251
|
Biundo R, Weis L, Fiorenzato E, Pistonesi F, Cagnin A, Bertoldo A, Anglani M, Cecchin D, Antonini A. The contribution of beta-amyloid to dementia in Lewy body diseases: a 1-year follow-up study. Brain Commun 2021; 3:fcab180. [PMID: 34458730 PMCID: PMC8390473 DOI: 10.1093/braincomms/fcab180] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 11/16/2022] Open
Abstract
Dementia in Lewy Body Diseases (Parkinson’s disease and dementia with Lewy Bodies) affects progression of disabilities, quality of life and well-being. Understanding its pathogenetic mechanisms is critical to properly implement disease-modifying strategies. It has been hypothesized that synuclein- and amyloid-pathology act synergistically aggravating cognitive decline in elderly patients but their precise contribution to dementia is debated. In this study, we aimed at exploring if presence of amyloid deposits influences clinical, cognitive and neuroanatomical correlates of mental decline in a cohort of 40 Parkinson’s disease patients with normal cognition (n = 5), mild cognitive impairment (n = 22), and dementia (n = 13) as well as in Dementia with Lewy Bodies (n = 10). Patients underwent simultaneous 3 T PET/MRI with [18F]-flutemetamol and were assessed with an extensive baseline motor and neuropsychological examination, which allowed level II diagnosis of mild cognitive impairment and dementia. The role of amyloid positivity on each cognitive domain, and on the rate of conversion to dementia at 1-year follow-up was explored. A Kaplan Meier and the Log Rank (Mantel–Cox) test were used to assess the pairwise differences in time-to-develop dementia in Parkinson’s disease patients with and without significant amyloidosis. Furthermore, the presence of an Alzheimer’s dementia-like morphological pattern was evaluated using visual and automated assessment of T1-weighted and T2-weighted MRI images. We observed similar percentage of amyloid deposits in Parkinson’s disease dementia and dementia with Lewy Bodies cohorts (50% in each group) with an overall prevalence of 34% of significant amyloid depositions in Lewy Body Diseases. PET amyloid positivity was associated with worse global cognition (Montreal Cognitive Assessment and Mini Mental State Examination), executive and language difficulties. At 12-month follow-up, amyloid positive Parkinson’s disease patients were more likely to have become demented than those without amyloidosis. Moreover, there was no difference in the presence of an Alzheimer’s disease-like atrophy pattern and in vascular load (at Fazekas scale) between Lewy Body Diseases with and without significant amyloid deposits. Our findings suggest that in Lewy Body Diseases, amyloid deposition enhances cognitive deficits, particularly attention-executive and language dysfunctions. However, the large number of patients without significant amyloid deposits among our cognitively impaired patients indicates that synuclein pathology itself plays a critical role in the development of dementia in Lewy Body Diseases.
Collapse
Affiliation(s)
- Roberta Biundo
- Department of General Psychology, University of Padua, Padua, Italy.,Study Center for Neurodegeneration (CESNE), University of Padua, Padua, Italy
| | - Luca Weis
- Parkinson and Movement Disorders Unit, Department of Neuroscience, University of Padua, Padua, Italy
| | | | - Francesca Pistonesi
- Parkinson and Movement Disorders Unit, Department of Neuroscience, University of Padua, Padua, Italy
| | - Annachiara Cagnin
- Department of Neuroscience, University of Padua, Padua, Italy.,Padova Neuroscience Center, University of Padua, Padua, Italy
| | | | | | - Diego Cecchin
- Padova Neuroscience Center, University of Padua, Padua, Italy.,Nuclear Medicine Unit, Department of Medicine-DIMED, Padua University Hospital, Padua, Italy
| | - Angelo Antonini
- Parkinson and Movement Disorders Unit, Department of Neuroscience, University of Padua, Padua, Italy.,Padova Neuroscience Center, University of Padua, Padua, Italy
| |
Collapse
|
252
|
Bassil F, Meymand ES, Brown HJ, Xu H, Cox TO, Pattabhiraman S, Maghames CM, Wu Q, Zhang B, Trojanowski JQ, Lee VMY. α-Synuclein modulates tau spreading in mouse brains. J Exp Med 2021; 218:211481. [PMID: 33091110 PMCID: PMC7588140 DOI: 10.1084/jem.20192193] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/12/2020] [Accepted: 07/24/2020] [Indexed: 12/17/2022] Open
Abstract
α-Synuclein (α-syn) and tau aggregates are the neuropathological hallmarks of Parkinson’s disease (PD) and Alzheimer’s disease (AD), respectively, although both pathologies co-occur in patients with these diseases, suggesting possible crosstalk between them. To elucidate the interactions of pathological α-syn and tau, we sought to model these interactions. We show that increased accumulation of tau aggregates occur following simultaneous introduction of α-syn mousepreformed fibrils (mpffs) and AD lysate–derived tau seeds (AD-tau) both in vitro and in vivo. Interestingly, the absence of endogenous mouse α-syn in mice reduces the accumulation and spreading of tau, while the absence of tau did not affect the seeding or spreading capacity of α-syn. These in vivo results are consistent with our in vitro data wherein the presence of tau has no synergistic effects on α-syn. Our results point to the important role of α-syn as a modulator of tau pathology burden and spreading in the brains of AD, PDD, and DLB patients.
Collapse
Affiliation(s)
- Fares Bassil
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.,AbbVie, Foundational Neuroscience Center, Cambridge, MA
| | - Emily S Meymand
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Hannah J Brown
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Hong Xu
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Timothy O Cox
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Shankar Pattabhiraman
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Chantal M Maghames
- The Department of Cancer Biology and Abramson Family Cancer Research Institute, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Qihui Wu
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Bin Zhang
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - John Q Trojanowski
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Virginia M-Y Lee
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
253
|
ASL expression in ALDH1A1 + neurons in the substantia nigra metabolically contributes to neurodegenerative phenotype. Hum Genet 2021; 140:1471-1485. [PMID: 34417872 PMCID: PMC8460544 DOI: 10.1007/s00439-021-02345-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 08/12/2021] [Indexed: 12/29/2022]
Abstract
Argininosuccinate lyase (ASL) is essential for the NO-dependent regulation of tyrosine hydroxylase (TH) and thus for catecholamine production. Using a conditional mouse model with loss of ASL in catecholamine neurons, we demonstrate that ASL is expressed in dopaminergic neurons in the substantia nigra pars compacta, including the ALDH1A1 + subpopulation that is pivotal for the pathogenesis of Parkinson disease (PD). Neuronal loss of ASL results in catecholamine deficiency, in accumulation and formation of tyrosine aggregates, in elevation of α-synuclein, and phenotypically in motor and cognitive deficits. NO supplementation rescues the formation of aggregates as well as the motor deficiencies. Our data point to a potential metabolic link between accumulations of tyrosine and seeding of pathological aggregates in neurons as initiators for the pathological processes involved in neurodegeneration. Hence, interventions in tyrosine metabolism via regulation of NO levels may be therapeutic beneficial for the treatment of catecholamine-related neurodegenerative disorders.
Collapse
|
254
|
Niu F, Sharma A, Wang Z, Feng L, Muresanu DF, Sahib S, Tian ZR, Lafuente JV, Buzoianu AD, Castellani RJ, Nozari A, Menon PK, Patnaik R, Wiklund L, Sharma HS. Nanodelivery of oxiracetam enhances memory, functional recovery and induces neuroprotection following concussive head injury. PROGRESS IN BRAIN RESEARCH 2021; 265:139-230. [PMID: 34560921 DOI: 10.1016/bs.pbr.2021.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Military personnel are the most susceptible to concussive head injury (CHI) caused by explosion, blast or missile or blunt head trauma. Mild to moderate CHI could induce lifetime functional and cognitive disturbances causing significant decrease in quality of life. Severe CHI leads to instant death and lifetime paralysis. Thus, further exploration of novel therapeutic agents or new features of known pharmacological agents are needed to enhance quality of life of CHI victims. Previous reports from our laboratory showed that mild CHI induced by weight drop technique causing an impact of 0.224N results in profound progressive functional deficit, memory impairment and brain pathology from 5h after trauma that continued over several weeks of injury. In this investigation we report that TiO2 nanowired delivery of oxiracetam (50mg/kg, i.p.) daily for 5 days after CHI resulted in significant improvement of functional deficit on the 8th day. This was observed using Rota Rod treadmill, memory improvement assessed by the time spent in finding hidden platform under water. The motor function improvement is seen in oxiracetam treated CHI group by placing forepaw on an inclined mesh walking and foot print analysis for stride length and distance between hind feet. TiO2-nanowired oxiracetam also induced marked improvements in the cerebral blood flow, reduction in the BBB breakdown and edema formation as well as neuroprotection of neuronal, glial and myelin damages caused by CHI at light and electron microscopy on the 7th day after 5 days TiO2 oxiracetam treatment. Adverse biochemical events such as upregulation of CSF nitrite and nitrate, IL-6, TNF-a and p-Tau are also reduced significantly in oxiracetam treated CHI group. On the other hand post treatment of 100mg/kg dose of normal oxiracetam in identical conditions after CHI is needed to show slight but significant neuroprotection together with mild recovery of memory function and functional deficits on the 8th day. These observations are the first to point out that nanowired delivery of oxiracetam has superior neuroprotective ability in CHI. These results indicate a promising clinical future of TiO2 oxiracetam in treating CHI patients for better quality of life and neurorehabilitation, not reported earlier.
Collapse
Affiliation(s)
- Feng Niu
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, China
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Zhenguo Wang
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, China
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - Preeti K Menon
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
255
|
The extracellular chaperone Clusterin enhances Tau aggregate seeding in a cellular model. Nat Commun 2021; 12:4863. [PMID: 34381050 PMCID: PMC8357826 DOI: 10.1038/s41467-021-25060-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
Spreading of aggregate pathology across brain regions acts as a driver of disease progression in Tau-related neurodegeneration, including Alzheimer’s disease (AD) and frontotemporal dementia. Aggregate seeds released from affected cells are internalized by naïve cells and induce the prion-like templating of soluble Tau into neurotoxic aggregates. Here we show in a cellular model system and in neurons that Clusterin, an abundant extracellular chaperone, strongly enhances Tau aggregate seeding. Upon interaction with Tau aggregates, Clusterin stabilizes highly potent, soluble seed species. Tau/Clusterin complexes enter recipient cells via endocytosis and compromise the endolysosomal compartment, allowing transfer to the cytosol where they propagate aggregation of endogenous Tau. Thus, upregulation of Clusterin, as observed in AD patients, may enhance Tau seeding and possibly accelerate the spreading of Tau pathology. Variants of the extracellular chaperone Clusterin are associated with Alzheimer’s disease (AD) and Clusterin levels are elevated in AD patient brains. Here, the authors show that Clusterin binds to oligomeric Tau, which enhances the seeding capacity of Tau aggregates upon cellular uptake. They also demonstrate that Tau/Clusterin complexes enter cells via the endosomal pathway, resulting in damage to endolysosomes and entry into the cytosol, where they induce the aggregation of endogenous, soluble Tau.
Collapse
|
256
|
Yin P, Bai D, Zhu L, Deng F, Guo X, Li B, Chen L, Li S, Li XJ. Cytoplasmic TDP-43 impairs the activity of the ubiquitin-proteasome system. Exp Neurol 2021; 345:113833. [PMID: 34363810 DOI: 10.1016/j.expneurol.2021.113833] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 11/25/2022]
Abstract
The cytoplasmic inclusions of nuclear TAR DNA-binding protein 43 (TDP-43) are a pathologic hallmark in amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration (FTD), and other neurological disorders. We reported that expressing mutant TDP-43(M337V) in rhesus monkeys can mimic the cytoplasmic mislocalization of mutant TDP-43 seen in patient brains. Here we investigated how cytoplasmic mutant TDP-43 mediates neuropathology. We found that C-terminal TDP-43 fragments are primarily localized in the cytoplasm and that the age-dependent elevated UBE2N promotes the accumulation of cytoplasmic C-terminal TDP-43 via K63 ubiquitination. Immunoprecipitation and mass spectrometry revealed that cytoplasmic mutant TDP-43 interacts with proteasome assembly proteins PSMG2 and PSD13, which might lead to the impairment of the proteasomal activity. Our findings suggest that cytoplasmic TDP-43 may participate in age-dependent accumulation of misfolded proteins in the brain by inhibiting the UPS activity.
Collapse
Affiliation(s)
- Peng Yin
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China.
| | - Dazhang Bai
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Longhong Zhu
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Fuyu Deng
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Xiangyu Guo
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Bang Li
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Laiqiang Chen
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Shihua Li
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China.
