family. Eur J Med Res 2025; 30:157. [PMID: 40055834 PMCID: PMC11889931 DOI: 10.1186/s40001-025-02418-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 02/27/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND/PURPOSE GRN mutations act as causative factors in patients with FTD clinical phenotype or FTD pathology and exhibit high clinical heterogeneity. The discovery of these mutations and the analysis of their associations with resembling Alzheimer's disease should be critical to understand the pathogenesis of FTD. METHODS Clinical analysis, neuroimaging, target region capture and high-throughput sequencing were performed in a family of 3 generations. The underlying Alzheimer's pathology was evaluated by using biomarker evidence obtained from cerebrospinal fluid (CSF) amyloid testing, 18F-florbetapir (AV-45) PET imaging and FDG18-positron emission tomography imaging. RESULTS Through target region capture and high-throughput sequencing, a three-generation family was able to identify a heterozygous G to A point mutation at position 490 (c.1468)G > A, which led to a valine to methionine substitution (V490M) at exon 12. This unique missense mutation was found at codon 1468. Eight members of the proband's family-two sisters and the proband himself-had the mutation found by Sanger sequencing. Interestingly, biomarker tests for amyloid in the proband's cerebrospinal fluid (CSF) indicated pathology consistent with Alzheimer's disease (AD). The mutation was expected to have a high likelihood of being pathogenic. CONCLUSIONS We firstly reported a novel mutation in the GRN gene at codon 490 (V490M) in exon 12 in a China FTD family. The CSF biomarker alterations of the proband revealed a reduction in Aβ42 and the Aβ42/Aβ40 ratio. The analysis of mutation might support the role of GRN in patients with FTD and contribute to the discovery of a new pathological mechanism underlying the disease.
Collapse
Affiliation(s)
- Mingrong Xia
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Chenhao Gao
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Junkui Shang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Dan Li
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Ali Yang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Weizhou Zang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China.
| | - Jiewen Zhang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China.
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
3
|
McAnnis KE, Mbydeen TH, Jonnakuti VS, Davila PA, Laylani NA, Jurkute N, Ondo WG, Lee AG. Candidate GRN Variant in Patient With Progressive Supranuclear Palsy. J Neuroophthalmol 2024:00041327-990000000-00664. [PMID: 38913951 DOI: 10.1097/wno.0000000000002192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Affiliation(s)
- Kathryn E McAnnis
- Western University of Pacific Health Sciences (KEM), Lebanon, Oregon; Department of Neurology (THM), University of Leicester, Kettering General Hospital, Kettering, United Kingdom; Medical Scientist Training Program (VSJ), Baylor College of Medicine, Houston, Texas; Department of Ophthalmology and Neurology (PAD, NAL, WO, AGL), Houston Methodist Hospital, Houston, Texas; Department of Neuro-ophthalmology (NJ), Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom; Department of Neuro-ophthalmology (NJ), National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, United Kingdom; Institute of Ophthalmology (NJ), University College London, London, United Kingdom; Departments of Ophthalmology, Neurology, and Neurosurgery (WO, AGL), Weill Cornell Medicine, New York, New York; Department of Ophthalmology (AGL), University of Texas Medical Branch, Galveston, Texas; University of Texas MD Anderson Cancer Center (AGL), Houston, Texas; Texas A and M College of Medicine (AGL), Bryan, Texas; and Department of Ophthalmology (AGL), The University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Nguyen HD, Hoang TL, Vu GH. An in silico investigation of the toxicological effects and biological activities of 3-phenoxybenzoic acid and its metabolite products. Xenobiotica 2024; 54:322-341. [PMID: 38833509 DOI: 10.1080/00498254.2024.2361457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/12/2024] [Accepted: 05/26/2024] [Indexed: 06/06/2024]
