501
|
Zhou X, Zhao P, Guo X, Wang J, Wang R. Effectiveness of aerobic and resistance training on the motor symptoms in Parkinson's disease: Systematic review and network meta-analysis. Front Aging Neurosci 2022; 14:935176. [PMID: 35978948 PMCID: PMC9376630 DOI: 10.3389/fnagi.2022.935176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background/objectives Aerobic and resistance training are common complementary therapies to improve motor symptoms in people with Parkinson's disease (PD), and there is still a lack of advice on which intensity and period of aerobic or resistance training is more appropriate for people with PD. Therefore, a network meta-analysis was conducted to assess the comparative efficacy of aerobic and resistance training of different intensities and cycles on motor symptoms in patients with Parkinson's disease. Methods Based on several biomedical databases, a search strategy system was conducted to retrieve randomized controlled trials (RCTs) without language restrictions. A network meta-analysis with a frequentist approach was conducted to estimate the efficacy and probability rankings of aerobic and resistance training on Parkinson's patients. What's more, a range of analyses and assessments, such as routine meta-analyses and risk of bias, were performed as well. Results Twenty trials with 719 patients evaluating 18 different therapies were identified. Through the Unified Parkinson's Disease Motor Rating Scale, (UPDRS III); 6-minute walk test, (6MWT); 10-meter walk test, (TWM); and time up and go (TUG) and Quality of Life Scale-39 (PDQ-39), to explore the effects of different intensity resistance and aerobic exercise on PD. As a result, short period high intensity resistance movement (standard mean difference (SMD) = -0.95, 95% confidence interval (CI) -1.68 to -0.22) had significantly decreased the Unified Parkinson's Disease Motor Rating Scale (UPDRS III). Short period high intensity resistance exercise showed similar superiority in other indices; also, aerobic and resistance training of different cycle intensities produced some efficacy in PD patients, both in direct and indirect comparisons. Conclusion For patients with moderate to mild Parkinson's symptoms, short periods high intensity resistance training may provide complementary therapy for PD, and aerobic or resistance training of varying intensity and periodicity may be recommended as exercise prescription for PD patients. However, more large scale and high quality clinical trials are needed to confirm the effectiveness of this exercise therapy in the future. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/, identifier: CRD42022324824.
Collapse
Affiliation(s)
- Xiao Zhou
- Sports Rehabilitation Research Center, China Institute of Sport Science, Beijing, China
| | - Peng Zhao
- Sports Rehabilitation Research Center, China Institute of Sport Science, Beijing, China
| | - Xuanhui Guo
- College of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Jialin Wang
- Sports Rehabilitation Research Center, China Institute of Sport Science, Beijing, China
| | - Ruirui Wang
- Sports Rehabilitation Research Center, China Institute of Sport Science, Beijing, China
| |
Collapse
|
502
|
Chen T, Edwards TS, Hinson VK, Soler ZM. Prevalence of Rhinorrhea in Parkinson Disease: A Systematic Review and Meta-analysis. Neurol Clin Pract 2022; 12:e75-e81. [PMID: 36382119 PMCID: PMC9647823 DOI: 10.1212/cpj.0000000000001180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 03/24/2022] [Indexed: 11/15/2022]
Abstract
Purpose of Review To perform a systematic review and determine the prevalence of rhinorrhea in Parkinson disease (PD). Recent Findings Of 451 patients with PD and 233 controls, the pooled prevalence of rhinorrhea in patients with PD was 45.0% (95% confidence interval 33.94-56.40), which was significantly greater than that in controls (p < 0.001). The prevalence of self-reported olfactory dysfunction was greater in patients with PD; however, a pooled analysis of studies using objective scores showed no difference. The mean age of patients with PD was greater than that of controls (p = 0.002). The mean age of patients with PD with rhinorrhea was also greater than that of patients with PD without rhinorrhea (p < 0.001), but disease characteristics did not differ. Summary There is a high prevalence of rhinorrhea in patients with PD; therefore, providers should query for rhinorrhea during visits and understand the treatment options available. Future studies should explore the pathophysiology of rhinorrhea in PD and the relationship between rhinorrhea and disease severity and duration, as well as determine whether treatment-specific outcomes differ in patients with PD.
Collapse
Affiliation(s)
- Tiffany Chen
- Department of Otolaryngology-Head and Neck Surgery (TC, TSE, ZMS), Medical University of South Carolina; and Department of Neurology (VKH), Medical University of South Carolina, Charleston
| | - Thomas S Edwards
- Department of Otolaryngology-Head and Neck Surgery (TC, TSE, ZMS), Medical University of South Carolina; and Department of Neurology (VKH), Medical University of South Carolina, Charleston
| | - Vanessa K Hinson
- Department of Otolaryngology-Head and Neck Surgery (TC, TSE, ZMS), Medical University of South Carolina; and Department of Neurology (VKH), Medical University of South Carolina, Charleston
| | - Zachary M Soler
- Department of Otolaryngology-Head and Neck Surgery (TC, TSE, ZMS), Medical University of South Carolina; and Department of Neurology (VKH), Medical University of South Carolina, Charleston
| |
Collapse
|
503
|
He C, Rong S, Zhang P, Li R, Li X, Li Y, Wang L, Zhang Y. Metabolite changes in prefrontal lobes and the anterior cingulate cortex correlate with processing speed and executive function in Parkinson disease patients. Quant Imaging Med Surg 2022; 12:4226-4238. [PMID: 35919059 PMCID: PMC9338382 DOI: 10.21037/qims-21-1126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 05/23/2022] [Indexed: 11/06/2022]
Abstract
Background Processing speed and executive function can be impaired in patients with Parkinson disease (PD). However, the neural factors related to the slowdown in processing speed and dysexecutive function in PD are not completely understood. The objective of this study is to investigate the metabolic changes of the frontal and anterior cingulate cortex (ACC) through the use of 1H magnetic resonance spectroscopy and to explore the association between cognitive function and metabolic ratios. Methods In this retrospective case-control study, we conducted neuropsychological assessments of executive function and information processing speed in healthy controls (HCs) and in patients with PD. Chemical information was obtained for the of N-acetyl-aspartate (NAA):creatine (Cr) ratio and the choline-containing compounds (Cho):Cr ratio within the bilateral prefrontal cortex and ACC. Using hierarchical multiple regression analysis, we analyzed the relationship between cognitive function and metabolic ratios in the bilateral prefrontal lobe and ACC in patients with PD. Results In all, 59 patients with PD and 30 HCs were recruited. Patients with PD showed worse performance in executive function and processing speed compared with HCs (P<0.001). In patients with PD, the Cho:Cr ratios in the ACC (Z=2.20, P=0.028) and the right prefrontal cortex (t=2.16, P=0.034) were significantly increased. The hierarchical multiple regressions in patients with PD showed that the NAA:Cr ratio in the ACC correlated with the Stroop A completion times (P<0.05) and that the NAA:Cr ratio of the right prefrontal cortex correlated with the scores of the Wechsler Adult Intelligence Scale (WAIS)-Digit symbol test (P<0.05). Conclusions Information processing speed and executive function are impaired in patients with PD. Neuronal integrity and membrane turnover in the ACC and the right prefrontal cortex may be important factors in the slowdown of the information processing speed in patients with PD.
Collapse
Affiliation(s)
- Chentao He
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Siming Rong
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Piao Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ruitao Li
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaohong Li
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yan Li
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lijuan Wang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuhu Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
504
|
Guo M, Ji X, Liu J. Hypoxia and Alpha-Synuclein: Inextricable Link Underlying the Pathologic Progression of Parkinson's Disease. Front Aging Neurosci 2022; 14:919343. [PMID: 35959288 PMCID: PMC9360429 DOI: 10.3389/fnagi.2022.919343] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease, with typical motor symptoms as the main clinical manifestations. At present, there are about 10 million patients with PD in the world, and its comorbidities and complications are numerous and incurable. Therefore, it is particularly important to explore the pathogenesis of PD and find possible therapeutic targets. Because the etiology of PD is complex, involving genes, environment, and aging, finding common factors is the key to identifying intervention targets. Hypoxia is ubiquitous in the natural environment and disease states, and it is considered to be closely related to the etiology of PD. Despite research showing that hypoxia increases the expression and aggregation of alpha-synuclein (α-syn), the most important pathogenic protein, there is still a lack of systematic studies on the role of hypoxia in α-syn pathology and PD pathogenesis. Considering that hypoxia is inextricably linked with various causes of PD, hypoxia may be a co-participant in many aspects of the PD pathologic process. In this review, we describe the risk factors for PD, and we discuss the possible role of hypoxia in inducing PD pathology by these risk factors. Furthermore, we attribute the pathological changes caused by PD etiology to oxygen uptake disorder and oxygen utilization disorder, thus emphasizing the possibility of hypoxia as a critical link in initiating or promoting α-syn pathology and PD pathogenesis. Our study provides novel insight for exploring the pathogenesis and therapeutic targets of PD.