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
257
|
Yan M, Zhang S, Li C, Liu Y, Zhao J, Wang Y, Yang Y, Zhang L. 5-Lipoxygenase as an emerging target against age-related brain disorders. Ageing Res Rev 2021; 69:101359. [PMID: 33984528 DOI: 10.1016/j.arr.2021.101359] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/30/2021] [Accepted: 05/07/2021] [Indexed: 12/15/2022]
Abstract
Neuroinflammation is a common feature of age-related brain disorders including Alzheimer's disease (AD), Parkinson's disease (PD) and cerebral ischemia. 5-lipoxygenase (5-LOX), a proinflammatory enzyme, modulates inflammation by generating leukotrienes. Abnormal activation of 5-LOX and excessive production of leukotrienes have been detected in the development of age-related brain pathology. In this review, we provide an update on the current understanding of 5-LOX activation and several groups of functionally related inhibitors. In addition, the modulatory roles of 5-LOX in the pathogenesis and progression of the age-related brain disorders have been comprehensively highlighted and discussed. Inhibition of 5-LOX activation may represent a promising therapeutic strategy for AD, PD and cerebral ischemia.
Collapse
|
258
|
Kakoty V, K C S, Dubey SK, Yang CH, Taliyan R. Neuroprotective Effects of Trehalose and Sodium Butyrate on Preformed Fibrillar Form of α-Synuclein-Induced Rat Model of Parkinson's Disease. ACS Chem Neurosci 2021; 12:2643-2660. [PMID: 34197084 DOI: 10.1021/acschemneuro.1c00144] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Therapeutic options for Parkinson's disease (PD) are limited to a symptomatic approach, making it a global threat. Targeting aggregated alpha-synuclein (α-syn) clearance is a gold standard for ameliorating PD pathology, bringing autophagy into the limelight. Expression of autophagy related genes are under the regulation by histone modifications, however, its relevance in PD is yet to be established. Here, preformed fibrillar form (PFF) of α-syn was used to induce PD in wistar rats, which were thereafter subjected to treatment with trehalose (tre, 4g/kg, orally), a potent autophagy inducer and sodium butyrate (SB, 300 mg/kg, orally), a pan histone deacetylase inhibitor alone as well as in combination. The combination treatment significantly reduced motor deficits as evidenced after rotarod, narrow beam walk, and open field tests. Novel object location and recognition tests were performed to govern cognitive abnormality associated with advanced stage PD, which was overcome by the combination treatment. Additionally, with the combination, the level of pro-inflammatory cytokines were significantly reduced, along with elevated levels of dopamine and histone H3 acetylation. Further, mRNA analysis revealed that levels of certain autophagy related genes and proteins implicated in PD pathogenesis significantly improved after administration of both tre and SB. Immunofluorescence and H&E staining in the substantia nigra region mirrored a potential improvement after treatment with both tre and SB. Therefore, outcomes of the present study were adequate to prove that combinatorial efficacy with tre and SB may prove to be a formidable insight into ameliorating PD exacerbated by PFF α-syn as compared to its individual efficacy.
Collapse
Affiliation(s)
- Violina Kakoty
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, 333031 Pilani, Rajasthan, India
| | - Sarathlal K C
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, 333031 Pilani, Rajasthan, India
| | - Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, 333031 Pilani, Rajasthan, India
| | - Chih-Hao Yang
- Department of Pharmacology, Taipei Medical University, Taipei 110, Taiwan
| | - Rajeev Taliyan
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, 333031 Pilani, Rajasthan, India
| |
Collapse
|
259
|
Meyer A, Handabaka I, Ehrensperger MM, Gschwandtner U, Hatz F, Monsch AU, Stieglitz RD, Fuhr P. A Comparison of Serial Position Effects in Patients with Mild Cognitive Impairment due to Parkinson's Disease or to Alzheimer's Disease. Dement Geriatr Cogn Disord 2021; 49:170-178. [PMID: 32634809 DOI: 10.1159/000507757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/06/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The first (primacy region) and last (recency region) items of a word list are generally better memorized than items from the middle region. The recency effect depends on short-term memory (STM) and the primacy effect on long-term memory (LTM), where verbal information is transferred from STM into LTM by maintenance rehearsal. We compared the serial position effects (SPE) between patients with mild cognitive impairment (MCI) due to Parkinson's disease (PD), i.e., PD-MCI, and patients with MCI due to Alzheimer's disease (AD-MCI), and evaluated the influence of SPE and frontostriatal deficits on verbal memory recall. METHODS Four similar groups of subjects participated in the study: 26 PD-MCI patients, 26 cognitively normal patients with PD (PD-CN), 26 AD-MCI patients, and 26 normal controls (NC). Verbal episodic memory, verbal span, attentional capacity, executive functions, and verbal working memory performance were assessed. Measures for primacy and recency regions were defined at the first trial of a 16-items word list. Hierarchical regression models were used to investigate the contribution of frontostriatal deficits beyond SPE on verbal memory recall performance ("long-delay free recall") in PD and AD patients. RESULTS Primacy effects were significantly diminished in both PD-MCI and AD-MCI patients relative to NC and PD-CN (all p < 0.01). Compared to PD-MCI patients, AD-MCI patients exhibited significantly worse "delayed-recall 'savings'." Reduced primacy effect was predictive for decreased recall performance in PD and AD. The conducted hierarchical regression model revealed that in PD, but not in AD patients, performance of attention and executive function significantly increased the prediction of free recalled words. CONCLUSIONS Reduced recall performance is likely due to impaired transition of newly learned material from STM into LTM in AD and in PD. Whereas AD-MCI patients suffer from a storage deficit, the similarly reduced recall performance found in patients with PD-MCI may additionally be related to deficient attentional and executive capacity.
Collapse
Affiliation(s)
- Antonia Meyer
- Department of Neurology, University Hospital of Basel, Basel, Switzerland,
| | - Ivana Handabaka
- Department of Neurology, University Hospital of Basel, Basel, Switzerland
| | - Michael M Ehrensperger
- Memory Clinic, University Center for Medicine of Aging Basel, Felix Platter Hospital, Basel, Switzerland
| | - Ute Gschwandtner
- Department of Neurology, University Hospital of Basel, Basel, Switzerland
| | - Florian Hatz
- Department of Neurology, University Hospital of Basel, Basel, Switzerland
| | - Andreas U Monsch
- Memory Clinic, University Center for Medicine of Aging Basel, Felix Platter Hospital, Basel, Switzerland
| | - Rolf D Stieglitz
- Department of Psychology, University of Basel, Basel, Switzerland
| | - Peter Fuhr
- Department of Neurology, University Hospital of Basel, Basel, Switzerland
| |
Collapse
|
260
|
Baek MS, Lee MJ, Kim HK, Lyoo CH. Temporal trajectory of biofluid markers in Parkinson's disease. Sci Rep 2021; 11:14820. [PMID: 34285331 PMCID: PMC8292456 DOI: 10.1038/s41598-021-94345-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/09/2021] [Indexed: 11/18/2022] Open
Abstract
Full dynamics of biofluid biomarkers have been unknown in patients with Parkinson’s disease (PD). Using data from 396 PD patients and 182 controls in the Parkinson's Progression Markers Initiative (PPMI) database, we estimated long-term temporal trajectories of CSF α-synuclein (α-syn), amyloid-β (Aβ), total tau (t-tau), phosphorylated tau (p-tau) and serum neurofilament light chain (NfL) by integrating function between the baseline levels and annual changes. At baseline, PD patients showed lower CSF α-syn, Aβ, t-tau and p-tau levels than those of the controls. In all PD patients, CSF α-syn and Aβ decreased in a negative exponential pattern before the onset of motor symptoms, whereas CSF t-tau and p-tau, and serum NfL increased. Patients with cognitive impairment exhibited faster decline of Aβ and α-syn and faster rise of t-tau, p-tau and NfL, when compared to those without. Similarly, low Aβ group showed earlier decline of α-syn, faster rise of t-tau, p-tau and NfL, and faster decline of cognitive performances, when compared to high Aβ group. Our results suggest that longitudinal changes in biomarkers can be influenced by cognitive impairment and Aβ burden at baseline. PD patients with Aβ pathology may be associated with early appearance of α-synuclein pathology, rapid progression of axonal degeneration and neurodegeneration, and consequently greater cognitive decline.
Collapse
Affiliation(s)
- Min Seok Baek
- Department of Neurology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Gangwon do, Republic of Korea.,Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, Republic of Korea
| | - Myung Jun Lee
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Gudeok-ro 179, Seo-gu, Busan, 49241, Republic of Korea.
| | - Han-Kyeol Kim
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, Republic of Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, Republic of Korea
| |
Collapse
|
261
|
Querfurth H, Lee HK. Mammalian/mechanistic target of rapamycin (mTOR) complexes in neurodegeneration. Mol Neurodegener 2021; 16:44. [PMID: 34215308 PMCID: PMC8252260 DOI: 10.1186/s13024-021-00428-5] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Novel targets to arrest neurodegeneration in several dementing conditions involving misfolded protein accumulations may be found in the diverse signaling pathways of the Mammalian/mechanistic target of rapamycin (mTOR). As a nutrient sensor, mTOR has important homeostatic functions to regulate energy metabolism and support neuronal growth and plasticity. However, in Alzheimer's disease (AD), mTOR alternately plays important pathogenic roles by inhibiting both insulin signaling and autophagic removal of β-amyloid (Aβ) and phospho-tau (ptau) aggregates. It also plays a role in the cerebrovascular dysfunction of AD. mTOR is a serine/threonine kinase residing at the core in either of two multiprotein complexes termed mTORC1 and mTORC2. Recent data suggest that their balanced actions also have implications for Parkinson's disease (PD) and Huntington's disease (HD), Frontotemporal dementia (FTD) and Amyotrophic Lateral Sclerosis (ALS). Beyond rapamycin; an mTOR inhibitor, there are rapalogs having greater tolerability and micro delivery modes, that hold promise in arresting these age dependent conditions.
Collapse
Affiliation(s)
- Henry Querfurth
- Department of Neurology, Tufts Medical Center, Boston, Massachusetts, USA.
| | - Han-Kyu Lee
- Department of Neurology, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
262
|
Rai SK, Savastano A, Singh P, Mukhopadhyay S, Zweckstetter M. Liquid-liquid phase separation of tau: From molecular biophysics to physiology and disease. Protein Sci 2021; 30:1294-1314. [PMID: 33930220 PMCID: PMC8197432 DOI: 10.1002/pro.4093] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 12/14/2022]
Abstract
Biomolecular condensation via liquid-liquid phase separation (LLPS) of intrinsically disordered proteins/regions (IDPs/IDRs), with and without nucleic acids, has drawn widespread interest due to the rapidly unfolding role of phase-separated condensates in a diverse range of cellular functions and human diseases. Biomolecular condensates form via transient and multivalent intermolecular forces that sequester proteins and nucleic acids into liquid-like membrane-less compartments. However, aberrant phase transitions into gel-like or solid-like aggregates might play an important role in neurodegenerative and other diseases. Tau, a microtubule-associated neuronal IDP, is involved in microtubule stabilization, regulates axonal outgrowth and transport in neurons. A growing body of evidence indicates that tau can accomplish some of its cellular activities via LLPS. However, liquid-to-solid transition resulting in the abnormal aggregation of tau is associated with neurodegenerative diseases. The physical chemistry of tau is crucial for governing its propensity for biomolecular condensation which is governed by various intermolecular and intramolecular interactions leading to simple one-component and complex multi-component condensates. In this review, we aim at capturing the current scientific state in unveiling the intriguing molecular mechanism of phase separation of tau. We particularly focus on the amalgamation of existing and emerging biophysical tools that offer unique spatiotemporal resolutions on a wide range of length- and time-scales. We also discuss the link between quantitative biophysical measurements and novel biological insights into biomolecular condensation of tau. We believe that this account will provide a broad and multidisciplinary view of phase separation of tau and its association with physiology and disease.
Collapse
Affiliation(s)
- Sandeep K. Rai
- Centre for Protein Science, Design and Engineering, Department of Biological Sciences, and Department of Chemical SciencesIndian Institute of Science Education and Research (IISER)MohaliIndia
| | - Adriana Savastano
- Research group Translational Structural BiologyGerman Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
| | - Priyanka Singh
- Centre for Protein Science, Design and Engineering, Department of Biological Sciences, and Department of Chemical SciencesIndian Institute of Science Education and Research (IISER)MohaliIndia
| | - Samrat Mukhopadhyay
- Centre for Protein Science, Design and Engineering, Department of Biological Sciences, and Department of Chemical SciencesIndian Institute of Science Education and Research (IISER)MohaliIndia
| | - Markus Zweckstetter
- Research group Translational Structural BiologyGerman Center for Neurodegenerative Diseases (DZNE)GöttingenGermany
- Department for NMR‐based Structural BiologyMax Planck Institute for Biophysical ChemistryGöttingenGermany
| |
Collapse
|
263
|
Choi A, Hallett M, Ehrlich D. Nutritional Ketosis in Parkinson's Disease - a Review of Remaining Questions and Insights. Neurotherapeutics 2021; 18:1637-1649. [PMID: 34235637 PMCID: PMC8608995 DOI: 10.1007/s13311-021-01067-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2021] [Indexed: 02/04/2023] Open
Abstract
Nutritional ketosis has promise for treating Parkinson's disease. Three previous studies explored the use of a ketogenic diet in cohorts with Parkinson's disease, and, while not conclusive, the data suggest non-motor symptom benefit. Before the ketogenic diet can be considered as a therapeutic option, it is important to establish with greater certainty that there is a reliable symptomatic benefit: which symptoms or groups of symptoms are impacted (if non-motor symptoms, which ones, and by which mechanism), what timescale is needed to obtain benefit, and how large an effect size can be achieved? To accomplish this, further investigation into the disease mechanisms based on pre-clinical data and hints from the clinical outcomes to date is useful to understand target engagement and gauge which mechanism could lead to a testable hypothesis. We review research pertaining to ketogenic diet, exogenous ketones, fasting, clinical studies, and theoretical review papers regarding therapeutic mechanisms from direct ketone body signaling and indirect metabolic effects. Through discussion of these findings and consideration of whether the ketogenic diet can be regarded as therapeutically useful for adjunctive therapy for Parkinson's disease, we identify remaining questions for the clinician to consider prior to recommending this diet.