Abstract
We aimed to elucidate the toxic effects and biological activities of 3-phenoxybenzoic acid (3PBA) and its metabolite products. Numerous in silico methods were used to identify the toxic effects and biological activities of 3PBA, including PASS online, molecular docking, ADMETlab 2.0, ADMESWISS, MetaTox, and molecular dynamic simulation. Ten metabolite products were identified via Phase II reactions (O-glucuronidation, O-sulfation, and methylation). All of the investigated compounds were followed by Lipinski's rule, indicating that they were stimulants or inducers of hazardous processes. Because of their high gastrointestinal absorption and ability to reach the blood-brain barrier, the studied compounds' physicochemical and pharmacokinetic properties matched existing evidence of harmful effects, including haematemesis, reproductive dysfunction, allergic dermatitis, toxic respiration, and neurotoxicity. The studied compounds have been linked to the apoptotic pathway, the reproductivity system, neuroendocrine disruptors, phospholipid-translocating ATPase inhibitors, and JAK2 expression. An O-glucuronidation metabolite product demonstrated higher binding affinity and interaction with CYP2C9, CYP3A4, caspase 3, and caspase 8 than 3PBA and other metabolite products, whereas metabolite products from methylation were predominant and more toxic. Our in silico findings partly meet the 3Rs principle by minimizing animal testing before more study is needed to identify the detrimental effects of 3PBA on other organs (liver, kidneys). Future research directions may involve experimental validation of in silico predictions, elucidation of molecular mechanisms, and exploration of therapeutic interventions. These findings contribute to our understanding of the toxicological profile of 3PBA and its metabolites, which has implications for risk assessment and regulatory decisions.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Division of Microbiology, Tulane National Private Research Center, Tulane University, Covington, LA, USA
| | - Thuy Linh Hoang
- College of Pharmacy, California Northstate University College of Pharmacy, CA, USA
| | - Giang Huong Vu
- Department of Public Heath, Hong Bang Health Center, Hai Phong, Vietnam
| |
Collapse
|
5
|
Chu M, Nan H, Jiang D, Liu L, Huang A, Wang Y, Wu L. Progranulin Gene Mutations in Chinese Patients with Frontotemporal Dementia: A Case Report and Literature Review. J Alzheimers Dis 2023; 93:225-234. [PMID: 36970912 DOI: 10.3233/jad-230052] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
BACKGROUND Progranulin (GRN) mutations in frontotemporal dementia (FTD) have been less frequently reported in China than in Western countries. OBJECTIVE This study reports a novel GRN mutation and summarizes the genetic and clinical features of patients with GRN mutations in China. METHODS Comprehensive clinical, genetic, and neuroimaging examinations were conducted on a 58-year-old female patient diagnosed with semantic variant primary progressive aphasia. A literature review was also conducted and clinical and genetic features of patients with GRN mutations in China were summarized. RESULTS Neuroimaging revealed marked lateral atrophy and hypometabolism in the left frontal, temporal, and parietal lobes. The patient was negative for pathologic amyloid and tau deposition by positron emission tomography. A novel heterozygous 45-bp deletion (c.1414-14_1444delCCCTTCCCCGCCAGGCTGTGTGCTGCGAGGATCGCCAGCACTGCT) was detected by whole-exome sequencing of the patient's genomic DNA. Nonsense-mediated mRNA decay was presumed to be involved in the degradation of the mutant gene transcript. The mutation was deemed pathogenic according to American College of Medical Genetics and Genomics criteria. The patient had a reduced plasma GRN level. In the literature, there were reports of 13 Chinese patients - mostly female - with GRN mutations; the prevalence was 1.2% -2.6% and patients mostly had early disease onset. CONCLUSION Our findings expand the mutation profile of GRN in China, which can aid the diagnosis and treatment of FTD.