Collapse
Affiliation(s)
- Mengyuan Guo
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Xunming Ji
| | - Jia Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
- *Correspondence: Jia Liu
| |
Collapse
|
505
|
Liu XL, Liu SY, Barret O, Tamagnan GD, Qiao HW, Song TB, Lu J, Chan P. Diagnostic value of striatal 18F-FP-DTBZ PET in Parkinson’s disease. Front Aging Neurosci 2022; 14:931015. [PMID: 35936768 PMCID: PMC9355024 DOI: 10.3389/fnagi.2022.931015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/30/2022] [Indexed: 12/25/2022] Open
Abstract
Background18F-FP-DTBZ has been proven as a biomarker for quantifying the concentration of presynaptic vesicular monoamine transporter 2 (VMAT2). However, its clinical application is still limited.ObjectivesTo evaluate the difference in dopaminergic integrity between patients with Parkinson’s disease (PD) and healthy controls (HC) using 18F-FP-DTBZ PET in vivo and to determine the diagnostic value of standardized uptake value ratios (SUVRs) using the Receiver Operating Characteristic (ROC) curve.MethodsA total of 34 PD and 31 HC participants were enrolled in the PET/MR derivation cohort, while 89 PD and 18 HC participants were recruited in the PET/CT validation cohort. The Hoehn–Yahr Scale and the third part of the MDS-Unified Parkinson’s Disease Rating Scale (MDSUPDRS-III) were used to evaluate the disease staging and severity. All assessments and PET scanning were performed in drug-off states. The striatum was segmented into five subregions as follows: caudate, anterior dorsal putamen (ADP), anterior ventral putamen (AVP), posterior dorsal putamen (PDP), and posterior ventral putamen (PVP) using automatic pipeline built with the PMOD software (version 4.105). The SUVRs of the targeted subregions were calculated using the bilateral occipital cortex as the reference region.ResultsRegarding the diagnostic value, ROC curve and blind validation showed that the contralateral PDP (SUVR = 3.43) had the best diagnostic accuracy (AUC = 0.973; P < 0.05), with a sensitivity of 97.1% (95% CI: 82.9–99.8%), specificity of 100% (95% CI: 86.3–100%), positive predictive value (PPV) of 100% (95% CI: 87.0–100%), negative predictive value (NPV) of 96.9% (95% CI: 82.0–99.8%), and an accuracy of 98.5% for the diagnosis of PD in the derivation cohort. Blind validation of 18F-FP-DTBZ PET imaging diagnosis was done using the PET/CT cohort, where participants with a SUVR of the PDP <3.43 were defined as PD. Kappa test showed a consistency of 0.933 (P < 0.05) between clinical diagnosis and imaging diagnosis, with a sensitivity of 98.9% (95% CI: 93.0–99.9%), specificity of 94.4% (95% CI: 70.6–99.7%), PPV of 98.9% (95% CI: 93.0–99.9%), NPV of 94.4% (95% CI: 70.6–99.7%), and a diagnostic accuracy of 98.1%.ConclusionsOur results showed that an SUVR threshold of 3.43 in the PDP could effectively distinguish patients with PD from HC.
Collapse
Affiliation(s)
- Xiu-Lin Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shu-Ying Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research (CIBR), Beijing, China
- Shu-Ying Liu,
| | - Olivier Barret
- CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Gilles D. Tamagnan
- Mental Health PET Radioligand Development (MHPRD) Program, Yale University, New Haven, CT, United States
| | - Hong-Wen Qiao
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tian-Bin Song
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Piu Chan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Beijing, China
- *Correspondence: Piu Chan,
| |
Collapse
|
506
|
Assessment of Vestibular-Evoked Myogenic Potentials in Parkinson’s Disease: A Systematic Review and Meta-Analysis. Brain Sci 2022; 12:brainsci12070956. [PMID: 35884762 PMCID: PMC9313211 DOI: 10.3390/brainsci12070956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/03/2022] [Accepted: 07/19/2022] [Indexed: 02/06/2023] Open
Abstract
(1) Background: The brainstem plays an essential role in the early stage of Parkinson’s disease (PD), but it is not widely tested in clinical examinations of PD. Vestibular-evoked myogenic potentials (VEMPs) are recognized as fundamental tools in the assessment of brainstem function. The aim of our meta-analysis was to assess the abnormal findings of VEMPs in patients with PD. (2) Methods: Up to 14 February 2022, PubMed, Embase, and Web of Science were searched to evaluate VEMPs in patients with PD in comparison with respective controls. The study protocol was registered at PROSPERO (CRD42022311103). (3) Results: A total of 15 studies were finally included in our meta-analysis. The absence rates of VEMPs in patients with PD were significantly higher than those of control groups (cVEMP: OR = 6.77; oVEMP: OR = 13.9; mVEMP: OR = 7.52). A delayed P13 latency, a decreased peak-to-peak amplitude, and an increased AAR of cVEMP, and a delayed oVEMP P15 latency were also found in patients with PD. (4) Conclusions: Our meta-analysis indicates abnormal VEMP findings in patients with PD, revealing the dysfunction of the brainstem in PD. VEMP tests, especially cVEMP tests, could be a helpful method for the early detection of PD.
Collapse
|
507
|
Minakawa EN. Bidirectional Relationship Between Sleep Disturbances and Parkinson's Disease. Front Neurol 2022; 13:927994. [PMID: 35923835 PMCID: PMC9342689 DOI: 10.3389/fneur.2022.927994] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/15/2022] [Indexed: 12/01/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease (AD). Both diseases share common clinical and pathological features: the gradual progression of neurological and psychiatric symptoms caused by neuronal dysfunction and neuronal cell death due to the accumulation of misfolded and neurotoxic proteins. Furthermore, both of them are multifactorial diseases in which both genetic and non-genetic factors contribute to the disease course. Non-genetic factors are of particular interest for the development of preventive and therapeutic approaches for these diseases because they are modifiable; of these, sleep is a particularly intriguing factor. Sleep disturbances are highly prevalent among both patients with AD and PD. To date, research has suggested that sleep disturbances are a consequence as well as a risk factor for the onset and progression of AD, which implies a bidirectional relationship between sleep and AD. Whether such a relationship exists in PD is less certain, albeit highly plausible given the shared pathomechanisms. This review examines the current evidence for the bidirectional relationship between sleep and PD. It includes research in both humans and animal models, followed by a discussion of the current understanding of the mechanisms underlying this relationship. Finally, potential avenues of research toward achieving disease modification to treat or prevent PD are proposed. Although further efforts are crucial for preventing the onset and slowing the progress of PD, it is evident that sleep is a valuable candidate target for future interventions to improve the outcomes of PD patients.
Collapse
Affiliation(s)
- Eiko N. Minakawa
- Department of Neurophysiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
- Parkinson Disease and Movement Disorder Center, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Japan
- Sleep Disorder Center, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Japan
- Research Center for Neurocognitive Disorders, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Japan
- *Correspondence: Eiko N. Minakawa
| |
Collapse
|
508
|
Villar-Conde S, Astillero-Lopez V, Gonzalez-Rodriguez M, Saiz-Sanchez D, Ubeda-Banon I, Flores-Cuadrado A, Martinez-Marcos A. Neuronal and glial characterization in the rostrocaudal axis of the human anterior olfactory nucleus: Involvement in Parkinson’s disease. Front Neuroanat 2022; 16:907373. [PMID: 35923614 PMCID: PMC9339613 DOI: 10.3389/fnana.2022.907373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/27/2022] [Indexed: 12/02/2022] Open
Abstract
Hyposmia is one of the prodromal symptoms of Parkinson’s disease (PD) and a red flag in clinical diagnosis. Neuropathologically, this sign correlates with α-synuclein involvement in the anterior olfactory nucleus (AON). Neurodegeneration, microgliosis, and astrogliosis in AON are poorly studied, and bulbar AON is the focus of these studies with contradictory results. Additionally, male sex is a risk marker for developing PD, but sexual dimorphism of neural and glial populations in the AON has rarely been considered. The aim of this study was to analyze the density of NeuN, Iba-1, GFAP, and Lewy bodies (LBs), as well as the relationship of these cell type markers with pathology along the rostrocaudal axis of the AON (bulbar, retrobulbar, cortical anterior, and posterior divisions). Cavalieri, optical fractionator, and area fraction fractionator stereological approaches were used for the volume, cell populations and LBs densities, area fraction, and percentage of overlap. Iba-1 and α-syn intensities were measured using ImageJ. In non-PD (NPD) cases, the volume was lower in the AON at the extremes of the rostrocaudal axis than in the intermediate divisions. Cortical anterior AON volume decreased in PD compared with NPD cases. NeuN density decreased rostrocaudally in AON portions in NPD and PD cases. This occurred similarly in Iba-1 but only in PD samples. Iba-1 intensity significantly increased in bulbar AON between PD and NPD. No changes were found in astrocytes. Eight percent of NeuN, 0.1% of Iba-1, and 0.1% of GFAP areas overlapped with LBs area along the AON portions. The data indicate that bulbar AON, which is the most rostral portion in this axis, could play a major role in the pathology. This could be related to the larger area occupied by LBs in these divisions.