Collapse
Affiliation(s)
- Alexander Choi
- Human Motor Control Section, Medical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, USA.
| | - Mark Hallett
- Human Motor Control Section, Medical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, USA
| | - Debra Ehrlich
- Office of the Clinical Director, Parkinson Disease Clinic, National Institute of Neurological Disorders and Stroke, Bethesda, USA
| |
Collapse
|
264
|
Davis AA, Inman CE, Wargel ZM, Dube U, Freeberg BM, Galluppi A, Haines JN, Dhavale DD, Miller R, Choudhury FA, Sullivan PM, Cruchaga C, Perlmutter JS, Ulrich JD, Benitez BA, Kotzbauer PT, Holtzman DM. APOE genotype regulates pathology and disease progression in synucleinopathy. Sci Transl Med 2021; 12:12/529/eaay3069. [PMID: 32024799 DOI: 10.1126/scitranslmed.aay3069] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022]
Abstract
Apolipoprotein E (APOE) ε4 genotype is associated with increased risk of dementia in Parkinson's disease (PD), but the mechanism is not clear, because patients often have a mixture of α-synuclein (αSyn), amyloid-β (Aβ), and tau pathologies. APOE ε4 exacerbates brain Aβ pathology, as well as tau pathology, but it is not clear whether APOE genotype independently regulates αSyn pathology. In this study, we generated A53T αSyn transgenic mice (A53T) on Apoe knockout (A53T/EKO) or human APOE knockin backgrounds (A53T/E2, E3, and E4). At 12 months of age, A53T/E4 mice accumulated higher amounts of brainstem detergent-insoluble phosphorylated αSyn compared to A53T/EKO and A53T/E3; detergent-insoluble αSyn in A53T/E2 mice was undetectable. By immunohistochemistry, A53T/E4 mice displayed a higher burden of phosphorylated αSyn and reactive gliosis compared to A53T/E2 mice. A53T/E2 mice exhibited increased survival and improved motor performance compared to other APOE genotypes. In a complementary model of αSyn spreading, striatal injection of αSyn preformed fibrils induced greater accumulation of αSyn pathology in the substantia nigra of A53T/E4 mice compared to A53T/E2 and A53T/EKO mice. In two separate cohorts of human patients with PD, APOE ε4/ε4 individuals showed the fastest rate of cognitive decline over time. Our results demonstrate that APOE genotype directly regulates αSyn pathology independent of its established effects on Aβ and tau, corroborate the finding that APOE ε4 exacerbates pathology, and suggest that APOE ε2 may protect against αSyn aggregation and neurodegeneration in synucleinopathies.
Collapse
Affiliation(s)
- Albert A Davis
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA. .,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Casey E Inman
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Zachary M Wargel
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Umber Dube
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University, St. Louis, MO 63110, USA
| | - Brittany M Freeberg
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Alexander Galluppi
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Jessica N Haines
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Dhruva D Dhavale
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Rebecca Miller
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Fahim A Choudhury
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Patrick M Sullivan
- Department of Medicine, Duke University Medical Center, Durham VAMC and Geriatric Research Clinical Center, Durham, NC 27705, USA
| | - Carlos Cruchaga
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University, St. Louis, MO 63110, USA
| | - Joel S Perlmutter
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA.,Departments of Neuroscience and Radiology, Programs in Physical and Occupational Therapy, Washington University, St. Louis, MO 63110, USA
| | - Jason D Ulrich
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Bruno A Benitez
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University, St. Louis, MO 63110, USA
| | - Paul T Kotzbauer
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - David M Holtzman
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA. .,Department of Neurology, Washington University, St. Louis, MO 63110, USA.,Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| |
Collapse
|
265
|
Kempster P. Prodromal and advanced non-motor features of Parkinson's disease. BMJ Neurol Open 2021; 3:e000168. [PMID: 34250486 PMCID: PMC8217910 DOI: 10.1136/bmjno-2021-000168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Peter Kempster
- Neurosciences Department, Monash Medical Centre, Clayton, Victoria, Australia
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
266
|
Huang X, Wang C, Chen L, Zhang T, Leung KL, Wong G. Human amyloid beta and α-synuclein co-expression in neurons impair behavior and recapitulate features for Lewy body dementia in Caenorhabditis elegans. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166203. [PMID: 34146705 DOI: 10.1016/j.bbadis.2021.166203] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022]
Abstract
Amyloid β (Aβ), a product of APP, and SNCA (α-synuclein (α-syn)) are two of the key proteins found in lesions associated with the age-related neurodegenerative disorders Alzheimer's disease (AD) and Parkinson's disease (PD), respectively. Previous clinical studies uncovered Aβ and α-syn co-expression in the brains of patients, which lead to Lewy body dementia (LBD), a disease encompassing Dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD). To explore the pathogenesis and define the relationship between Aβ and α-syn for LBD, we established a C. elegans model which co-expresses human Aβ and α-syn with alanine 53 to threonine mutant (α-syn(A53T)) in pan-neurons. Compared to α-syn(A53T) single transgenic animals, pan-neuronal Aβ and α-syn(A53T) co-expression further enhanced the thrashing, egg laying, serotonin and cholinergic signaling deficits, and dopaminergic neuron damage in C. elegans. In addition, Aβ increased α-syn expression in transgenic animals. Transcriptome analysis of both Aβ;α-syn(A53T) strains and DLB patients showed common downregulation in lipid metabolism and lysosome function genes, suggesting that a decrease of lysosome function may reduce the clearance ability in DLB, and this may lead to the further pathogenic protein accumulation. These findings suggest that our model can recapitulate some features in LBD and provides a mechanism by which Aβ may exacerbate α-syn pathogenesis.
Collapse
Affiliation(s)
- Xiaobing Huang
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Changliang Wang
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Liang Chen
- Department of Computer Science, College of Engineering, Shantou University, Shantou 515063, China; Key Laboratory of Intelligent Manufacturing Technology of Ministry of Education, Shantou University, Shantou 515063, China
| | - Tianjiao Zhang
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Ka Lai Leung
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Garry Wong
- Cancer Centre, Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China.
| |
Collapse
|
267
|
miR-101-3p Contributes to α-Synuclein Aggregation in Neural Cells through the miR-101-3p/SKP1/PLK2 Pathway. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:6147434. [PMID: 34234930 PMCID: PMC8216813 DOI: 10.1155/2021/6147434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/02/2021] [Indexed: 11/17/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by progressive neuronal loss in different brain regions, including the dopaminergic (DA) neurons of the substantia nigra pars compacta (SNc). The aggregation of α-synuclein (α-Syn) plays an essential role in the progression of PD-related neuron toxicity. In this study, bioinformatic analysis was used to confirm differentially expressed genes between patients with PD and healthy donors. Immunofluorescence was used to study the aggregation of α-Syn. Flow cytometry was used to confirm the apoptosis of neurons. Western blot was used to investigate the underlying mechanism. Coimmunoprecipitation (co-IP) was used to verify the interaction between proteins. Luciferase activity assay was used to confirm the target gene of miRNA. In vitro protein ubiquitination assay was used to ascertain the role of S-phase kinase-associated protein 1 (SKP1) on the ubiquitination processes of polo-like kinase 2 (PLK2). The result indicated that miR-101-3p was overexpressed in the substantia nigra of the postmortem brains of patients with PD. The underlying role was investigated in the SH-SY5Y cell line. The overexpression of α-Syn did not result in toxicity or aggregation. However, the co-overexpression of miR-101-3p and α-Syn promoted aggregation and neuron toxicity. Luciferase activity assay indicated that SKP1 is a target gene of miR-101-3p. The co-IP experiment confirmed that SKP1 could directly interact with PLK2. In vitro protein ubiquitination assay confirmed that SKP1 could promote the ubiquitination and subsequent protein degradation of PLK2. We also observed that the cotransfection of short hairpin RNA that targets PLK2 and α-Syn overexpression plasmid results in the endoplasmic reticulum stress of neurons. Our results collectively provide evidence that miR-101-3p contributes to α-Syn aggregation in neurons through the miR-101-3p/SKP1/PLK2 pathway.
Collapse
|
268
|
T-Type Ca 2+ Enhancer SAK3 Activates CaMKII and Proteasome Activities in Lewy Body Dementia Mice Model. Int J Mol Sci 2021; 22:ijms22126185. [PMID: 34201181 PMCID: PMC8228122 DOI: 10.3390/ijms22126185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 12/28/2022] Open
Abstract
Lewy bodies are pathological characteristics of Lewy body dementia (LBD) and are composed of α-synuclein (α-Syn), which is mostly degraded via the ubiquitin–proteasome system. More importantly, 26S proteasomal activity decreases in the brain of LBD patients. We recently introduced a T-type calcium channel enhancer SAK3 (ethyl-8-methyl-2,4-dioxo-2-(piperidin-1-yl)- 2H-spiro[cyclopentane-1,3-imidazo [1,2-a]pyridin]-2-ene-3-carboxylate) for Alzheimer’s disease therapeutics. SAK3 enhanced the proteasome activity via CaMKII activation in amyloid precursor protein knock-in mice, promoting the degradation of amyloid-β plaques to improve cognition. At this point, we addressed whether SAK3 promotes the degradation of misfolded α-Syn and the aggregates in α-Syn preformed fibril (PFF)-injected mice. The mice were injected with α-Syn PFF in the dorsal striatum, and SAK3 (0.5 or 1.0 mg/kg) was administered orally for three months, either immediately or during the last month after injection. SAK3 significantly inhibited the accumulation of fibrilized phosphorylated-α-Syn in the substantia nigra. Accordingly, SAK3 significantly recovered mesencephalic dopamine neurons from cell death. Decreased α-Syn accumulation was closely associated with increased proteasome activity. Elevated CaMKII/Rpt-6 signaling possibly mediates the enhanced proteasome activity after SAK3 administration in the cortex and hippocampus. CaMKII/Rpt-6 activation also accounted for improved memory and cognition in α-Syn PFF-injected mice. These findings indicate that CaMKII/Rpt-6-dependent proteasomal activation by SAK3 recovers from α-Syn pathology in LBD.
Collapse
|
269
|
Kuebler L, Buss S, Leonov A, Ryazanov S, Schmidt F, Maurer A, Weckbecker D, Landau AM, Lillethorup TP, Bleher D, Saw RS, Pichler BJ, Griesinger C, Giese A, Herfert K. [ 11C]MODAG-001-towards a PET tracer targeting α-synuclein aggregates. Eur J Nucl Med Mol Imaging 2021; 48:1759-1772. [PMID: 33369690 PMCID: PMC8113290 DOI: 10.1007/s00259-020-05133-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/25/2020] [Accepted: 10/20/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE Deposition of misfolded alpha-synuclein (αSYN) aggregates in the human brain is one of the major hallmarks of synucleinopathies. However, a target-specific tracer to detect pathological aggregates of αSYN remains lacking. Here, we report the development of a positron emission tomography (PET) tracer based on anle138b, a compound shown to have therapeutic activity in animal models of neurodegenerative diseases. METHODS Specificity and selectivity of [3H]MODAG-001 were tested in in vitro binding assays using recombinant fibrils. After carbon-11 radiolabeling, the pharmacokinetic and metabolic profile was determined in mice. Specific binding was quantified in rats, inoculated with αSYN fibrils and using in vitro autoradiography in human brain sections of Lewy body dementia (LBD) cases provided by the Neurobiobank Munich (NBM). RESULTS [3H]MODAG-001 revealed a very high affinity towards pure αSYN fibrils (Kd = 0.6 ± 0.1 nM) and only a moderate affinity to hTau46 fibrils (Kd = 19 ± 6.4 nM) as well as amyloid-β1-42 fibrils (Kd = 20 ± 10 nM). [11C]MODAG-001 showed an excellent ability to penetrate the mouse brain. Metabolic degradation was present, but the stability of the parent compound improved after selective deuteration of the precursor. (d3)-[11C]MODAG-001 binding was confirmed in fibril-inoculated rat striata using in vivo PET imaging. In vitro autoradiography showed no detectable binding to aggregated αSYN in human brain sections of LBD cases, most likely, because of the low abundance of aggregated αSYN against background protein. CONCLUSION MODAG-001 provides a promising lead structure for future compound development as it combines a high affinity and good selectivity in fibril-binding assays with suitable pharmacokinetics and biodistribution properties.