Collapse
Affiliation(s)
- Min Chu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Haitian Nan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Deming Jiang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Li Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Anqi Huang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yihao Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Liyong Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Wang R, Gao H, Xie H, Jia Z, Chen Q. Molecular imaging biomarkers in familial frontotemporal lobar degeneration: Progress and prospects. Front Neurol 2022; 13:933217. [PMID: 36051222 PMCID: PMC9424494 DOI: 10.3389/fneur.2022.933217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/25/2022] [Indexed: 12/01/2022] Open
Abstract
Familial frontotemporal lobar degeneration (FTLD) is a pathologically heterogeneous group of neurodegenerative diseases with diverse genotypes and clinical phenotypes. Three major mutations were reported in patients with familial FTLD, namely, progranulin (GRN), microtubule-associated protein tau (MAPT), and the chromosome 9 open reading frame 72 (C9orf72) repeat expansion, which could cause neurodegenerative pathological changes years before symptom onset. Noninvasive quantitative molecular imaging with PET or single-photon emission CT (SPECT) allows for selective visualization of the molecular targets in vivo to investigate brain metabolism, perfusion, neuroinflammation, and pathophysiological changes. There was increasing evidence that several molecular imaging biomarkers tend to serve as biomarkers to reveal the early brain abnormalities in familial FTLD. Tau-PET with 18F-flortaucipir and 11C-PBB3 demonstrated the elevated tau position in patients with FTLD and also showed the ability to differentiate patterns among the different subtypes of the mutations in familial FTLD. Furthermore, dopamine transporter imaging with the 11C-DOPA and 11C-CFT in PET and the 123I-FP-CIT in SPECT revealed the loss of dopaminergic neurons in the asymptomatic and symptomatic patients of familial FTLD. In addition, PET imaging with the 11C-MP4A has demonstrated reduced acetylcholinesterase (AChE) activity in patients with FTLD, while PET with the 11C-DAA1106 and 11C-PK11195 revealed an increased level of microglial activation associated with neuroinflammation even before the onset of symptoms in familial FTLD. 18F-fluorodeoxyglucose (FDG)-PET indicated hypometabolism in FTLD with different mutations preceded the atrophy on MRI. Identifying molecular imaging biomarkers for familial FTLD is important for the in-vivo assessment of underlying pathophysiological changes with disease progression and future disease-modifying therapy. We review the recent progress of molecular imaging in familial FTLD with focused on the possible implication of these techniques and their prospects in specific mutation types.
Collapse
Affiliation(s)
- Ruihan Wang
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, China
| | - Hui Gao
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, China
| | - Hongsheng Xie
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Zhiyun Jia
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Qin Chen
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Qin Chen
| |
Collapse
|
7
|
Tao X, Zhou H, Mo D, Zhang W, Chang Z, Zeng Y, Luo Y, Wu S, Tang W, Yang C, Wang Q. Erythrocytes Are an Independent Protective Factor for Vascular Cognitive Impairment in Patients With Severe White Matter Hyperintensities. Front Aging Neurosci 2022; 14:789602. [PMID: 35250538 PMCID: PMC8894857 DOI: 10.3389/fnagi.2022.789602] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background and Purpose: Hemoglobin is one of the main proteins in erythrocytes. There are significant correlations between low hemoglobin and white matter hyperintensities (WMH) and cognitive impairment. This study explored whether erythrocytopenia has predictive value for vascular cognitive impairment (VCI) in patients with WMH. Method: We conducted a cross-sectional study of 302 patients, including 62 with cerebral small vessel disease and 240 with stroke. Basic demographic data and fasting blood were collected. First, all patients were divided into normal cognition (NC), mild VCI (mVCI), and severe VCI (sVCI) groups (subgroups later) based on cognitive behavior scores. Second, all patients were divided into mild WMH (mWMH) and severe WMH (sWMH) groups based on Fazekas scores. The differences in blood markers between different groups or subgroups with different cognitive levels were analyzed by univariate analysis. Then, binary logistic regression was used to analyze the diagnostic value of erythrocyte counts for VCI in the sWMH group, and ordinal logistic regression was used to analyze the predictive value of multiple variables for different cognitive levels. Results: Univariate analysis showed that erythrocytes, hemoglobin, high-sensitivity C-reactive protein, retinol binding protein and prealbumin were potential blood markers for different cognitive levels in sWMH patients. Among them, erythrocytopenia has good predictive value for the diagnosis of mVCI (AUC = 0.685, P = 0.008) or sVCI (AUC = 0.699, P = 0.003) in patients with sWMH. Multivariate joint analysis showed that erythrocytes were an independent protective factor reducing the occurrence of VCI in patients with sWMH (OR = 0.633, P = 0.045). Even after adjusting for age, there was still a significant difference (P = 0.047). Conclusion: Erythrocytes are an independent protective factor for VCI in patients with sWMH. Promoting hematopoietic function may have potential value for prevention of cognitive decline in patients with cerebrovascular disease.