Collapse
|
509
|
Peng L, Zhang Z, Chen X, Gao X. Alternation of the Rich-Club Organization of Individual Brain Metabolic Networks in Parkinson's Disease. Front Aging Neurosci 2022; 14:964874. [PMID: 35875793 PMCID: PMC9304954 DOI: 10.3389/fnagi.2022.964874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 06/23/2022] [Indexed: 11/18/2022] Open
Abstract
Objective The diagnosis of Parkinson's disease (PD) remains challenging. Although 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) has revealed the metabolic abnormalities associated with PD at systemic levels, the underlying rich-club organization of the metabolic connectome in these patients remains largely unknown. Materials and Methods The data of 49 PD patients and 49 well-matched healthy controls (HCs) were retrieved and assessed. An individual metabolic connectome based on the standard uptake value (SUV) was built using the Jensen-Shannon Divergence Similarity Estimation (JSSE) method to compare the rich-club properties between PD patients and HC. Results Our results showed the rich-club organization of metabolic networks (normalized rich-club coefficients > 1) in the PD and HC group were within a range of thresholds. Further, patients with PD demonstrated lower strength and degree in rich-club connections compared with HCs (strength: HCs = 55.70 ± 8.52, PDs = 52.03 ± 10.49, p = 0.028; degree: HCs = 56.55 ± 8.60, PDs = 52.85 ± 10.62, p = 0.029), but difference between their feeder and local connections was not significant. Conclusion Individual metabolic networks combined with rich club analysis indicated that PD patients had decreased rich club connections but similar feeder and local connections compared with HCs, indicating rich club connections as a promising marker for early diagnosis of PD.
Collapse
Affiliation(s)
- Liling Peng
- Shanghai Universal Medical Imaging Diagnostic Center, Shanghai, China
| | - Zhimin Zhang
- Department of Blood Transfusion, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaofeng Chen
- College of Mathematics and Statistics, Chongqing Jiaotong University, Chongqing, China
| | - Xin Gao
- Shanghai Universal Medical Imaging Diagnostic Center, Shanghai, China
| |
Collapse
|
510
|
Irisin, An Exercise-induced Bioactive Peptide Beneficial for Health Promotion During Aging Process. Ageing Res Rev 2022; 80:101680. [DOI: 10.1016/j.arr.2022.101680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 12/11/2022]
|
511
|
Zhu M, Liu X, Ye Y, Yan X, Cheng Y, Zhao L, Chen F, Ling Z. Gut Microbiota: A Novel Therapeutic Target for Parkinson’s Disease. Front Immunol 2022; 13:937555. [PMID: 35812394 PMCID: PMC9263276 DOI: 10.3389/fimmu.2022.937555] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/26/2022] [Indexed: 12/16/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease characterized by motor dysfunction. Growing evidence has demonstrated that gut dysbiosis is involved in the occurrence, development and progression of PD. Numerous clinical trials have identified the characteristics of the changed gut microbiota profiles, and preclinical studies in PD animal models have indicated that gut dysbiosis can influence the progression and onset of PD via increasing intestinal permeability, aggravating neuroinflammation, aggregating abnormal levels of α-synuclein fibrils, increasing oxidative stress, and decreasing neurotransmitter production. The gut microbiota can be considered promising diagnostic and therapeutic targets for PD, which can be regulated by probiotics, psychobiotics, prebiotics, synbiotics, postbiotics, fecal microbiota transplantation, diet modifications, and Chinese medicine. This review summarizes the recent studies in PD-associated gut microbiota profiles and functions, the potential roles, and mechanisms of gut microbiota in PD, and gut microbiota-targeted interventions for PD. Deciphering the underlying roles and mechanisms of the PD-associated gut microbiota will help interpret the pathogenesis of PD from new perspectives and elucidate novel therapeutic strategies for PD.
Collapse
Affiliation(s)
- Manlian Zhu
- Department of Geriatrics, Lishui Second People’s Hospital, Lishui, China
| | - Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yiru Ye
- Department of Respiratory Medicine, Lishui Central Hospital, Lishui, China
| | - Xiumei Yan
- Department of Laboratory Medicine, Lishui Second People’s Hospital, Lishui, China
| | - Yiwen Cheng
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Longyou Zhao
- Department of Laboratory Medicine, Lishui Second People’s Hospital, Lishui, China
| | - Feng Chen
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Zongxin Ling, ; ; Feng Chen,
| | - Zongxin Ling
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Zongxin Ling, ; ; Feng Chen,
| |
Collapse
|
512
|
Pérez-Carrión MD, Posadas I, Solera J, Ceña V. LRRK2 and Proteostasis in Parkinson's Disease. Int J Mol Sci 2022; 23:6808. [PMID: 35743250 PMCID: PMC9224256 DOI: 10.3390/ijms23126808] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 02/01/2023] Open
Abstract
Parkinson's disease is a neurodegenerative condition initially characterized by the presence of tremor, muscle stiffness and impaired balance, with the deposition of insoluble protein aggregates in Lewy's Bodies the histopathological hallmark of the disease. Although different gene variants are linked to Parkinson disease, mutations in the Leucine-Rich Repeat Kinase 2 (LRRK2) gene are one of the most frequent causes of Parkinson's disease related to genetic mutations. LRRK2 toxicity has been mainly explained by an increase in kinase activity, but alternative mechanisms have emerged as underlying causes for Parkinson's disease, such as the imbalance in LRRK2 homeostasis and the involvement of LRRK2 in aggregation and spreading of α-synuclein toxicity. In this review, we recapitulate the main LRRK2 pathological mutations that contribute to Parkinson's disease and the different cellular and therapeutic strategies devised to correct LRRK2 homeostasis. In this review, we describe the main cellular control mechanisms that regulate LRRK2 folding and aggregation, such as the chaperone network and the protein-clearing pathways such as the ubiquitin-proteasome system and the autophagic-lysosomal pathway. We will also address the more relevant strategies to modulate neurodegeneration in Parkinson's disease through the regulation of LRRK2, using small molecules or LRRK2 silencing.
Collapse
Affiliation(s)
- María Dolores Pérez-Carrión
- Unidad Asociada Neurodeath, Universidad de Castilla-La Mancha, 02006 Albacete, Spain; (M.D.P.-C.); (I.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Consorcio CIBER, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Inmaculada Posadas
- Unidad Asociada Neurodeath, Universidad de Castilla-La Mancha, 02006 Albacete, Spain; (M.D.P.-C.); (I.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Consorcio CIBER, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Solera
- Servicio de Medicina Interna, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain;
- Facultad de Medicina de Albacete, Universidad de Castilla-La Mancha, 02006 Albacete, Spain
| | - Valentín Ceña
- Unidad Asociada Neurodeath, Universidad de Castilla-La Mancha, 02006 Albacete, Spain; (M.D.P.-C.); (I.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Consorcio CIBER, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
513
|
Vitamin D Status and Parkinson's Disease. Brain Sci 2022; 12:brainsci12060790. [PMID: 35741675 PMCID: PMC9221008 DOI: 10.3390/brainsci12060790] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 02/04/2023] Open
Abstract
Parkinson’s disease (PD) is a complex and progressive neurodegenerative disease, characterized by resting tremor, rigidity, slowness of movement, and postural instability. Furthermore, PD is associated with a wide spectrum of non-motor symptoms that add to overall disability. In recent years, some investigations, from basic science to clinical applications, have focused on the role of vitamin D in PD, often with controversial findings. Vitamin D has widespread effects on several biological processes in the central nervous system, including neurotransmission in dopaminergic neural circuits. Various studies have recorded lower levels of vitamin D in PD patients than in healthy controls. Low vitamin D status has also been correlated with the risk for PD and motor severity, whereas less is known about the effects vitamin D has on cognitive function and other non-motor symptoms. This review aims to better characterize the correlation between vitamin D and PD, clarify the role of vitamin D in PD prevention and treatment, and discuss avenues for future research in this field.