Collapse
Affiliation(s)
- Laura Kuebler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Röntgenweg 13, 72076, Tübingen, Germany
| | - Sabrina Buss
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Röntgenweg 13, 72076, Tübingen, Germany
| | - Andrei Leonov
- MODAG GmbH, Mikroforum Ring 3, 55234, Wendelsheim, Germany
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077, Göttingen, Germany
| | - Sergey Ryazanov
- MODAG GmbH, Mikroforum Ring 3, 55234, Wendelsheim, Germany
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077, Göttingen, Germany
| | - Felix Schmidt
- MODAG GmbH, Mikroforum Ring 3, 55234, Wendelsheim, Germany
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Röntgenweg 13, 72076, Tübingen, Germany
| | | | - Anne M Landau
- Translational Neuropsychiatry Unit, Aarhus University, Norrebrogade 44, 8000, Aarhus, Denmark
- Department of Nuclear Medicine and PET-Centre, Aarhus University, Palle Juul-Jensens 165, J109, 8200, Aarhus, Denmark
| | - Thea P Lillethorup
- Department of Nuclear Medicine and PET-Centre, Aarhus University, Palle Juul-Jensens 165, J109, 8200, Aarhus, Denmark
| | - Daniel Bleher
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Röntgenweg 13, 72076, Tübingen, Germany
| | - Ran Sing Saw
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Röntgenweg 13, 72076, Tübingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Röntgenweg 13, 72076, Tübingen, Germany
| | - Christian Griesinger
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077, Göttingen, Germany.
- University Göttingen, Cluster of Excellence Multiscale Bioimaging Molecular Machines, 37077, Göttingen, Germany.
| | - Armin Giese
- MODAG GmbH, Mikroforum Ring 3, 55234, Wendelsheim, Germany.
| | - Kristina Herfert
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Röntgenweg 13, 72076, Tübingen, Germany.
| |
Collapse
|
270
|
Hippocampal miR-211-5p regulates neurogenesis and depression-like behaviors in the rat. Neuropharmacology 2021; 194:108618. [PMID: 34062164 DOI: 10.1016/j.neuropharm.2021.108618] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/13/2021] [Accepted: 05/15/2021] [Indexed: 01/22/2023]
Abstract
Emerging evidence has shown that microRNAs (miRNAs) contribute to the pathogenesis of depression, a potentially life-threatening and disabling mental disorder caused by the interaction of genetic and environmental factors. However, the specific miRNAs and their underlying molecular mechanisms as involved in the pathogenesis and development of depression remain largely unknown. In the present study, we screened miRNA expression profiles and found that miR-211-5p was significantly down-regulated within the dentate gyrus (DG) hippocampus in the chronic unpredictable mild stress (CUMS) induced rat model of depression. Deficits in miR-211-5p were accompanied with reductions in neurogenesis and increased apoptosis in these CUMS rats. In contrast, an up-regulation of miR-211-5p within the DG area in CUMS rats promoted neuronal neurogenesis, reduced neuronal apoptosis via suppression of the Dyrk1A/STAT3 signaling pathway and relieved depression-like behaviors in these CUMS rats. In rats subjected to a knock-down of miR-211-5p in the DG there was an increase in neuronal apoptosis and a decrease in neuronal regeneration, effects which were accompanied with an induction of depression-like behaviors. Taken together, the results of our study reveal that altered levels of miR-211-5p in the hippocampal DG area exert a significant impact on neurogenesis, apoptosis and thus depression-like behaviors in rats. These findings suggest that the miR-211-5p/Dyrk1A pathway plays an important role in the pathogenesis of depression and may serve as a potential therapeutic target for the treatment of depression.
Collapse
|
271
|
Kasho K, Krasauskas L, Smirnovas V, Stojkovič G, Morozova-Roche LA, Wanrooij S. Human Polymerase δ-Interacting Protein 2 (PolDIP2) Inhibits the Formation of Human Tau Oligomers and Fibrils. Int J Mol Sci 2021; 22:ijms22115768. [PMID: 34071254 PMCID: PMC8199196 DOI: 10.3390/ijms22115768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/13/2021] [Accepted: 05/26/2021] [Indexed: 11/16/2022] Open
Abstract
A central characteristic of Alzheimer’s disease (AD) and other tauopathies is the accumulation of aggregated and misfolded Tau deposits in the brain. Tau-targeting therapies for AD have been unsuccessful in patients to date. Here we show that human polymerase δ-interacting protein 2 (PolDIP2) interacts with Tau. With a set of complementary methods, including thioflavin-T-based aggregation kinetic assays, Tau oligomer-specific dot-blot analysis, and single oligomer/fibril analysis by atomic force microscopy, we demonstrate that PolDIP2 inhibits Tau aggregation and amyloid fibril growth in vitro. The identification of PolDIP2 as a potential regulator of cellular Tau aggregation should be considered for future Tau-targeting therapeutics.
Collapse
Affiliation(s)
- Kazutoshi Kasho
- Department of Medical Biochemistry and Biophysics, Umeå University, 90 187 Umeå, Sweden
| | - Lukas Krasauskas
- Life Sciences Center, Institute of Biotechnology, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Vytautas Smirnovas
- Life Sciences Center, Institute of Biotechnology, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Gorazd Stojkovič
- Department of Medical Biochemistry and Biophysics, Umeå University, 90 187 Umeå, Sweden
| | | | - Sjoerd Wanrooij
- Department of Medical Biochemistry and Biophysics, Umeå University, 90 187 Umeå, Sweden
| |
Collapse
|
272
|
Interactome Mapping Provides a Network of Neurodegenerative Disease Proteins and Uncovers Widespread Protein Aggregation in Affected Brains. Cell Rep 2021; 32:108050. [PMID: 32814053 DOI: 10.1016/j.celrep.2020.108050] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 02/15/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Interactome maps are valuable resources to elucidate protein function and disease mechanisms. Here, we report on an interactome map that focuses on neurodegenerative disease (ND), connects ∼5,000 human proteins via ∼30,000 candidate interactions and is generated by systematic yeast two-hybrid interaction screening of ∼500 ND-related proteins and integration of literature interactions. This network reveals interconnectivity across diseases and links many known ND-causing proteins, such as α-synuclein, TDP-43, and ATXN1, to a host of proteins previously unrelated to NDs. It facilitates the identification of interacting proteins that significantly influence mutant TDP-43 and HTT toxicity in transgenic flies, as well as of ARF-GEP100 that controls misfolding and aggregation of multiple ND-causing proteins in experimental model systems. Furthermore, it enables the prediction of ND-specific subnetworks and the identification of proteins, such as ATXN1 and MKL1, that are abnormally aggregated in postmortem brains of Alzheimer's disease patients, suggesting widespread protein aggregation in NDs.
Collapse
|
273
|
Chung CC, Chan L, Chen JH, Hung YC, Hong CT. Plasma Extracellular Vesicle α-Synuclein Level in Patients with Parkinson's Disease. Biomolecules 2021; 11:biom11050744. [PMID: 34067663 PMCID: PMC8155846 DOI: 10.3390/biom11050744] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The most established pathognomonic protein of Parkinson's disease (PD), α-synuclein, is extensively investigated for disease diagnosis and prognosis; however, investigations into whether the free form of α-synuclein in the blood functions as a PD biomarker have not been fruitful. Extracellular vesicles (EVs) secreted from cells and present in blood transport molecules are novel platforms for biomarker identification. In blood EVs, α-synuclein originates predominantly from the brain without the interference of the blood-brain barrier. The present study investigated the role of plasma EV-borne α-synuclein as a biomarker of PD. METHODS Patients with mild to moderate stages of PD (n = 116) and individuals without PD (n = 46) were recruited to serve as the PD study group and the control group, respectively. Plasma EVs were isolated, and immunomagnetic reduction-based immunoassay was used to assess EV α-synuclein levels. Conventional statistical analysis was performed using SPSS 25.0, and p < 0.05 was considered significant. RESULTS Compared with controls, we observed significantly lower plasma EV α-synuclein levels in the patients with PD (PD: 56.0 ± 3.7 fg/mL vs. control: 74.5 ± 4.3 fg/mL, p = 0.009), and the significance remained after adjustment for age and sex. Plasma EV α-synuclein levels in the patients with PD did not correlate with age, disease duration, Part I and II scores of the Unified Parkinson's Disease Rating Scale (UPDRS), or the Mini-Mental State Examination scores. However, such levels were significantly correlated with UPDRS Part III score, which assesses motor dysfunction. Furthermore, the severity of akinetic-rigidity symptoms, but not tremor, was inversely associated with plasma EV α-synuclein level. CONCLUSION Plasma EV α-synuclein was significantly different between the control and PD group and was associated with akinetic-rigidity symptom severity in patients with PD. This study corroborates the possible diagnostic and subtyping roles of plasma EV α-synuclein in patients with PD, and it further provides a basis for this protein's clinical relevance and feasibility as a PD biomarker.
Collapse
Affiliation(s)
- Chen-Chih Chung
- Department of Neurology, Taipei Medical University–Shuang Ho Hospital, New Taipei City 23561, Taiwan; (C.-C.C.); (L.C.); (J.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei 11031, Taiwan
| | - Lung Chan
- Department of Neurology, Taipei Medical University–Shuang Ho Hospital, New Taipei City 23561, Taiwan; (C.-C.C.); (L.C.); (J.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Jia-Hung Chen
- Department of Neurology, Taipei Medical University–Shuang Ho Hospital, New Taipei City 23561, Taiwan; (C.-C.C.); (L.C.); (J.-H.C.)
| | - Yi-Chieh Hung
- Department of Neurosurgery, Department of Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
- Department of Recreation and Healthcare Management, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan
- Correspondence: (Y.-C.H.); (C.-T.H.); Tel.: +886-2-224-900-88 (ext. 811) (C.-T.H.)
| | - Chien-Tai Hong
- Department of Neurology, Taipei Medical University–Shuang Ho Hospital, New Taipei City 23561, Taiwan; (C.-C.C.); (L.C.); (J.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (Y.-C.H.); (C.-T.H.); Tel.: +886-2-224-900-88 (ext. 811) (C.-T.H.)
| |
Collapse
|
274
|
Shin EJ, Park JW. Nanoaggregates Derived from Amyloid-beta and Alpha-synuclein Characterized by Sequential Quadruple Force Mapping. NANO LETTERS 2021; 21:3789-3797. [PMID: 33845574 DOI: 10.1021/acs.nanolett.1c00058] [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] [Indexed: 06/12/2023]
Abstract
Overlapping of Alzheimer's disease and Parkinson's disease is associated with the formation of hetero-oligomers derived from amyloid-beta and alpha-synuclein. However, the structural identity of the hetero-oligomer has yet to be elucidated, particularly at high resolution. Here, with atomic force microscopy, the surface structure of hetero-oligomer was examined with four AFM tips tethering one of the selected antibodies recognizing N-terminus or C-terminus of each peptide. All aggregates were found to be hetero-oligomers, and probability of recognizing the termini is higher than that for the homo-oligomers, suggesting that the termini of the former have a greater tendency to be located at the surface or the termini have more freedom to be recognized, probably through loose packing. The methodology in this study provides us with a new approach to elucidate the structure of such aggregates at the single-molecule level, allowing the exploration of other intrinsically disordered proteins frequently found in nature.
Collapse
Affiliation(s)
- Eun Ji Shin
- Department of Chemistry, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Joon Won Park
- Department of Chemistry, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
- Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
275
|
Tong Q, Chen L. Associations of Alzheimer's Disease Neuropathologic Changes with Clinical Presentations of Parkinson's Disease. J Alzheimers Dis 2021; 81:201-207. [PMID: 33720903 DOI: 10.3233/jad-210114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Parkinson's disease (PD) and Alzheimer's disease (AD) are the two most prevalent neurodegenerative diseases associated with age. Pathological studies have shown that these two diseases share a certain degree of neuropathological overlap. AD neuropathologic change contributes to cognitive impairment in PD. However, the impact of AD pathology on other clinical phenotypes in PD remains largely unknown. OBJECTIVE Herein we aimed to assess the impact of co-occurring AD neuropathologic change on the clinical phenotypes of PD. METHODS We examined 46 autopsy brains of PD patients and available clinical information to retrospectively assess the effects of comorbid AD pathology on dementia, hallucinations, and dyskinesia commonly seen in advanced PD. RESULTS AD neuropathology significantly increased the risk of hallucinations and dementia, but not dyskinesia in PD patients. Surprisingly, diffuse Lewy body pathology, but not AD pathology, was associated with the occurrence of dementia and hallucinations. Most importantly, we reported that the severity of neuronal loss in the locus coeruleus (LC), but not the severity of neuronal loss in the substantia nigra (SN), was associated with the occurrence of dyskinesia in advanced PD patients, while neither Lewy body scores in SN nor LC had significant effects. CONCLUSION We show for the first time that neuronal loss in LC contributes to dyskinesia. Understanding the relationships between the two distinct pathologies and their relevant clinical phenotypes will be crucial in the development of effective disease-modifying therapies for PD.