Collapse
Affiliation(s)
- Xi Tao
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Neurological Rehabilitation, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Hang Zhou
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Danheng Mo
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Neurological Rehabilitation, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Wenjie Zhang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zihan Chang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yiheng Zeng
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuqi Luo
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Siyuan Wu
- Department of Neurological Rehabilitation, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Wenjing Tang
- Department of Neurological Rehabilitation, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Chen Yang
- Department of Neurological Rehabilitation, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Zhou ZD, Saw WT, Ho PGH, Zhang ZW, Zeng L, Chang YY, Sun AXY, Ma DR, Wang HY, Zhou L, Lim KL, Tan EK. The role of tyrosine hydroxylase-dopamine pathway in Parkinson's disease pathogenesis. Cell Mol Life Sci 2022; 79:599. [PMID: 36409355 PMCID: PMC9678997 DOI: 10.1007/s00018-022-04574-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by selective and progressive dopamine (DA) neuron loss in the substantia nigra and other brain regions, with the presence of Lewy body formation. Most PD cases are sporadic, whereas monogenic forms of PD have been linked to multiple genes, including Leucine kinase repeat 2 (LRRK2) and PTEN-induced kinase 1 (PINK1), two protein kinase genes involved in multiple signaling pathways. There is increasing evidence to suggest that endogenous DA and DA-dependent neurodegeneration have a pathophysiologic role in sporadic and familial PD. METHODS We generated patient-derived dopaminergic neurons and human midbrain-like organoids (hMLOs), transgenic (TG) mouse and Drosophila models, expressing both mutant and wild-type (WT) LRRK2 and PINK1. Using these models, we examined the effect of LRRK2 and PINK1 on tyrosine hydroxylase (TH)-DA pathway. RESULTS We demonstrated that PD-linked LRRK2 mutations were able to modulate TH-DA pathway, resulting in up-regulation of DA early in the disease which subsequently led to neurodegeneration. The LRRK2-induced DA toxicity and degeneration were abrogated by wild-type (WT) PINK1 (but not PINK1 mutations), and early treatment with a clinical-grade drug, α-methyl-L-tyrosine (α-MT), a TH inhibitor, was able to reverse the pathologies in human neurons and TG Drosophila models. We also identified opposing effects between LRRK2 and PINK1 on TH expression, suggesting that functional balance between these two genes may regulate the TH-DA pathway. CONCLUSIONS Our findings highlight the vital role of the TH-DA pathway in PD pathogenesis. LRRK2 and PINK1 have opposing effects on the TH-DA pathway, and its balance affects DA neuron survival. LRRK2 or PINK1 mutations can disrupt this balance, promoting DA neuron demise. Our findings provide support for potential clinical trials using TH-DA pathway inhibitors in early or prodromic PD.
Collapse
Affiliation(s)
- Zhi Dong Zhou
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioural Disorders, 8 College Road, Singapore, 169857 Singapore
| | - Wuan Ting Saw
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Patrick Ghim Hoe Ho
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Zhi Wei Zhang
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Li Zeng
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Ya Yin Chang
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioural Disorders, 8 College Road, Singapore, 169857 Singapore
| | - Alfred Xu Yang Sun
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioural Disorders, 8 College Road, Singapore, 169857 Singapore
| | - Dong Rui Ma
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Hong Yan Wang
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioural Disorders, 8 College Road, Singapore, 169857 Singapore
| | - Lei Zhou
- Ocular Proteomics Laboratory, Singapore Eye Research Institute, Singapore, 169856 Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077 Singapore
- Ophthalmology and Visual Sciences Academic Clinical Research Program, Duke-NUS Medical School, Singapore, 169857 Singapore
| | - Kah Leong Lim
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
- Developmental of Stem Cell Biology and Regenerative Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232 Singapore
| | - Eng-King Tan
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
- Department of Neurology, Singapore General Hospital, National Neuroscience Institute, Outram Road, Singapore, 169608 Singapore
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioural Disorders, 8 College Road, Singapore, 169857 Singapore
| |
Collapse
|