Collapse
|
514
|
Wang ZB, Qu J, Yang ZY, Liu DY, Jiang SL, Zhang Y, Yang ZQ, Mao XY, Liu ZQ. Integrated Analysis of Expression Profile and Potential Pathogenic Mechanism of Temporal Lobe Epilepsy With Hippocampal Sclerosis. Front Neurosci 2022; 16:892022. [PMID: 35784838 PMCID: PMC9243442 DOI: 10.3389/fnins.2022.892022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To investigate the potential pathogenic mechanism of temporal lobe epilepsy with hippocampal sclerosis (TLE+HS) by analyzing the expression profiles of microRNA/ mRNA/ lncRNA/ DNA methylation in brain tissues. Methods Brain tissues of six patients with TLE+HS and nine of normal temporal or parietal cortices (NTP) of patients undergoing internal decompression for traumatic brain injury (TBI) were collected. The total RNA was dephosphorylated, labeled, and hybridized to the Agilent Human miRNA Microarray, Release 19.0, 8 × 60K. The cDNA was labeled and hybridized to the Agilent LncRNA+mRNA Human Gene Expression Microarray V3.0,4 × 180K. For methylation detection, the DNA was labeled and hybridized to the Illumina 450K Infinium Methylation BeadChip. The raw data was extracted from hybridized images using Agilent Feature Extraction, and quantile normalization was performed using the Agilent GeneSpring. P-value < 0.05 and absolute fold change >2 were considered the threshold of differential expression data. Data analyses were performed using R and Bioconductor. BrainSpan database was used to screen for signatures that were not differentially expressed in normal human hippocampus and cortex (data from BrainSpan), but differentially expressed in TLE+HS’ hippocampus and NTP’ cortex (data from our cohort). The strategy “Guilt by association” was used to predict the prospective roles of each important hub mRNA, miRNA, or lncRNA. Results A significantly negative correlation (r < −0.5) was found between 116 pairs of microRNA/mRNA, differentially expressed in six patients with TLE+HS and nine of NTP. We examined this regulation network’s intersection with target gene prediction results and built a lncRNA-microRNA-Gene regulatory network with structural, and functional significance. Meanwhile, we found that the disorder of FGFR3, hsa-miR-486-5p, and lnc-KCNH5-1 plays a key vital role in developing TLE+HS.
Collapse
Affiliation(s)
- Zhi-Bin Wang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha, China
| | - Jian Qu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhuan-Yi Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ding-Yang Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Shi-Long Jiang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha, China
| | - Ying Zhang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Zhi-Quan Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Zhi-Quan Yang,
| | - Xiao-Yuan Mao
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha, China
- Xiao-Yuan Mao,
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha, China
- *Correspondence: Zhao-Qian Liu,
| |
Collapse
|
515
|
Romo-Vaquero M, Fernández-Villalba E, Gil-Martinez AL, Cuenca-Bermejo L, Espín JC, Herrero MT, Selma MV. Urolithins: potential biomarkers of gut dysbiosis and disease stage in Parkinson's patients. Food Funct 2022; 13:6306-6316. [PMID: 35611932 DOI: 10.1039/d2fo00552b] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Gut microbiota alteration (gut dysbiosis) occurs during the onset and progression of Parkinson's disease. Gut dysbiosis biomarkers could be relevant to prodromal disease. Urolithins, anti-inflammatory metabolites produced from some dietary polyphenols by specific gut microbial ecologies (urolithin metabotypes), have been proposed as biomarkers of gut microbiota composition and functionality. However, this has not been explored in Parkinson's disease patients. The current study aimed to assess associations between urolithin metabotypes, gut dysbiosis and disease severity in Parkinson's disease patients. Participants (52 patients and 117 healthy controls) provided stool samples for microbiota sequencing and urine samples for urolithin profiling before and after consuming 30 g of walnuts for three days. Data on demographics, medication, disease duration and Hoehn and Yahr disease stage were collected. We observed a significant gradual increase of urolithin non-producers (metabotype-0) as the disease severity increased. The gut microbiome of metabotype-0 patients and patients with the greatest severity was characterized by a more altered bacterial composition, i.e., increased pro-inflammatory Enterobacteriaceae and reduced protective bacteria against autoimmune and inflammatory processes, including butyrate and urolithin-producing bacteria (Lachnospiraceae members and Gordonibacter). Besides, their microbiome was characterized by predictive functions of lipopolysaccharide biosynthesis and metabolism of glutathione, cysteine and methionine that could indirectly reflect the gut pro-inflammatory status. Urolithin detection in urine is a feasible, non-invasive and fast approach that can reflect gut microbiome dysbiosis and intestinal inflammation in Parkinson's disease patients. Our current study could provide novel strategies for improving diagnostics, and for preventing and treating disease progression in microbiota-based interventions.
Collapse
Affiliation(s)
- María Romo-Vaquero
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, Campus de Espinardo, Murcia 30100, Spain.
| | - Emiliano Fernández-Villalba
- Institute of Biomedical Research of Murcia (IMIB), Campus de Ciencias de la Salud, Carretera Buenavista s/n, El Palmar, Murcia 30120, Spain.,Clinical & Experimental Neuroscience, Institute for Aging Research, School of Medicine, University of Murcia, Murcia, Spain.
| | - Ana-Luisa Gil-Martinez
- Institute of Biomedical Research of Murcia (IMIB), Campus de Ciencias de la Salud, Carretera Buenavista s/n, El Palmar, Murcia 30120, Spain.,Clinical & Experimental Neuroscience, Institute for Aging Research, School of Medicine, University of Murcia, Murcia, Spain.
| | - Lorena Cuenca-Bermejo
- Institute of Biomedical Research of Murcia (IMIB), Campus de Ciencias de la Salud, Carretera Buenavista s/n, El Palmar, Murcia 30120, Spain.,Clinical & Experimental Neuroscience, Institute for Aging Research, School of Medicine, University of Murcia, Murcia, Spain.
| | - Juan Carlos Espín
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, Campus de Espinardo, Murcia 30100, Spain.
| | - María Trinidad Herrero
- Institute of Biomedical Research of Murcia (IMIB), Campus de Ciencias de la Salud, Carretera Buenavista s/n, El Palmar, Murcia 30120, Spain.,Clinical & Experimental Neuroscience, Institute for Aging Research, School of Medicine, University of Murcia, Murcia, Spain.
| | - María Victoria Selma
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, Campus de Espinardo, Murcia 30100, Spain.
| |
Collapse
|
516
|
Chen L, Huang T, Ma D, Chen YC. Altered Default Mode Network Functional Connectivity in Parkinson’s Disease: A Resting-State Functional Magnetic Resonance Imaging Study. Front Neurosci 2022; 16:905121. [PMID: 35720728 PMCID: PMC9204219 DOI: 10.3389/fnins.2022.905121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeWhether the intrinsic functional connectivity pattern of the default mode network (DMN) is involved in the progression of cognitive decline in Parkinson’s disease (PD) remains unclear. This study aimed to investigate the intrinsic functional connectivity (FC) pattern of the DMN anchored on the posterior cingulate cortex (PCC) in patients with PD by resting-state functional magnetic resonance imaging (fMRI).MethodsFifty patients with PD and 50 healthy controls (HCs) were included for resting-state fMRI scanning. A seed-based FC method was used to reveal FC patterns in the DMN with region of interest (ROI) in the PCC. Relationships between FC patterns and disease severity (UPDRS-III) were detected.ResultsCompared with the HCs, the patients with PD showed increased FC between the PCC and the right precuneus, left cuneus, and right angular gyrus. In the PD group, the increased FC values in the right precuneus were significantly and positively correlated with motor severity as assessed with UPDRS-III scores (rho = 0.337, p = 0.02).ConclusionOur result highlights that the patients with PD showed increased FC between the PCC and the right precuneus, left cuneus, and right angular gyrus in the DMN. The altered connectivity pattern in the DMN may play a crucial role in the neurophysiological mechanism of cognitive decline in patients with PD. These findings might provide new insights into neural mechanisms of cognitive decline in PD.