Collapse
Affiliation(s)
- Qiang Tong
- Department of Neurology, the Affiliated Huaian First People's Hospital of Nanjing Medical University, Jiangsu, China.,Division of Neuropathology, Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Liam Chen
- Division of Neuropathology, Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
276
|
Baik K, Yang JJ, Jung JH, Lee YH, Chung SJ, Yoo HS, Sohn YH, Lee PH, Lee JM, Ye BS. Structural connectivity networks in Alzheimer's disease and Lewy body disease. Brain Behav 2021; 11:e02112. [PMID: 33792194 PMCID: PMC8119831 DOI: 10.1002/brb3.2112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 02/14/2021] [Accepted: 02/17/2021] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE We evaluated disruption of the white matter (WM) network related with Alzheimer's disease (AD) and Lewy body disease (LBD), which includes Parkinson's disease and dementia with Lewy bodies. METHODS We consecutively recruited 37 controls and 77 patients with AD-related cognitive impairment (ADCI) and/or LBD-related cognitive impairment (LBCI). Diagnoses of ADCI and LBCI were supported by amyloid PET and dopamine transporter PET, respectively. There were 22 patients with ADCI, 19 patients with LBCI, and 36 patients with mixed ADCI/LBCI. We investigated the relationship between ADCI, LBCI, graph theory-based network measures on diffusion tensor images, and cognitive dysfunction using general linear models after controlling for age, sex, education, deep WM hyperintensities (WMH), periventricular WMH, and intracranial volume. RESULTS LBCI, especially mixed with ADCI, was associated with increased normalized path length and decreased normalized global efficiency. LBCI was related to the decreased nodal degree of left caudate, which was further associated with broad cognitive dysfunction. Decreased left caudate nodal degree was associated with decreased fractional anisotropy (FA) in the brain regions vulnerable to LBD. Compared with the control group, the LBCI group had an increased betweenness centrality in the occipital nodes, which was associated with decreased FA in the WM adjacent to the striatum and visuospatial dysfunction. CONCLUSION Concomitant ADCI and LBCI are associated with the accentuation of LBCI-related WM network disruption centered in the left caudate nucleus. The increase of occipital betweenness centrality could be a characteristic biologic change associated with visuospatial dysfunction in LBCI.
Collapse
Affiliation(s)
- Kyoungwon Baik
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Jin-Ju Yang
- Department of Biomedical Engineering, Hanyang University, Seoul, Korea
| | - Jin Ho Jung
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Yang Hyun Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Seok Jong Chung
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Han Soo Yoo
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Young H Sohn
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Jong-Min Lee
- Department of Biomedical Engineering, Hanyang University, Seoul, Korea
| | - Byoung Seok Ye
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
277
|
Wang H, Zhang M, Wei T, Zhou J, Zhang Y, Guo D. Long non-coding RNA SNHG1 mediates neuronal damage in Parkinson's disease model cells by regulating miR-216a-3p/Bcl-2-associated X protein. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:851. [PMID: 34164485 PMCID: PMC8184415 DOI: 10.21037/atm-21-1613] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background Parkinson’s disease (PD) is a common central nervous system degenerative disease in middle-aged and elderly people. Our study aimed to illuminate the relationship and mechanism of long-chain non-coding RNA SNHG1 and miRNA (miR)-216a-3p in PD. Methods Human neuroblastoma cell lines were treated with MPP+ to construct a PD model. Real-time fluorescent quantitative PCR was used to detect the cellular expression of SNHG1. Neuronal cell activity and apoptosis were compared before and after SNHG1 knock-down, as was neuronal miR-216a-3p expression. Further, a luciferase reporter gene experiment was performed to verify BAX as the target of miR-216a-3p. Anti-miR-216a-3p and BAX were co-transfected into PD model cells, and neuronal cellular activity and apoptosis were observed. Finally, the potential regulatory network of SNHG1/miR-216a-3p/BAX in PD was investigated. Results The expression of miR-216a-3p was decreased in the PD model cells, and re-expression reversed the high apoptotic rate and cell vitality inhibition in PD model cells. SNHG1 interacted with miR-216a-3p and negatively regulated its upstream molecules, while miR-216a-3p attenuated the effect of SNHG1 knock-down on neurons. The overexpression of BAX in the PD cell model blocked the damage by miR-216a-3p to neurons. At the same time, SNHG1 acted as a coordinator, mediating the regulation of BAX via miR-216a-3p, thereby affecting the activity and apoptotic rate of neurons in the PD model. Conclusions SNHG1 interacts with miR-216a-3p to regulate the expression of BAX. This SNHG1/miR-216a-3p/BAX molecular regulatory network is implicated in the pathogenesis of PD.
Collapse
Affiliation(s)
- Hai Wang
- Department of Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Meng Zhang
- Department of Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Taofeng Wei
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Zhou
- Center for Medicinal Resources Research, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Yongle Zhang
- Department of Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Dengjun Guo
- Department of Neurology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| |
Collapse
|
278
|
Nies YH, Mohamad Najib NH, Lim WL, Kamaruzzaman MA, Yahaya MF, Teoh SL. MicroRNA Dysregulation in Parkinson's Disease: A Narrative Review. Front Neurosci 2021; 15:660379. [PMID: 33994934 PMCID: PMC8121453 DOI: 10.3389/fnins.2021.660379] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is a severely debilitating neurodegenerative disease, affecting the motor system, leading to resting tremor, cogwheel rigidity, bradykinesia, walking and gait difficulties, and postural instability. The severe loss of dopaminergic neurons in the substantia nigra pars compacta causes striatal dopamine deficiency and the presence of Lewy bodies indicates a pathological hallmark of PD. Although the current treatment of PD aims to preserve dopaminergic neurons or to replace dopamine depletion in the brain, it is notable that complete recovery from the disease is yet to be achieved. Given the complexity and multisystem effects of PD, the underlying mechanisms of PD pathogenesis are yet to be elucidated. The advancement of medical technologies has given some insights in understanding the mechanism and potential treatment of PD with a special interest in the role of microRNAs (miRNAs) to unravel the pathophysiology of PD. In PD patients, it was found that striatal brain tissue and dopaminergic neurons from the substantia nigra demonstrated dysregulated miRNAs expression profiles. Hence, dysregulation of miRNAs may contribute to the pathogenesis of PD through modulation of PD-associated gene and protein expression. This review will discuss recent findings on PD-associated miRNAs dysregulation, from the regulation of PD-associated genes, dopaminergic neuron survival, α-synuclein-induced inflammation and circulating miRNAs. The next section of this review also provides an update on the potential uses of miRNAs as diagnostic biomarkers and therapeutic tools for PD.
Collapse
Affiliation(s)
- Yong Hui Nies
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Nor Haliza Mohamad Najib
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Wei Ling Lim
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Selangor, Malaysia
| | - Mohd Amir Kamaruzzaman
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
279
|
Zhang Y, Zhang C, Fan Y, Liu Z, Hu F, Zhao YS. Smart Protein-Based Biolasers: An Alternative Way to Protein Conformation Detection. ACS APPLIED MATERIALS & INTERFACES 2021; 13:19187-19192. [PMID: 33871261 DOI: 10.1021/acsami.0c22270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Detecting conformational changes in protein is imperative due to its major role in neurodegenerative disorders. Here, we propose an alternative strategy for monitoring the structural change of proteins based on biological microlasers. Smart responsive protein-based microscale biolasers were constructed by incorporating organic gain medium into the microspheres of silk fibroin via emulsion-solvent evaporation. The lasing characteristic of the biolasers exhibited a sensitive response to the structural transformation of the silk fibroin. With narrowed linewidth, the as-prepared biolasers as sensing signals enable highly sensitive protein conformation detection. These results offer an effective approach to monitoring the protein conformational changes and provide valuable guidance for a better understanding of the relationship between bio-microstructures and their photonic properties.
Collapse
Affiliation(s)
- Yue Zhang
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Chunhuan Zhang
- Key Laboratory of Photochemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Yuqing Fan
- Key Laboratory of Photochemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhen Liu
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Fengqin Hu
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Yong Sheng Zhao
- Key Laboratory of Photochemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
280
|
Hou Y, Li X, Liu C, Zhang M, Zhang X, Ge S, Zhao L. Neuroprotective effects of short-chain fatty acids in MPTP induced mice model of Parkinson's disease. Exp Gerontol 2021; 150:111376. [PMID: 33905875 DOI: 10.1016/j.exger.2021.111376] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/09/2023]
Abstract
Gut microbial metabolites, SCFAs, were related with the occurrence and development of Parkinson's disease (PD). But the effects of different short-chain fatty acids (SCFAs) on PD and involving mechanisms are still undefined. In this study we evaluate the effects of three dominant SCFAs (acetate, propionate and butyrate) on motor damage, dopaminergic neuronal degeneration and underlying neuroinflammation related mechanisms in 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced PD mice. High (2.0 g/kg) or low doses (0.2 g/kg) of sodium acetate (NaA), sodium propionate (NaP) or sodium butyrate (NaB) were gavaged into PD mice for 6 weeks. High doses of NaA reduced the turning time of PD mice. NaB significantly reduced the turning and total time in pole test, and increased the average velocity in open field test when compared with PD mice, indicating the most effective alleviation of PD-induced motor disorder. Low and high doses of NaB significantly increased the content of tyrosine hydroxylase (TH) by 12.3% and 20.2%, while reduced α-synuclein activation by 159.4% and 132.7% in the substantia nigra pars compacta (SNpc), compared with PD groups. Butyrate reached into the midbrain SNpc and suppressed microglia over-activation. It inhibited the levels of pro-inflammatory factors (IL-6, IL-1β and TNF-α) (P < 0.01) and iNOS. Besides, butyrate inhibited the activation of NF-κB and MAPK signaling pathways in the SNpc region. Consequently, sodium butyrate could inhibit neuroinflammation and alleviate neurological damage of PD.
Collapse
Affiliation(s)
- Yichao Hou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; Key Laboratory of Precision Nutrition and Food Quality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Xingqi Li
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Chang Liu
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Ming Zhang
- School of Food and Health, Beijing Technology and Business University, Beijing, 100048, China
| | - Xiaoying Zhang
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot 010080, China
| | - Shaoyang Ge
- Hebei Engineering Research Center of Animal Product, Sanhe 065200, China
| | - Liang Zhao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; Key Laboratory of Precision Nutrition and Food Quality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
281
|
Kee TR, Espinoza Gonzalez P, Wehinger JL, Bukhari MZ, Ermekbaeva A, Sista A, Kotsiviras P, Liu T, Kang DE, Woo JAA. Mitochondrial CHCHD2: Disease-Associated Mutations, Physiological Functions, and Current Animal Models. Front Aging Neurosci 2021; 13:660843. [PMID: 33967741 PMCID: PMC8100248 DOI: 10.3389/fnagi.2021.660843] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/31/2021] [Indexed: 12/19/2022] Open
Abstract
Rare mutations in the mitochondrial protein coiled-coil-helix-coiled-coil-helix domain containing 2 (CHCHD2) are associated with Parkinson's disease (PD) and other Lewy body disorders. CHCHD2 is a bi-organellar mediator of oxidative phosphorylation, playing crucial roles in regulating electron flow in the mitochondrial electron transport chain and acting as a nuclear transcription factor for a cytochrome c oxidase subunit (COX4I2) and itself in response to hypoxic stress. CHCHD2 also regulates cell migration and differentiation, mitochondrial cristae structure, and apoptosis. In this review, we summarize the known disease-associated mutations of CHCHD2 in Asian and Caucasian populations, the physiological functions of CHCHD2, how CHCHD2 mutations contribute to α-synuclein pathology, and current animal models of CHCHD2. Further, we discuss the necessity of continued investigation into the divergent functions of CHCHD2 and CHCHD10 to determine how mutations in these similar mitochondrial proteins contribute to different neurodegenerative diseases.
Collapse
Affiliation(s)
- Teresa R Kee
- USF Health Byrd Alzheimer's Center and Research Institute, Tampa, FL, United States.,Department of Molecular Pharmacology and Physiology, USF Health Morsani College of Medicine, Tampa, FL, United States
| | | | - Jessica L Wehinger
- USF Health Byrd Alzheimer's Center and Research Institute, Tampa, FL, United States
| | - Mohammed Zaheen Bukhari
- USF Health Byrd Alzheimer's Center and Research Institute, Tampa, FL, United States.,Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, United States
| | - Aizara Ermekbaeva
- USF Health Byrd Alzheimer's Center and Research Institute, Tampa, FL, United States
| | - Apoorva Sista
- USF Health Byrd Alzheimer's Center and Research Institute, Tampa, FL, United States
| | - Peter Kotsiviras
- USF Health Byrd Alzheimer's Center and Research Institute, Tampa, FL, United States
| | - Tian Liu
- USF Health Byrd Alzheimer's Center and Research Institute, Tampa, FL, United States.,Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, United States
| | - David E Kang
- USF Health Byrd Alzheimer's Center and Research Institute, Tampa, FL, United States.,Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, United States.,James A. Haley Veterans Administration Hospital, Tampa, FL, United States
| | - Jung-A A Woo
- USF Health Byrd Alzheimer's Center and Research Institute, Tampa, FL, United States.,Department of Molecular Pharmacology and Physiology, USF Health Morsani College of Medicine, Tampa, FL, United States
| |
Collapse
|
282
|
Cordone S, Scarpelli S, Alfonsi V, De Gennaro L, Gorgoni M. Sleep-Based Interventions in Alzheimer's Disease: Promising Approaches from Prevention to Treatment along the Disease Trajectory. Pharmaceuticals (Basel) 2021; 14:383. [PMID: 33921870 PMCID: PMC8073746 DOI: 10.3390/ph14040383] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/10/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023] Open
Abstract
The multifactorial nature of Alzheimer's disease (AD) has led scientific researchers to focus on the modifiable and treatable risk factors of AD. Sleep fits into this context, given the bidirectional relationship with AD confirmed by several studies over the last years. Sleep disorders appear at an early stage of AD and continue throughout the entire course of the pathology. Specifically, sleep abnormalities, such as more fragmented sleep, increase in time of awakenings, worsening of sleep quality and primary sleep disorders raise with the severity and progression of AD. Intervening on sleep, therefore, means acting both with prevention strategies in the pre-clinical phase and with treatments during the course of the disease. This review explores sleep disturbances in the different stages of AD, starting from the pre-clinical stage. Particular attention is given to the empirical evidence investigating obstructive sleep apnea (OSA) disorder and the mechanisms overlapping and sharing with AD. Next, we discuss sleep-based intervention strategies in the healthy elderly population, mild cognitive impairment (MCI) and AD patients. We mention interventions related to behavioral strategies, combination therapies, and bright light therapy, leaving extensive space for new and raising evidence on continuous positive air pressure (CPAP) treatment effectiveness. Finally, we clarify the role of NREM sleep across the AD trajectory and consider the most recent studies based on the promising results of NREM sleep enhancement, which use innovative experimental designs and techniques.
Collapse
Affiliation(s)
- Susanna Cordone
- UniCamillus, Saint Camillus International University of Health Sciences, 00131 Rome, Italy;
| | - Serena Scarpelli
- Department of Psychology, University of Rome “Sapienza”, 00185 Rome, Italy; (S.S.); (M.G.)
| | | | - Luigi De Gennaro
- Department of Psychology, University of Rome “Sapienza”, 00185 Rome, Italy; (S.S.); (M.G.)
- IRCCS Fondazione Santa Lucia, 00179 Rome, Italy;
| | - Maurizio Gorgoni
- Department of Psychology, University of Rome “Sapienza”, 00185 Rome, Italy; (S.S.); (M.G.)
| |
Collapse
|
283
|
Hirano S. Clinical implications for dopaminergic and functional neuroimage research in cognitive symptoms of Parkinson's disease. Mol Med 2021; 27:40. [PMID: 33858320 PMCID: PMC8048076 DOI: 10.1186/s10020-021-00301-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Evidence from dopaminergic image and cerebral blood flow/metabolism images have shed light on symptomatology of cognitive aspects in brain physiology of healthy human as well as patients with Parkinson's disease. Cognitive impairment in Parkinson's disease is characterized by executive, visuospatial, attentional disturbances. Dopaminergic system includes triadic parallel pathways. The mesostriatal pathway consist of posterolateral putamen and motor areas, the mesocortical pathway of dorsal caudate nucleus and dorsolateral prefrontal cortex, and the mesolimbic pathway of ventral striatum, anterior cingulate cortex. The mesocortical pathway is responsible for the executive function which may change by administration of dopaminergic medication. The mesolimbic pathway is associated with motivation and reward prediction which may result in depression or apathy when dopamine level was suboptimal, impulse control disorder and punding when dopamine was over the optimal level. Abnormal brain metabolism/perfusion related to cognitive impairment in Parkinson's disease are relatively reduced activity located in frontal and parietal association areas and relatively increased activity in the cerebellum. In the anterior brain, the mesocortical pathway, is responsible for verbal memory and executive function, which originates with caudate dopaminergic system and account for mild cognitive impairment of Parkinson's disease. The posterior brain system which includes the parietal, temporal, and occipital cortices, is responsible for the memory and visuospatial function, and related to cholinergic dysfunction and possibly glucocerebrosidase gene variants, relating to dementia in Parkinson's disease. The role of cerebellum in Parkinson's disease remains unclear but emerging evidence suggests that it may relate to the sequencing detection and affective symptoms. The dual syndrome hypothesis is helpful for understanding the mechanism of cognitive impairment in Parkinson's disease and optimal symptom management.
Collapse
Affiliation(s)
- Shigeki Hirano
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
284
|
Tangrodchanapong T, Sornkaew N, Yurasakpong L, Niamnont N, Nantasenamat C, Sobhon P, Meemon K. Beneficial Effects of Cyclic Ether 2-Butoxytetrahydrofuran from Sea Cucumber Holothuria scabra against Aβ Aggregate Toxicity in Transgenic Caenorhabditis elegans and Potential Chemical Interaction. Molecules 2021; 26:molecules26082195. [PMID: 33920352 PMCID: PMC8070609 DOI: 10.3390/molecules26082195] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/04/2021] [Accepted: 04/07/2021] [Indexed: 12/24/2022] Open
Abstract
The pathological finding of amyloid-β (Aβ) aggregates is thought to be a leading cause of untreated Alzheimer’s disease (AD). In this study, we isolated 2-butoxytetrahydrofuran (2-BTHF), a small cyclic ether, from Holothuria scabra and demonstrated its therapeutic potential against AD through the attenuation of Aβ aggregation in a transgenic Caenorhabditis elegans model. Our results revealed that amongst the five H. scabra isolated compounds, 2-BTHF was shown to be the most effective in suppressing worm paralysis caused by Aβ toxicity and in expressing strong neuroprotection in CL4176 and CL2355 strains, respectively. An immunoblot analysis showed that CL4176 and CL2006 treated with 2-BTHF showed no effect on the level of Aβ monomers but significantly reduced the toxic oligomeric form and the amount of 1,4-bis(3-carboxy-hydroxy-phenylethenyl)-benzene (X-34)-positive fibril deposits. This concurrently occurred with a reduction of reactive oxygen species (ROS) in the treated CL4176 worms. Mechanistically, heat shock factor 1 (HSF-1) (at residues histidine 63 (HIS63) and glutamine 72 (GLN72)) was shown to be 2-BTHF’s potential target that might contribute to an increased expression of autophagy-related genes required for the breakdown of the Aβ aggregate, thus attenuating its toxicity. In conclusion, 2-BTHF from H. scabra could protect C. elegans from Aβ toxicity by suppressing its aggregation via an HSF-1-regulated autophagic pathway and has been implicated as a potential drug for AD.
Collapse
Affiliation(s)
- Taweesak Tangrodchanapong
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (T.T.); (L.Y.); (P.S.)
| | - Nilubon Sornkaew
- Department of Chemistry, Faculty of Science, King Mongkut’s University of Technology Thonburi, Bangkok 10140, Thailand; (N.S.); (N.N.)
| | - Laphatrada Yurasakpong
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (T.T.); (L.Y.); (P.S.)
| | - Nakorn Niamnont
- Department of Chemistry, Faculty of Science, King Mongkut’s University of Technology Thonburi, Bangkok 10140, Thailand; (N.S.); (N.N.)
| | - Chanin Nantasenamat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand;
| | - Prasert Sobhon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (T.T.); (L.Y.); (P.S.)
| | - Krai Meemon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (T.T.); (L.Y.); (P.S.)
- Correspondence: or ; Tel.: +66-22-015-407
| |
Collapse
|
285
|
Duwa R, Jeong JH, Yook S. Development of immunotherapy and nanoparticles-based strategies for the treatment of Parkinson’s disease. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2021. [DOI: 10.1007/s40005-021-00521-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
286
|
Discovery of a Novel Acetylcholinesterase Inhibitor by Fragment-Based Design and Virtual Screening. Molecules 2021; 26:molecules26072058. [PMID: 33916760 PMCID: PMC8038331 DOI: 10.3390/molecules26072058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/27/2021] [Accepted: 03/31/2021] [Indexed: 11/17/2022] Open
Abstract
Despite extensive and intensive research efforts in recent decades, there is still no effective treatment for neurodegenerative diseases. On this background, the use of drugs inhibiting the enzyme acetylcholinesterase (AChE) remains an eternal evergreen in the symptomatic treatment of mild to moderate cognitive impairments. Even more, the cholinergic hypothesis, somewhat forgotten in recent years due to the shift in focus on amyloid cascade, is back to life, and the search for new, more effective AChE inhibitors continues. We generated a fragment-based library containing aromatic moieties and linkers originating from a set of novel AChE inhibitors. We used this library to design 1220 galantamine (GAL) derivatives following the model GAL (binding core) - linker (L) - aromatic fragment (Ar). The newly designed compounds were screened virtually for blood–brain barrier (BBB) permeability and binding to AChE. Among the top 10 best-scored compounds, a representative lead molecule was selected and tested for anti-AChE activity and neurotoxicity. It was found that the selected compound was a powerful non-toxic AChE inhibitor, 68 times more active than GAL, and could serve as a lead molecule for further optimization and development.
Collapse
|
287
|
Brooks DJ. Imaging Familial and Sporadic Neurodegenerative Disorders Associated with Parkinsonism. Neurotherapeutics 2021; 18:753-771. [PMID: 33432494 PMCID: PMC8423977 DOI: 10.1007/s13311-020-00994-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2020] [Indexed: 11/24/2022] Open
Abstract
In this paper, the structural and functional imaging changes associated with sporadic and genetic Parkinson's disease and atypical Parkinsonian variants are reviewed. The role of imaging for supporting diagnosis and detecting subclinical disease is discussed, and the potential use and drawbacks of using imaging biomarkers for monitoring disease progression is debated. Imaging changes associated with nonmotor complications of PD are presented. The similarities and differences in imaging findings in Lewy body dementia, Parkinson's disease dementia, and Alzheimer's disease are discussed.
Collapse
Affiliation(s)
- David J Brooks
- Department of Nuclear Medicine, Aarhus University, Aarhus N, 8200, Denmark.
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK.
| |
Collapse
|
288
|
Kim R, Park S, Yoo D, Jun JS, Jeon B. Association of Physical Activity and APOE Genotype With Longitudinal Cognitive Change in Early Parkinson Disease. Neurology 2021; 96:e2429-e2437. [PMID: 33790041 DOI: 10.1212/wnl.0000000000011852] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 02/10/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether greater physical activity could modify the negative association of APOE ε4 with longitudinal cognitive changes in early Parkinson disease (PD) and to uncover the disease-specific mechanism for explaining such benefits of physical activity. METHODS We used data from the Parkinson's Progression Markers Initiative cohort. Because self-reported physical activity, measured by the Physical Activity Scale of the Elderly, was initiated at 2 years after enrollment, this longitudinal analysis was based on assessments performed at years 2, 3, and 4. Cognitive function was measured annually with the Montreal Cognitive Assessment (MoCA). Dopamine transporter (DAT) imaging was performed at years 2 and 4. We assessed the interactive associations between physical activity and the APOE ε4 allele on the longitudinal changes in MoCA scores and striatal DAT activities. RESULTS A total of 173 patients with early PD (age 63.3 ± 10.0 years, 27% APOE ε4 carriers) were included. The APOE ε4 allele showed a steeper rate of cognitive decline than the non-APOE ε4 allele (estimate -1.33, 95% confidence interval [CI] -2.12 to -0.47, p = 0.002). However, there was a significant interaction between physical activity and APOE ε4 such that higher physical activity was related to slower APOE ε4-related cognitive decline (estimate 0.007, 95% CI 0.003-0.011, p = 0.001). No significant interaction was found between physical activity and the APOE ε4 allele regarding the change in striatal DAT activities. CONCLUSION Increased physical activity attenuated APOE ε4-related vulnerability to early cognitive decline in patients with PD. This protective effect did not appear to be mediated by striatal dopaminergic function. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov Identifier: NCT01141023. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that increased physical activity was associated with decreased APOE ε4-related early cognitive decline in patients with PD.