Collapse
Affiliation(s)
- Lu Chen
- Department of Radiology, Nanjing Integrated Traditional Chinese and Western Medicine Hospital Affiliated With Nanjing University of Chinese Medicine, Nanjing, China
| | - Ting Huang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Di Ma
- College of Information Science and Technology, Nanjing Forestry University, Nanjing, China
| | - Yu-Chen Chen
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Yu-Chen Chen,
| |
Collapse
|
517
|
Circulating microRNAs as potential biomarkers for the diagnosis of Parkinson's disease: A meta-analysis. Neurologia 2022. [DOI: 10.1016/j.nrl.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
518
|
Freuer D, Meisinger C. Association between inflammatory bowel disease and Parkinson's disease: A Mendelian randomization study. NPJ Parkinsons Dis 2022; 8:55. [PMID: 35534507 PMCID: PMC9085764 DOI: 10.1038/s41531-022-00318-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 04/08/2022] [Indexed: 12/13/2022] Open
Abstract
Emerging evidence from observational studies suggests an increased risk of Parkinson's disease (PD) in patients with inflammatory bowel disease (IBD). However, to date it is not clear whether a causal relationship exists. To investigate whether IBD is causally related to PD, a two-sample Mendelian randomization study was carried out. Independent genetic instruments from the largest available genome-wide association study (GWAS) for IBD (7045 cases, 456,327 controls) including European participants were used to investigate the association with PD (56,306 cases; 1.4 million controls). The results were validated by using a second IBD sample (12,882 cases; 21,770 controls) including the main subtypes ulcerative colitis (UC; 6968 cases; 20,464 controls) and Crohn's disease (CD; 5956 cases; 14,927 controls). The radial inverse-variance weighted (IVW) approach was used in the primary analysis, and the robustness of the findings were confirmed in a number of sensitivity analyses. Finally, the recently proposed CAUSE approach was performed. There was no evidence of an association between IBD and PD (ORIVW = 0.98; 95% CI: [0.93; 1.04]; P = 0.48). This finding could be validated using a second sample of IBD cases (ORIVW = 0.98; 95% CI: [0.95; 1.02]; P = 0.36). Furthermore, MR analyses did not support a causal effect of CD (ORIVW = 1.00; 95% CI: [0.98; 1.03]; P = 0.96) or UC (ORIVW = 1.02; 95% CI: [0.98; 1.06]; P = 0.45) on PD. The present study suggests that neither IBD nor its subtypes CD and UC causally affect Parkinson's disease in the European population. Further research is necessary to investigate whether intestinal inflammation impacts the development of PD.
Collapse
Affiliation(s)
- D Freuer
- Chair of Epidemiology, University of Augsburg, University Hospital of Augsburg, Augsburg, Germany.
| | - C Meisinger
- Chair of Epidemiology, University of Augsburg, University Hospital of Augsburg, Augsburg, Germany
| |
Collapse
|
519
|
Action and emotion perception in Parkinson's disease: A neuroimaging meta-analysis. Neuroimage Clin 2022; 35:103031. [PMID: 35569229 PMCID: PMC9112018 DOI: 10.1016/j.nicl.2022.103031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 03/01/2022] [Accepted: 05/02/2022] [Indexed: 11/23/2022]
Abstract
The neural substrates for action and emotion perception deficits in PD are still unclear. We addressed this issue via coordinate-based meta-analyses of previous fMRI data. PD patients exhibit decreased response in the basal ganglia. PD patients exhibit a trend toward decreased response in the parietal areas. PD patients exhibit a trend toward increased activation in the posterior cerebellum.
Patients with Parkinson disease (PD) may show impairments in the social perception. Whether these deficits have been consistently reported, it remains to be clarified which brain alterations subtend them. To this aim, we conducted a neuroimaging meta-analysis to compare the brain activity during social perception in patients with PD versus healthy controls. Our results show that PD patients exhibit a significantly decreased response in the basal ganglia (putamen and pallidum) and a trend toward decreased activity in the mirror system, particularly in the left parietal cortex (inferior parietal lobule and intraparietal sulcus). This reduced activation may be tied to a disruption of cognitive resonance mechanisms and may thus constitute the basis of impaired others’ representations underlying action and emotion perception. We also found increased activation in the posterior cerebellum in PD, although only in a within-group analysis and not in comparison with healthy controls. This cerebellar activation may reflect compensatory mechanisms, an aspect that deserves further investigation. We discuss the clinical implications of our findings for the development of novel social skill training programs for PD patients.
Collapse
|
520
|
Otani RTV, Yamamoto JYS, Nunes DM, Haddad MS, Parmera JB. Magnetic resonance and dopamine transporter imaging for the diagnosis of Parkinson´s disease: a narrative review. ARQUIVOS DE NEURO-PSIQUIATRIA 2022; 80:116-125. [PMID: 35976320 PMCID: PMC9491424 DOI: 10.1590/0004-282x-anp-2022-s130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/29/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND the diagnosis of Parkinson's disease (PD) can be challenging, especially in the early stages, albeit its updated and validated clinical criteria. Recent developments on neuroimaging in PD, altogether with its consolidated role of excluding secondary and other neurodegenerative causes of parkinsonism, provide more confidence in the diagnosis across the different stages of the disease. This review highlights current knowledge and major recent advances in magnetic resonance and dopamine transporter imaging in aiding PD diagnosis. OBJECTIVE This study aims to review current knowledge about the role of magnetic resonance imaging and neuroimaging of the dopamine transporter in diagnosing Parkinson's disease. METHODS We performed a non-systematic literature review through the PubMed database, using the keywords "Parkinson", "magnetic resonance imaging", "diffusion tensor", "diffusion-weighted", "neuromelanin", "nigrosome-1", "single-photon emission computed tomography", "dopamine transporter imaging". The search was restricted to articles written in English, published between January 2010 and February 2022. RESULTS The diagnosis of Parkinson's disease remains a clinical diagnosis. However, new neuroimaging biomarkers hold promise for increased diagnostic accuracy, especially in earlier stages of the disease. CONCLUSION Future validation of new imaging biomarkers bring the expectation of an increased neuroimaging role in the diagnosis of PD in the following years.
Collapse
Affiliation(s)
- Rafael Tomio Vicentini Otani
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, São Paulo SP, Brazil
| | - Joyce Yuri Silvestre Yamamoto
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, São Paulo SP, Brazil
| | - Douglas Mendes Nunes
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departmento de Radiologia e Oncologia, Instituto de Radiologia, São Paulo SP, Brazil
| | - Mônica Santoro Haddad
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, São Paulo SP, Brazil
| | - Jacy Bezerra Parmera
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, São Paulo SP, Brazil
| |
Collapse
|
521
|
Morowitz JM, Pogson KB, Roque DA, Church FC. Role of SARS-CoV-2 in Modifying Neurodegenerative Processes in Parkinson's Disease: A Narrative Review. Brain Sci 2022; 12:536. [PMID: 35624923 PMCID: PMC9139310 DOI: 10.3390/brainsci12050536] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/12/2022] [Accepted: 04/21/2022] [Indexed: 12/12/2022] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, continues to impact global health regarding both morbidity and mortality. Although SARS-CoV-2 primarily causes acute respiratory distress syndrome (ARDS), the virus interacts with and influences other organs and tissues, including blood vessel endothelium, heart, gastrointestinal tract, and brain. We are learning much about the pathophysiology of SARS-CoV-2 infection; however, we are just beginning to study and understand the long-term and chronic health consequences. Since the pandemic's beginning in late 2019, older adults, those with pre-existing illnesses, or both, have an increased risk of contracting COVID-19 and developing severe COVID-19. Furthermore, older adults are also more likely to develop the neurodegenerative disorder Parkinson's disease (PD), with advanced age as the most significant risk factor. Thus, does SARS-CoV-2 potentially influence, promote, or accelerate the development of PD in older adults? Our initial focus was aimed at understanding SARS-CoV-2 pathophysiology and the connection to neurodegenerative disorders. We then completed a literature review to assess the relationship between PD and COVID-19. We described potential molecular and cellular pathways that indicate dopaminergic neurons are susceptible, both directly and indirectly, to SARS-CoV-2 infection. We concluded that under certain pathological circumstances, in vulnerable persons-with-Parkinson's disease (PwP), SARS-CoV-2 acts as a neurodegenerative enhancer to potentially support the development or progression of PD and its related motor and non-motor symptoms.