Collapse
Affiliation(s)
- Ryul Kim
- From the Department of Neurology (R.K.), Inha University Hospital, Incheon; Department of Neurology (S.P.), School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu; Department of Neurology (D.Y.), Kyung Hee University Hospital; Department of Neurology (J.-S.J.), Kangnam Sacred Heart Hospital, Hallym University College of Medicine; and Department of Neurology (B.J.), College of Medicine, Seoul National University Hospital, Korea
| | - Sangmin Park
- From the Department of Neurology (R.K.), Inha University Hospital, Incheon; Department of Neurology (S.P.), School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu; Department of Neurology (D.Y.), Kyung Hee University Hospital; Department of Neurology (J.-S.J.), Kangnam Sacred Heart Hospital, Hallym University College of Medicine; and Department of Neurology (B.J.), College of Medicine, Seoul National University Hospital, Korea
| | - Dallah Yoo
- From the Department of Neurology (R.K.), Inha University Hospital, Incheon; Department of Neurology (S.P.), School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu; Department of Neurology (D.Y.), Kyung Hee University Hospital; Department of Neurology (J.-S.J.), Kangnam Sacred Heart Hospital, Hallym University College of Medicine; and Department of Neurology (B.J.), College of Medicine, Seoul National University Hospital, Korea
| | - Jin-Sun Jun
- From the Department of Neurology (R.K.), Inha University Hospital, Incheon; Department of Neurology (S.P.), School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu; Department of Neurology (D.Y.), Kyung Hee University Hospital; Department of Neurology (J.-S.J.), Kangnam Sacred Heart Hospital, Hallym University College of Medicine; and Department of Neurology (B.J.), College of Medicine, Seoul National University Hospital, Korea.
| | - Beomseok Jeon
- From the Department of Neurology (R.K.), Inha University Hospital, Incheon; Department of Neurology (S.P.), School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu; Department of Neurology (D.Y.), Kyung Hee University Hospital; Department of Neurology (J.-S.J.), Kangnam Sacred Heart Hospital, Hallym University College of Medicine; and Department of Neurology (B.J.), College of Medicine, Seoul National University Hospital, Korea
| |
Collapse
|
289
|
Bellomo G, Indaco A, Chiasserini D, Maderna E, Paolini Paoletti F, Gaetani L, Paciotti S, Petricciuolo M, Tagliavini F, Giaccone G, Parnetti L, Di Fede G. Machine Learning Driven Profiling of Cerebrospinal Fluid Core Biomarkers in Alzheimer's Disease and Other Neurological Disorders. Front Neurosci 2021; 15:647783. [PMID: 33867925 PMCID: PMC8044304 DOI: 10.3389/fnins.2021.647783] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/08/2021] [Indexed: 12/19/2022] Open
Abstract
Amyloid-beta (Aβ) 42/40 ratio, tau phosphorylated at threonine-181 (p-tau), and total-tau (t-tau) are considered core biomarkers for the diagnosis of Alzheimer’s disease (AD). The use of fully automated biomarker assays has been shown to reduce the intra- and inter-laboratory variability, which is a critical factor when defining cut-off values. The calculation of cut-off values is often influenced by the composition of AD and control groups. Indeed, the clinically defined AD group may include patients affected by other forms of dementia, while the control group is often very heterogeneous due to the inclusion of subjects diagnosed with other neurological diseases (OND). In this context, unsupervised machine learning approaches may overcome these issues providing unbiased cut-off values and data-driven patient stratification according to the sole distribution of biomarkers. In this work, we took advantage of the reproducibility of automated determination of the CSF core AD biomarkers to compare two large cohorts of patients diagnosed with different neurological disorders and enrolled in two centers with established expertise in AD biomarkers. We applied an unsupervised Gaussian mixture model clustering algorithm and found that our large series of patients could be classified in six clusters according to their CSF biomarker profile, some presenting a typical AD-like profile and some a non-AD profile. By considering the frequencies of clinically defined OND and AD subjects in clusters, we subsequently computed cluster-based cut-off values for Aβ42/Aβ40, p-tau, and t-tau. This approach promises to be useful for large-scale biomarker studies aimed at providing efficient biochemical phenotyping of neurological diseases.
Collapse
Affiliation(s)
- Giovanni Bellomo
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Antonio Indaco
- Neurology 5/Neuropathology Unit, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | - Davide Chiasserini
- Section of Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emanuela Maderna
- Neurology 5/Neuropathology Unit, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | | | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Silvia Paciotti
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Maya Petricciuolo
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Fabrizio Tagliavini
- Neurology 5/Neuropathology Unit, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | - Giorgio Giaccone
- Neurology 5/Neuropathology Unit, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | - Lucilla Parnetti
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.,Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giuseppe Di Fede
- Neurology 5/Neuropathology Unit, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| |
Collapse
|
290
|
Paulus A, Engdahl A, Yang Y, Boza-Serrano A, Bachiller S, Torres-Garcia L, Svanbergsson A, Garcia MG, Gouras GK, Li JY, Deierborg T, Klementieva O. Amyloid Structural Changes Studied by Infrared Microspectroscopy in Bigenic Cellular Models of Alzheimer's Disease. Int J Mol Sci 2021; 22:3430. [PMID: 33810433 PMCID: PMC8037084 DOI: 10.3390/ijms22073430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease affects millions of lives worldwide. This terminal disease is characterized by the formation of amyloid aggregates, so-called amyloid oligomers. These oligomers are composed of β-sheet structures, which are believed to be neurotoxic. However, the actual secondary structure that contributes most to neurotoxicity remains unknown. This lack of knowledge is due to the challenging nature of characterizing the secondary structure of amyloids in cells. To overcome this and investigate the molecular changes in proteins directly in cells, we used synchrotron-based infrared microspectroscopy, a label-free and non-destructive technique available for in situ molecular imaging, to detect structural changes in proteins and lipids. Specifically, we evaluated the formation of β-sheet structures in different monogenic and bigenic cellular models of Alzheimer's disease that we generated for this study. We report on the possibility to discern different amyloid signatures directly in cells using infrared microspectroscopy and demonstrate that bigenic (amyloid-β, α-synuclein) and (amyloid-β, Tau) neuron-like cells display changes in β-sheet load. Altogether, our findings support the notion that different molecular mechanisms of amyloid aggregation, as opposed to a common mechanism, are triggered by the specific cellular environment and, therefore, that various mechanisms lead to the development of Alzheimer's disease.
Collapse
Affiliation(s)
- Agnes Paulus
- Medical Microspectroscopy Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (A.P.); (A.E.)
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (Y.Y.); (A.B.-S.); (S.B.); (M.G.G.)
| | - Anders Engdahl
- Medical Microspectroscopy Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (A.P.); (A.E.)
| | - Yiyi Yang
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (Y.Y.); (A.B.-S.); (S.B.); (M.G.G.)
| | - Antonio Boza-Serrano
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (Y.Y.); (A.B.-S.); (S.B.); (M.G.G.)
| | - Sara Bachiller
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (Y.Y.); (A.B.-S.); (S.B.); (M.G.G.)
| | - Laura Torres-Garcia
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (L.T.-G.); (G.K.G.)
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (A.S.); (J.-Y.L.)
| | - Alexander Svanbergsson
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (A.S.); (J.-Y.L.)
| | - Megg G. Garcia
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (Y.Y.); (A.B.-S.); (S.B.); (M.G.G.)
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (L.T.-G.); (G.K.G.)
| | - Gunnar K. Gouras
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (L.T.-G.); (G.K.G.)
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (A.S.); (J.-Y.L.)
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (Y.Y.); (A.B.-S.); (S.B.); (M.G.G.)
| | - Oxana Klementieva
- Medical Microspectroscopy Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (A.P.); (A.E.)
- Lund Institute for Advanced Neutron and X-ray Science (LINXS), 22370 Lund, Sweden
| |
Collapse
|
291
|
Dhakal S, Wyant CE, George HE, Morgan SE, Rangachari V. Prion-like C-Terminal Domain of TDP-43 and α-Synuclein Interact Synergistically to Generate Neurotoxic Hybrid Fibrils. J Mol Biol 2021; 433:166953. [PMID: 33771571 DOI: 10.1016/j.jmb.2021.166953] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 03/04/2021] [Accepted: 03/17/2021] [Indexed: 12/17/2022]
Abstract
Aberrant aggregation and amyloid formation of tar DNA binding protein (TDP-43) and α-synuclein (αS) underlie frontotemporal dementia (FTD) and Parkinson's disease (PD), respectively. Amyloid inclusions of TDP-43 and αS are also commonly co-observed in amyotrophic lateral sclerosis (ALS), dementia with Lewy bodies (DLB) and Alzheimer disease (AD). Emerging evidence from cellular and animal models show colocalization of the TDP-43 and αS aggregates, raising the possibility of direct interactions and co-aggregation between the two proteins. In this report, we set out to answer this question by investigating the interactions between αS and prion-like pathogenic C-terminal domain of TDP-43 (TDP-43 PrLD). PrLD is an aggregation-prone fragment generated both by alternative splicing as well as aberrant proteolytic cleavage of full length TDP-43. Our results indicate that two proteins interact in a synergistic manner to augment each other's aggregation towards hybrid fibrils. While monomers, oligomers and sonicated fibrils of αS seed TDP-43 PrLD monomers, TDP-43 PrLD fibrils failed to seed αS monomers indicating selectivity in interactions. Furthermore, αS modulates liquid droplets formed by TDP-43 PrLD and RNA to promote insoluble amyloid aggregates. Importantly, the cross-seeded hybrid aggregates show greater cytotoxicity as compared to the individual homotypic aggregates suggesting that the interactions between the two proteins have a discernable impact on cellular functions. Together, these results bring forth insights into TDP-43 PrLD - αS interactions that could help explain clinical and pathological presentations in patients with co-morbidities involving the two proteins.
Collapse
Affiliation(s)
- Shailendra Dhakal
- Department of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Courtney E Wyant
- Department of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Hannah E George
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Sarah E Morgan
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Vijayaraghavan Rangachari
- Department of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA; Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA.
| |
Collapse
|
292
|
Croce KR, Yamamoto A. Dissolving the Complex Role Aggregation Plays in Neurodegenerative Disease. Mov Disord 2021; 36:1061-1069. [PMID: 33755257 DOI: 10.1002/mds.28522] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/11/2021] [Accepted: 01/18/2021] [Indexed: 11/10/2022] Open
Abstract
Prominent neuropathological hallmarks of many adult-onset neurodegenerative diseases include the deposition and accumulation of misfolded proteins or conformers; however, their role in pathogenesis has remained unclear. This is in part due to the deceptive simplicity of the question and our limited understanding of how protein homeostasis is maintained in the compartmentalized cells of the central nervous system, especially in the context of the adult brain. Building on studies from simple cell-based systems and invertebrate animals, we recently identified a protein central to the specific and selective turnover of aggregated proteins in the adult brain, the autophagy-linked FYVE protein (Alfy)/Wdfy3. Depletion of Alfy levels in a mouse model of Huntington's disease showed that it accelerated the accumulation of the aggregated mutant huntingtin protein, as well as the onset of behavioral deficits. Although the motor dysfunction was accelerated in the model, this was in the absence of increasing overt cell loss, implicating protein aggregates as a modifier of circuit dysfunction rather than driving degeneration per se. We discuss these findings in the context of what is known about protein accumulation and how we can use proteins such as Alfy to determine if protein accumulation is a shared pathogenic event across different adult-onset diseases. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Katherine R Croce
- Doctoral Program in Pathobiology, Columbia University, New York, New York, USA
| | - Ai Yamamoto
- Departments of Neurology, Pathology and Cell Biology, Columbia University, New York, New York, USA
| |
Collapse
|
293
|
Cong C, Zhang W, Qian X, Qiu W, Ma C. Significant Overlap of α-Synuclein, Amyloid-β, and Phospho-Tau Pathologies in Neuropathological Diagnosis of Lewy-related Pathology: Evidence from China Human Brain Bank. J Alzheimers Dis 2021; 80:447-458. [PMID: 33554920 DOI: 10.3233/jad-201548] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Lewy-related pathology (LRP), primarily comprised of α-synuclein, is a typical neuropathological change that has been identified in many neurodegenerative disorders such as Parkinson's disease (PD), PD with dementia, and dementia with Lewy bodies. OBJECTIVE To investigate the distribution of LRP in the China Human Brain Bank, the co-occurrence of neuropathologic features of Alzheimer's disease (AD) in LRP cases, and LRP-related cognitive dysfunction. METHODS LRP neuropathological diagnosis was performed in 180 postmortem brains. AD neuropathological diagnosis was then performed in the 21 neuropathologically-diagnosed LRP cases. Antemortem cognitive functioning evaluation (Everyday Cognitive, ECog) was assessed for brain donors by the immediate kin of the donor within 24 hours after death. RESULTS 12% (21 in 180) postmortem brains were neuropathologically diagnosed as LRP cases. 86% (18 in 21) aged above 80, 81% (17 in 21) LRP cases combined with AD neuropathology, and 62% (13 in 21) combined with both the intermediate or high-level amyloid-β and phospho-tau pathologies. ECog scores showed significant differences between the groups of LRP brainstem-predominant type and LRP diffuse neocortical type, and between groups of AD and the combined LRP (diffuse neocortical type)-AD. CONCLUSION The overlap of neocortical α-synuclein, amyloid-β, phospho-tau, and neuritic plaques in LRP suggested the potential interplay among the common characteristics of proteinopathies in the late stage of neuropathological development of LRP in human brains. The anatomic progression of LRP, the process of α-synuclein spreading from the brainstem to limbic and neocortical regions, might aggravate the deterioration of cognitive function in addition to that effect of AD.
Collapse
Affiliation(s)
- Cong Cong
- Institute of Basic Medical Sciences, Neuroscience Center, National Human Brain Bank for Development and Function, Chinese Academy of Medical Sciences, Beijing, China.,Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wanying Zhang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,Department of Respiratory Medicine, Capital Medical University, Beijing, China
| | - Xiaojing Qian
- Institute of Basic Medical Sciences, Neuroscience Center, National Human Brain Bank for Development and Function, Chinese Academy of Medical Sciences, Beijing, China.,Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wenying Qiu
- Institute of Basic Medical Sciences, Neuroscience Center, National Human Brain Bank for Development and Function, Chinese Academy of Medical Sciences, Beijing, China.,Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Chao Ma
- Institute of Basic Medical Sciences, Neuroscience Center, National Human Brain Bank for Development and Function, Chinese Academy of Medical Sciences, Beijing, China.,Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Peking Union Medical College, Beijing, China.,Chinese Institute for Brain Research, Beijing, China
| |
Collapse
|
294
|
Dong MX, Wei YD, Hu L. Lipid metabolic dysregulation is involved in Parkinson's disease dementia. Metab Brain Dis 2021; 36:463-470. [PMID: 33433787 DOI: 10.1007/s11011-020-00665-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/25/2020] [Indexed: 12/13/2022]
Abstract
Dementia is very common in the late stage of patient with Parkinson's disease (PD). We aim to explore its underlying pathogenesis and identify candidate biomarkers using untargeted metabolomics analysis. Consecutive PD patients and healthy controls were recruited. Clinical data were assessed and patients were categorized into Parkinson's disease without dementia (PDND) and Parkinson's disease dementia (PDD). Fast plasma samples were obtained and untargeted liquid chromatography-mass spectrometry-based metabolomics analysis was performed. Based on the identified differentially-expressed metabolites from the metabolomics analysis, multivariate linear regression analyses and receiver operating characteristic (ROC) curves were further employed. According to the clinical data, the mean ages of PDND and PDD patients were significantly higher than those of healthy controls. The incidence of hypercholesterolemia was decreased in PDD patients. PDD patients also had lower levels of triglyceride, low-density lipoprotein cholesterol, and apolipoprotein B. There were 24 and 57 differentially expressed metabolites in PDD patients when compared with the healthy controls and PDND patients from the metabolomics analysis. Eleven lipid metabolites were simultaneously decreased between these two groups, and can be further subcategorized into fatty acyls, glycerolipids, glycerophospholipids, sphingolipids, and prenol lipids. The plasma levels of the eleven metabolites were positively correlated with MMSE score and can be candidate biomarkers for PDD patients with areas under the curve ranging from 0.724 to 0.806 based on the ROC curves. Plasma lipoproteins are significantly lower in PDD patients. A panel of eleven lipid metabolites were also decreased and can be candidate biomarkers for the diagnosis of PDD patients. Lipid metabolic dysregulation is involved in the pathogenesis of Parkinson's disease dementia.
Collapse
Affiliation(s)
- Mei-Xue Dong
- Department of Neurology, Renmin Hospital of Wuhan University, Hubei General Hospital, No. 99 Zhangzhidong Road, Wuchang District, Wuhan, Hubei, China
| | - You-Dong Wei
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Hu
- Department of Neurology, Renmin Hospital of Wuhan University, Hubei General Hospital, No. 99 Zhangzhidong Road, Wuchang District, Wuhan, Hubei, China.
| |
Collapse
|
295
|
Li D, Liu C. Hierarchical chemical determination of amyloid polymorphs in neurodegenerative disease. Nat Chem Biol 2021; 17:237-245. [PMID: 33432239 DOI: 10.1038/s41589-020-00708-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 11/10/2020] [Indexed: 01/28/2023]
Abstract
Amyloid aggregation, which disrupts protein homeostasis, is a common pathological event occurring in human neurodegenerative diseases (NDs). Numerous evidences have shown that the structural diversity, so-called polymorphism, is decisive to the amyloid pathology and is closely associated with the onset, progression, and phenotype of ND. But how could one protein form so many stable structures? Recently, atomic structural evidence has been rapidly mounting to depict the involvement of chemical modifications in the amyloid fibril formation. In this Perspective, we aim to present a hierarchical regulation of chemical modifications including covalent post-translational modifications (PTMs) and noncovalent cofactor binding in governing the polymorphic amyloid formation, based mainly on the latest α-synuclein and Tau fibril structures. We hope to emphasize the determinant role of chemical modifications in amyloid assembly and pathology and to evoke chemical biological approaches to lead the fundamental and therapeutic research on protein amyloid state and the associated NDs.
Collapse
Affiliation(s)
- Dan Li
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China. .,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
296
|
Mani I. CRISPR-Cas9 for treating hereditary diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 181:165-183. [PMID: 34127193 DOI: 10.1016/bs.pmbts.2021.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
This chapter analyzes to use of the genome editing tool to the treatment of various genetic diseases. The genome editing method could be used to change the DNA in cells or organisms to understand their physiological response. Therefore, a key objective is to present general information about the use of the genome editing tool in a pertinent way. An emerging genome editing technology like a clustered regularly short palindromic repeats (CRISPR) is an extensively expended in biological sciences. CRISPR and CRISPR-associated protein 9 (CRISPR-Cas9) technique is being utilized to edit any DNA mutations associated with hereditary diseases to study in cells (in vitro) and animals (in vivo). Interestingly, CRISPR-Cas9 could be used to the investigation of treatments of various human hereditary diseases such as hemophila, β-thalassemia, cystic fibrosis, Alzheimer's, Huntington's, Parkinson's, tyrosinemia, Duchnene muscular dystrophy, Tay-Sachs, and fragile X syndrome disorders. Furthermore, CRISPR-Cas9 could also be used in other diseases to the improvement of human health. Finally, this chapter discuss current progress to treatment for hereditary diseases using CRISPR-Cas9 technology and highlights associated challenges and future prospects.
Collapse
Affiliation(s)
- Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| |
Collapse
|
297
|
CD33 rs3865444 as a risk factor for Parkinson's disease. Neurosci Lett 2021; 748:135709. [PMID: 33582190 DOI: 10.1016/j.neulet.2021.135709] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Alzheimer's (AD) and Parkinson's diseases (PD) share a few elements of their clinical, pathological and genetic backgrounds. The CD33 rs3865444 has emerged as a strong genetic locus associated with AD through genome-wide association study (GWAS). However, little is known for its role in PD. OBJECTIVE To assess the role of CD33 rs3865444 on PD risk. METHODS We genotyped 358 patients with PD and 358 healthy controls for theCD33 rs3865444. Odds ratios (ORs) with the respective 95% confidence intervals (CIs)], were calculated with the SNPStats software, assuming five genetic models (co-dominant, dominant, recessive, over-dominant and log-additive), with the G allele as the reference allele. RESULTS The CD33 rs3865444 was associated with decreased PD risk in the dominant [GG vs GT + TT; OR (95% CI) = 0.61 (0.45-0.82), p = 0.001], the over-dominant [GG + TT vs GT; OR (95% CI) = 0.65 (0.48-0.89), p = 0.0061], log-additive [OR (95% CI) = 0.67 (0.52-0.86), p = 0.0014], and co-dominant [with overall p = 0.0043, and OR (95% CI) = 0.62 (0.45-0.84) for the TG genotype compared to the GG], modes of inheritance. CONCLUSIONS The CD33 rs3865444 is associated with decreased PD risk, and larger studies investigating the role of CD33 rs3865444 on PD are needed.
Collapse
|
298
|
Wang X, Ding D, Wu L, Jiang T, Wu C, Ge Y, Guo X. PHB blocks endoplasmic reticulum stress and apoptosis induced by MPTP/MPP + in PD models. J Chem Neuroanat 2021; 113:101922. [PMID: 33581266 DOI: 10.1016/j.jchemneu.2021.101922] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 01/23/2021] [Accepted: 01/24/2021] [Indexed: 01/27/2023]
Abstract
Ample empirical evidence suggests that mitochondrial dysfunction and endoplasmic reticulum (ER) stress play a crucial role in the pathogenesis of Parkinson's disease (PD). Prohibitin (PHB), a mitochondrial inner-membrane protein involved in mitochondrial homeostasis and function, may be involved in the pathogenesis of PD. We investigated the functional role of PHB in mitochondrial biogenesis and ER stress in methyl-4-phenylpyridinium (MPP +)-induced in vivo and in vitro models of PD. The overexpression of PHB in SH-SY5Y cells block ed cell death and the apoptosis induced by MPP + incubation. PHB also block ed the activation of ER stress markers, including glucose-regulated protein 78, while increasing the expression of Xbox- binding protein 1 and caspase-12. Moreover, the intracerebroventricular administration of the PHB overexpression vector greatly block ed motor dysfunction and the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-mediated neurodegeneration in the mouse model of PD. The production of reactive oxygen species, ER stress, and autophagic stress induced by MPTP were also significantly block ed in PD mice overexpressing PHB. Our results suggest that PHB blocks the dopaminergic-neuron depletion by preserving mitochondrial function and inhibiting ER stress. The genetic manipulation of PHB may feature potential as a treatment for PD.
Collapse
Affiliation(s)
- Xiaohong Wang
- School of Medicine, Yangzhou University, Yangzhou 225001, China; Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA ResearchNoncoding RNA Center, Yangzhou University, YangZhou 225001, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Dongyi Ding
- School of Medicine, Yangzhou University, Yangzhou 225001, China; Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA ResearchNoncoding RNA Center, Yangzhou University, YangZhou 225001, China
| | - Lei Wu
- School of Medicine, Yangzhou University, Yangzhou 225001, China
| | - Tianlin Jiang
- School of Medicine, Yangzhou University, Yangzhou 225001, China
| | - Chenghao Wu
- School of Medicine, Yangzhou University, Yangzhou 225001, China
| | - Yue Ge
- School of Medicine, Yangzhou University, Yangzhou 225001, China
| | | |
Collapse
|
299
|
Compta Y, Revesz T. Neuropathological and Biomarker Findings in Parkinson's Disease and Alzheimer's Disease: From Protein Aggregates to Synaptic Dysfunction. JOURNAL OF PARKINSONS DISEASE 2021; 11:107-121. [PMID: 33325398 PMCID: PMC7990431 DOI: 10.3233/jpd-202323] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There is mounting evidence that Parkinson’s disease (PD) and Alzheimer’s disease (AD) share neuropathological hallmarks, while similar types of biomarkers are being applied to both. In this review we aimed to explore similarities and differences between PD and AD at both the neuropathology and the biomarker levels, specifically focusing on protein aggregates and synapse dysfunction. Thus, amyloid-β peptide (Aβ) and tau lesions of the Alzheimer-type are common in PD and α-synuclein Lewy-type aggregates are frequent findings in AD. Modern neuropathological techniques adding to routine immunohistochemistry might take further our knowledge of these diseases beyond protein aggregates and down to their presynaptic and postsynaptic terminals, with potential mechanistic and even future therapeutic implications. Translation of neuropathological discoveries to the clinic remains challenging. Cerebrospinal fluid (CSF) and positron emission tomography (PET) markers of Aβ and tau have been shown to be reliable for AD diagnosis. Conversely, CSF markers of α-synuclein have not been that consistent. In terms of PET markers, there is no PET probe available for α-synuclein yet, while the AD PET markers range from consistent evidence of their specificity (amyloid imaging) to greater uncertainty of their reliability due to off-target binding (tau imaging). CSF synaptic markers are attractive, still needing more evidence, which currently suggests those might be non-specific markers of disease progression. It can be summarized that there is neuropathological evidence that protein aggregates of AD and PD are present both at the soma and the synapse. Thus, a number of CSF and PET biomarkers beyond α-synuclein, tau and Aβ might capture these different faces of protein-related neurodegeneration. It remains to be seen what the longitudinal outcomes and the potential value as surrogate markers of these biomarkers are.
Collapse
Affiliation(s)
- Yaroslau Compta
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic / IDIBAPS / CIBERNED, Barcelona, Catalonia, Spain.,Institut de Neurociències, Maextu's excellence center, University of Barcelona, Barcelona, Catalonia, Spain
| | - Tamas Revesz
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, UK.,Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, UK.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK
| |
Collapse
|
300
|
Castonguay AM, Gravel C, Lévesque M. Treating Parkinson's Disease with Antibodies: Previous Studies and Future Directions. JOURNAL OF PARKINSONS DISEASE 2021; 11:71-92. [PMID: 33104039 PMCID: PMC7990466 DOI: 10.3233/jpd-202221] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Parkinson's disease is a neurodegenerative disorder mainly characterized by the degeneration of dopaminergic neurons in the substantia nigra. Degenerating neurons contain abnormal aggregates called Lewy bodies, that are predominantly composed of the misfolded and/or mutated alpha-synuclein protein. Post-translational modifications, cellular stress, inflammation and gene mutations are thought to trigger its pathological misfolding and aggregation. With alpha-synuclein pathology being strongly associated with dopaminergic neuronal toxicity, strategies aimed to reduce its burden are expected to be beneficial in slowing disease progression. Moreover, multiple sources of evidence suggest a cell-to-cell transmission of pathological alpha-synuclein in a prion-like manner. Therefore, antibodies targeting extra- or intracellular alpha-synuclein could be efficient in limiting the aggregation and transmission. Several active and passive immunization strategies have been explored to target alpha-synuclein. Here, we summarize immunotherapeutic approaches that were tested in pre-clinical or clinical studies in the last two decades in an attempt to treat Parkinson's disease.
Collapse
Affiliation(s)
- Anne-Marie Castonguay
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.,CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, QC, Canada
| | - Claude Gravel
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.,CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, QC, Canada
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.,CERVO Brain Research Centre, 2601, chemin de la Canardière, Québec, QC, Canada
| |
Collapse
|