Collapse
Affiliation(s)
- Jeremy M. Morowitz
- Developmental and Stem Cell Biology Program, Duke University, Durham, NC 27708, USA;
| | - Kaylyn B. Pogson
- School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Daniel A. Roque
- Department of Neurology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA;
| | - Frank C. Church
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
522
|
Redenšek S, Kristanc T, Blagus T, Trošt M, Dolžan V. Genetic Variability of the Vitamin D Receptor Affects Susceptibility to Parkinson’s Disease and Dopaminergic Treatment Adverse Events. Front Aging Neurosci 2022; 14:853277. [PMID: 35517045 PMCID: PMC9063754 DOI: 10.3389/fnagi.2022.853277] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
Vitamin D is a lipid-soluble molecule and an important transcriptional regulator in many tissues and organs, including the brain. Its role has been demonstrated also in Parkinson’s disease (PD) pathogenesis. Vitamin D receptor (VDR) is responsible for the initiation of vitamin D signaling cascade. The aim of this study was to assess the associations of VDR genetic variability with PD risk and different PD-related phenotypes. We genotyped 231 well characterized PD patients and 161 healthy blood donors for six VDR single nucleotide polymorphisms, namely rs739837, rs4516035, rs11568820, rs731236, rs2228570, and rs1544410. We observed that VDR rs2228570 is associated with PD risk (p < 0.001). Additionally, we observed associations of specific VDR genotypes with adverse events of dopaminergic treatment. VDR rs1544410 (GG vs. GA + AA: p = 0.005; GG vs. GA: p = 0.009) was associated with the occurrence of visual hallucinations and VDR rs739837 (TT vs. GG: p = 0.036), rs731236 (TT vs. TC + CC: p = 0.011; TT vs. TC: p = 0.028; TT vs. CC: p = 0.035), and rs1544410 (GG vs. GA: p = 0.014) with the occurrence of orthostatic hypotension. We believe that the reported study may support personalized approach to PD treatment, especially in terms of monitoring vitamin D level and vitamin D supplementation in patients with high risk VDR genotypes.
Collapse
Affiliation(s)
- Sara Redenšek
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tilen Kristanc
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tanja Blagus
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Maja Trošt
- Department of Neurology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Vita Dolžan,
| |
Collapse
|
523
|
Esfandiary A, Finkelstein DI, Voelcker NH, Rudd D. Clinical Sphingolipids Pathway in Parkinson’s Disease: From GCase to Integrated-Biomarker Discovery. Cells 2022; 11:cells11081353. [PMID: 35456032 PMCID: PMC9028315 DOI: 10.3390/cells11081353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 02/01/2023] Open
Abstract
Alterations in the sphingolipid metabolism of Parkinson’s Disease (PD) could be a potential diagnostic feature. Only around 10–15% of PD cases can be diagnosed through genetic alterations, while the remaining population, idiopathic PD (iPD), manifest without validated and specific biomarkers either before or after motor symptoms appear. Therefore, clinical diagnosis is reliant on the skills of the clinician, which can lead to misdiagnosis. IPD cases present with a spectrum of non-specific symptoms (e.g., constipation and loss of the sense of smell) that can occur up to 20 years before motor function loss (prodromal stage) and formal clinical diagnosis. Prodromal alterations in metabolites and proteins from the pathways underlying these symptoms could act as biomarkers if they could be differentiated from the broad values seen in a healthy age-matched control population. Additionally, these shifts in metabolites could be integrated with other emerging biomarkers/diagnostic tests to give a PD-specific signature. Here we provide an up-to-date review of the diagnostic value of the alterations in sphingolipids pathway in PD by focusing on the changes in definitive PD (postmortem confirmed brain data) and their representation in “probable PD” cerebrospinal fluid (CSF) and blood. We conclude that the trend of holistic changes in the sphingolipid pathway in the PD brain seems partly consistent in CSF and blood, and could be one of the most promising pathways in differentiating PD cases from healthy controls, with the potential to improve early-stage iPD diagnosis and distinguish iPD from other Parkinsonism when combined with other pathological markers.
Collapse
Affiliation(s)
- Ali Esfandiary
- Drug Delivery, Disposition and Dynamics, Monash University, Parkville, VIC 3052, Australia; (A.E.); (N.H.V.)
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC 3168, Australia
| | | | - Nicolas Hans Voelcker
- Drug Delivery, Disposition and Dynamics, Monash University, Parkville, VIC 3052, Australia; (A.E.); (N.H.V.)
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC 3168, Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, VIC 3168, Australia
- Materials Science and Engineering, Monash University, Clayton, VIC 3168, Australia
| | - David Rudd
- Drug Delivery, Disposition and Dynamics, Monash University, Parkville, VIC 3052, Australia; (A.E.); (N.H.V.)
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC 3168, Australia
- Correspondence: ; Tel.: +61-3-9903-9581
| |
Collapse
|
524
|
Zeng Y, Huang J, Tang X, Wang T, Chen S. The Impact of Triangle Hierarchical Management on Self-Management Behavior and Quality of Survival in Parkinson's Patients. Front Surg 2022; 9:878477. [PMID: 35495769 PMCID: PMC9051069 DOI: 10.3389/fsurg.2022.878477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTo investigate the effect of Triangle tiered and graded management on the self-management behavior and quality of survival of Parkinson's Disease (PD) patients.MethodsEighty ambulatory PD patients admitted to the neurology outpatient clinic of our hospital from June 2020 to January 2021 were selected for the study. Eighty patients were divided into 40 cases each in the test group and the control group using the random number table method. Patients in the control group were given conventional treatment and care, while in the test group, Triangle hierarchical management was applied on the basis of the control group. Non-motor symptoms [assessed by the Montreal Cognitive Inventory (MoCA), the Scale for Outcomes in PD for Autonomic Symptoms disability Scale (SCOPA-DS) and the Nocturnal Scale (SCOPA-NS)], motor symptoms [assessed by the Functional Gait Assessment (FGA), the Modified Ashworth Scale, and the Unified Parkinson's Disease Rating Scale (UPDRS-III)], quality of life (assessed by Barthel Index), medication adherence (self-administered medication adherence questionnaire), quality of survival (assessed by the 39-item Parkinson's Disease Quality of Survival Questionnaire, PDQ-39), and self-management effectiveness (assessed by the Chronic Disease Self-Efficacy Scale, symptom management and disease co-management) were compared between the two groups before and after the intervention. The two groups were also observed for satisfaction with care.ResultsAfter the intervention, the MoCA score, FGA score, Barthel Index, Medication adherence and all scores of self-management effectiveness were significantly higher in the test group than in the control group (P < 0.05); the SCOPA-DS score, SCOPA-NS score, Ashworth score, UPDRS-III score and PDQ-39 score were significantly lower than in the control group (P < 0.05). Satisfaction with nursing care was significantly higher in the test group than in the control group (P < 0.05).ConclusionThe application of Triangle's tiered and graded management to the home care of ambulatory PD patients was effective in improving their non-motor and motor symptoms, their ability to perform daily activities, medication adherence and self-management effectiveness, and their overall survival outcome.
Collapse
Affiliation(s)
- Yahua Zeng
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang, China
| | - Jianghua Huang
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang, China
| | - Xuan Tang
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang, China
| | - Ting Wang
- The First Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, China
| | - Shuangqin Chen
- The First Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, China
- *Correspondence: Shuangqin Chen
| |
Collapse
|
525
|
Thiankhaw K, Chattipakorn K, Chattipakorn SC, Chattipakorn N. Roles of humanin and derivatives on the pathology of neurodegenerative diseases and cognition. Biochim Biophys Acta Gen Subj 2022; 1866:130097. [DOI: 10.1016/j.bbagen.2022.130097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/13/2022] [Accepted: 01/20/2022] [Indexed: 10/19/2022]
|
526
|
Lu S, Jiang H, Shi Y. Association between irritable bowel syndrome and Parkinson's disease: A systematic review and meta-analysis. Acta Neurol Scand 2022; 145:442-448. [PMID: 34908158 DOI: 10.1111/ane.13570] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Growing evidence suggests that irritable bowel syndrome (IBS) and Parkinson's disease (PD) share similar pathological mechanisms and risk factors. METHODS We performed a systematic review and meta-analysis of the evidence for a relationship between IBS and PD. Risk estimates from individual studies were pooled using random-effects models. RESULTS Six articles involving 58,645 patients with PD were included in our meta-analysis. The overall risk for PD in IBS patients was significantly higher than that in the general population (odds ratio [OR], 1.5; 95% confidence interval [CI], 1.29-1.75; p < .001). Subgroup analysis revealed no significant differences in risk between men (OR = 1.47, 95% CI: 1.3-1.67; p < .001) and women (OR = 1.51, 95% CI: 1.29-1.75; p < .001); however, older (≥65 years) IBS patients (OR = 1.44, 95% CI: 1.3-1.59; p < .001) may be at higher risk for PD than younger (40-64 years) patients (OR = 1.32, 95% CI: 1.05-1.64; p = .017). CONCLUSION Overall, the PD risk was higher in IBS patients than others, indicating that the intestinal disorder may serve as a warning sign for PD.
Collapse
Affiliation(s)
- Sen Lu
- Department of Colorectal and Anal Surgery The First Affiliated Hospital of College of Medicine Zhejiang University Hangzhou China
| | - Hai‐yin Jiang
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases State Key Laboratory for Diagnosis and Treatment of Infectious Diseases The First Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
| | - Yu‐dan Shi
- Department of Chinese Internal Medicine Taizhou First People's Hospital Taizhou China
| |
Collapse
|
527
|
Provitera V, Iodice V, Manganelli F, Mozzillo S, Caporaso G, Stancanelli A, Borreca I, Esposito M, Dubbioso R, Iodice R, Vitale F, Koay S, Vichayanrat E, Valerio F, Santoro L, Nolano M. Postganglionic Sudomotor Assessment in Early Stage of Multiple System Atrophy and Parkinson Disease: A Morpho-functional Study. Neurology 2022; 98:e1282-e1291. [PMID: 35017309 PMCID: PMC8967330 DOI: 10.1212/wnl.0000000000013300] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/27/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Sudomotor impairment has been recognized as a key feature in differentiating Parkinson disease (PD) and multiple system atrophy-parkinsonian type (MSA-P), with the latter characterized by diffuse anhidrosis in prospective study, including patients in late stage of disease. We aimed to evaluate morphologic and functional postganglionic sudomotor involvement in patients with newly diagnosed MSA-P and PD to identify possible biomarkers that might be of help in differentiating the 2 conditions in the early stage. METHODS One hundred patients with parkinsonism within 2 years from onset of motor symptoms were included in the study. At the time of recruitment, questionnaires to assess nonmotor, autonomic, and small fiber symptoms were administered, and patients underwent postganglionic sudomotor function assessment by the dynamic sweat test and punch skin biopsy from the distal leg. Skin samples were processed for indirect immunofluorescence with a panel of antibodies, including noradrenergic and cholinergic markers. The density of intraepidermal, sudomotor, and pilomotor nerve fibers was measured on confocal images with dedicated software. A follow-up visit 12 months after recruitment was performed to confirm the diagnosis. RESULTS We recruited 57 patients with PD (M/F 36/21, age 63.5 ± 9.4 years) and 43 patients with MSA-P (M/F 27/16, age 62.3 ± 9.0 years). Clinical scales and questionnaires showed a more severe clinical picture in patients with MSA-P compared to those with PD. Sweating output and intraepidermal, pilomotor, and sudomotor nerve densities, compared to controls, were lower in both groups but with a greater impairment in patients with MSA-P. Pilomotor and sudomotor nerve density correlated with sweating function and with nonmotor clinical symptoms. A composite sudomotor parameter defined as the arithmetic product of sweat production multiplied by the density of sudomotor fibers efficiently separated the 2 populations; the receiver operating characteristics curve showed an area under the curve of 0.83. DISCUSSION Dynamic sweat test and the quantification of cutaneous autonomic nerves proved to be a sensitive morpho-functional approach to assess the postganglionic component of the sudomotor pathway, revealing a more severe involvement in MSA-P than in PD early in the disease course. This approach can be applied to differentiate the 2 conditions early. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that postganglionic sudomotor morpho-functional assessment accurately distinguish patients with PD from patients with MSA-P.
Collapse
Affiliation(s)
- Vincenzo Provitera
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy.
| | - Valeria Iodice
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| | - Fiore Manganelli
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| | - Stefania Mozzillo
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| | - Giuseppe Caporaso
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| | - Annamaria Stancanelli
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| | - Ilaria Borreca
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| | - Marcello Esposito
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| | - Raffaele Dubbioso
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| | - Rosa Iodice
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| | - Floriana Vitale
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| | - Shiwen Koay
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| | - Ekawat Vichayanrat
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| | - Fernanda Valerio
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| | - Lucio Santoro
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| | - Maria Nolano
- From the Neurology Department (V.P., S.M., G.C., A.S., I.B., M.N.), Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy; Department of Brain, Repair and Rehabilitation (V.I., S.K.), University College London Queen Square Institute of Neurology; Autonomic Unit (V.I., S.K., E.V., F. Valerio), National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.D., R.I., F. Vitale, L.S., M.N.), University Federico II of Naples; and Clinical Neurophysiology Unit (M.E.), Cardarelli Hospital, Naples, Italy
| |
Collapse
|
528
|
Cascaded Deep Learning Frameworks in Contribution to the Detection of Parkinson’s Disease. Bioengineering (Basel) 2022; 9:bioengineering9030116. [PMID: 35324805 PMCID: PMC8945200 DOI: 10.3390/bioengineering9030116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/10/2022] [Accepted: 03/10/2022] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disorder characterized by motor impairment, as well as tremors, stiffness, and rigidity. Besides the typical motor symptomatology, some Parkinsonians experience non-motor symptoms such as hyposmia, constipation, urinary dysfunction, orthostatic hypotension, memory loss, depression, pain, and sleep disturbances. The correct diagnosis of PD cannot be easy since there is no standard objective approach to it. After the incorporation of machine learning (ML) algorithms in medical diagnoses, the accuracy of disease predictions has improved. In this work, we have used three deep-learning-type cascaded neural network models based on the audial voice features of PD patients, called Recurrent Neural Networks (RNN), Multilayer Perception (MLP), and Long Short-Term Memory (LSTM), to estimate the accuracy of PD diagnosis. A performance comparison between the three models was performed on a sample of the subjects’ voice biomarkers. Experimental outcomes suggested that the LSTM model outperforms others with 99% accuracy. This study has also presented loss function curves on the relevance of good-fitting models to the detection of neurodegenerative diseases such as PD.
Collapse
|
529
|
Berdowska I, Matusiewicz M, Krzystek-Korpacka M. HDL Accessory Proteins in Parkinson’s Disease—Focusing on Clusterin (Apolipoprotein J) in Regard to Its Involvement in Pathology and Diagnostics—A Review. Antioxidants (Basel) 2022; 11:antiox11030524. [PMID: 35326174 PMCID: PMC8944556 DOI: 10.3390/antiox11030524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 02/04/2023] Open
Abstract
Parkinson’s disease (PD)—a neurodegenerative disorder (NDD) characterized by progressive destruction of dopaminergic neurons within the substantia nigra of the brain—is associated with the formation of Lewy bodies containing mainly α-synuclein. HDL-related proteins such as paraoxonase 1 and apolipoproteins A1, E, D, and J are implicated in NDDs, including PD. Apolipoprotein J (ApoJ, clusterin) is a ubiquitous, multifunctional protein; besides its engagement in lipid transport, it modulates a variety of other processes such as immune system functionality and cellular death signaling. Furthermore, being an extracellular chaperone, ApoJ interacts with proteins associated with NDD pathogenesis (amyloid β, tau, and α-synuclein), thus modulating their properties. In this review, the association of clusterin with PD is delineated, with respect to its putative involvement in the pathological mechanism and its application in PD prognosis/diagnosis.
Collapse
Affiliation(s)
- Izabela Berdowska
- Correspondence: (I.B.); (M.M.); Tel.: +48-71-784-13-92 (I.B.); +48-71-784-13-70 (M.M.)
| | | | | |
Collapse
|
530
|
Efficacy of Acupuncture for Parkinson's Disease Anxiety: Two-Stage Protocol for a Randomized Controlled Clinical Trial. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5180193. [PMID: 35186098 PMCID: PMC8849893 DOI: 10.1155/2022/5180193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/25/2021] [Accepted: 01/21/2022] [Indexed: 01/01/2023]
Abstract
Parkinson's disease anxiety (PDA) is a nonmotor symptom of Parkinson's disease (PD) that is often neglected. PDA poses a far-reaching challenge to the treatment of PD. Acupuncture could be successful in the treatment of PDA. However, the evidence for this is still limited. We propose a two-stage clinical trial. In stage 1, a total of 70 volunteers with PDA will be randomly assigned to either acupuncture (manual acupuncture) or control group (sham acupuncture) in a 1 : 1 ratio. Treatments will be performed for four weeks. The change in the Hamilton Rating Scale for Anxiety (HAMA) score from baseline to week 4 and week 12 will be the primary outcome. The levels of adrenocorticotropic hormone (ACTH), cortisol (CORT), serotonin (5-HT), and corticotropin-releasing factor (CRH) in the patients' serum and the scores on the Hoehn–Yahr Rating Scale and the Unified Parkinson's Disease Rating Scale (UPDRS) will all be considered among the secondary outcomes. Participants will be followed up until week 12. In stage 2, a total of 82 volunteers with PDA will be randomly assigned to either an acupuncture (manual acupuncture) or a control group (anti-Parkinson drugs only) in a 1 : 1 ratio. HAMA score will be the primary outcome. Universality, feasibility and cost effectiveness, Hoehn–Yahr Rating Scale, UPDRS, and serological indicators will be secondary outcomes. Participants will be followed up until week 4. The statistical analysis will include all the allocated individuals. The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine's Research Ethical Committee authorized this procedure, and the trial is registered with ChiCTR2100047253.
Collapse
|
531
|
Chang Z, Xie F, Li H, Yuan F, Zeng L, Shi L, Zhu S, Lu X, Wei X, Wang Q. Retinal Nerve Fiber Layer Thickness and Associations With Cognitive Impairment in Parkinson’s Disease. Front Aging Neurosci 2022; 14:832768. [PMID: 35222000 PMCID: PMC8867012 DOI: 10.3389/fnagi.2022.832768] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/20/2022] [Indexed: 01/18/2023] Open
Abstract
ObjectiveThis study intended to investigate whether retinal nerve fiber layer (RNFL) thickness could become a potential marker in patients with Parkinson’s disease with cognitive impairment (PD-CI).MethodsFifty-seven PD patients and 45 age-matched healthy controls (HCs) were recruited in our cross-sectional study and completed optical coherence tomography (OCT) evaluations. PD with normal cognition (PD-NC) and cognitive impairment (PD-CI) patients were divided following the 2015 Movement Disorder Society criteria. RNFL thickness was quantified in subfields of the 3.0-mm circle surrounding the optic disk; while a battery of neuropsychiatric assessments was conducted to estimate the Parkinsonism severity. General linear models and one-way ANOVA were adopted to assess RNFL thickness between subgroups with different cognitive statuses; logistic regression analyses were applied to determine the relation between RNFL and PD-CI cases.ResultsCompared with HCs, more thinning of the RNFL was observed in the inferior and temporal sectors in PD patients, especially in the PD-CI group. Inferior RNFL thickness was reduced in PD-CI compared with PD-NC patients. Logistic regression analysis found that inferior RNFL thickness was independently associated with PD-CI cases (odds ratio = 0.923, p = 0.014). Receiver operating characteristic analysis showed that the RNFL-involved combined model provided a high accuracy in screening cognitive deficiency in PD cases (area under the curve = 0.85, p < 0.001).ConclusionReduced RNFL thickness especially in the inferior sector is independently associated with PD-CI patients. Our study present new perspectives into verifying possible indicators for neuropathological processes or disease severity in Parkinsonians with cognitive dysfunction.
Collapse
Affiliation(s)
- Zihan Chang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fen Xie
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hualing Li
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Feilan Yuan
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lina Zeng
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lin Shi
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Shuzhen Zhu
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaohe Lu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Xiaohe Lu,
| | - Xiaobo Wei
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Xiaobo Wei,
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Qing Wang, ;
| |
Collapse
|
532
|
Solana-Manrique C, Sanz FJ, Torregrosa I, Palomino-Schätzlein M, Hernández-Oliver C, Pineda-Lucena A, Paricio N. Metabolic Alterations in a Drosophila Model of Parkinson's Disease Based on DJ-1 Deficiency. Cells 2022; 11:cells11030331. [PMID: 35159141 PMCID: PMC8834223 DOI: 10.3390/cells11030331] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/13/2022] Open
Abstract
Parkinson’s disease (PD) is the second-most common neurodegenerative disorder, whose physiopathology is still unclear. Moreover, there is an urgent need to discover new biomarkers and therapeutic targets to facilitate its diagnosis and treatment. Previous studies performed in PD models and samples from PD patients already demonstrated that metabolic alterations are associated with this disease. In this context, the aim of this study is to provide a better understanding of metabolic disturbances underlying PD pathogenesis. To achieve this goal, we used a Drosophila PD model based on inactivation of the DJ-1β gene (ortholog of human DJ-1). Metabolomic analyses were performed in 1-day-old and 15-day-old DJ-1β mutants and control flies using 1H nuclear magnetic resonance spectroscopy, combined with expression and enzymatic activity assays of proteins implicated in altered pathways. Our results showed that the PD model flies exhibited protein metabolism alterations, a shift fromthe tricarboxylic acid cycle to glycolytic pathway to obtain ATP, together with an increase in the expression of some urea cycle enzymes. Thus, these metabolic changes could contribute to PD pathogenesis and might constitute possible therapeutic targets and/or biomarkers for this disease.
Collapse
Affiliation(s)
- Cristina Solana-Manrique
- Departamento de Genética, Facultad CC Biológicas, Instituto Universitario de Biotecnología y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain; (C.S.-M.); (F.J.S.); (I.T.)
| | - Francisco José Sanz
- Departamento de Genética, Facultad CC Biológicas, Instituto Universitario de Biotecnología y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain; (C.S.-M.); (F.J.S.); (I.T.)
| | - Isabel Torregrosa
- Departamento de Genética, Facultad CC Biológicas, Instituto Universitario de Biotecnología y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain; (C.S.-M.); (F.J.S.); (I.T.)
| | | | - Carolina Hernández-Oliver
- Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain; (C.H.-O.); (A.P.-L.)
| | - Antonio Pineda-Lucena
- Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain; (C.H.-O.); (A.P.-L.)
- Programa de Terapias Moleculares, Centro de Investigación Médica Aplicada, Universidad de Navarra, 31008 Pamplona, Spain
| | - Nuria Paricio
- Departamento de Genética, Facultad CC Biológicas, Instituto Universitario de Biotecnología y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain; (C.S.-M.); (F.J.S.); (I.T.)
- Correspondence: ; Tel.: +34-96-354-3005; Fax: +34-96-354-3029
| |
Collapse
|
533
|
Garrido A, Santamaría E, Fernández-Irigoyen J, Soto M, Simonet C, Fernández M, Obiang D, Tolosa E, Martí MJ, Padmanabhan S, Malagelada C, Ezquerra M, Fernández-Santiago R. Differential Phospho-Signatures in Blood Cells Identify LRRK2 G2019S Carriers in Parkinson's Disease. Mov Disord 2022; 37:1004-1015. [PMID: 35049090 PMCID: PMC9306798 DOI: 10.1002/mds.28927] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/14/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022] Open
Abstract
Background The clinicopathological phenotype of G2019S LRRK2‐associated Parkinson's disease (L2PD) is similar to idiopathic Parkinson's disease (iPD), and G2019S LRRK2 nonmanifesting carriers (L2NMCs) are at increased risk for development of PD. With various therapeutic strategies in the clinical and preclinical pipeline, there is an urgent need to identify biomarkers that can aid early diagnosis and patient enrichment for ongoing and future LRRK2‐targeted trials. Objective The objective of this work was to investigate differential protein and phospho‐protein changes related to G2019S mutant LRRK2 in peripheral blood mononuclear cells from G2019S L2PD patients and G2019S L2NMCs, identify specific phospho‐protein changes associated with the G2019S mutation and with disease status, and compare findings with patients with iPD. Methods We performed an unbiased phospho‐proteomic study by isobaric label–based mass spectrometry using peripheral blood mononuclear cell group pools from a LRRK2 cohort from Spain encompassing patients with G2019S L2PD (n = 20), G2019S L2NMCs (n = 20), healthy control subjects (n = 30), patients with iPD (n = 15), patients with R1441G L2PD (n = 5), and R1441G L2NMCs (n = 3) (total N = 93). Results Comparing G2019S carriers with healthy controls, we identified phospho‐protein changes associated with the G2019S mutation. Moreover, we uncovered a specific G2019S phospho‐signature that changes with disease status and can discriminate patients with G2019S L2PD, G2019S L2NMCs, and healthy controls. Although patients with iPD showed a differential phospho‐proteomic profile, biological enrichment analyses revealed similar changes in deregulated pathways across the three groups. Conclusions We found a differential phospho‐signature associated with LRRK2 G2019S for which, consistent with disease status, the phospho‐profile from PD at‐risk G2019S L2NMCs was more similar to healthy controls than patients with G2019S L2PD with the manifested disease. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society
Collapse
Affiliation(s)
- Alicia Garrido
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - Enrique Santamaría
- Proteored-ISCIII, Proteomics Platform, Clinical Neuroproteomics Unit, Navarrabiomed, Departamento de Salud, UPNA, IdiSNA, Pamplona, Navarra, Spain
| | - Joaquín Fernández-Irigoyen
- Proteored-ISCIII, Proteomics Platform, Clinical Neuroproteomics Unit, Navarrabiomed, Departamento de Salud, UPNA, IdiSNA, Pamplona, Navarra, Spain
| | - Marta Soto
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - Cristina Simonet
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - Manel Fernández
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain.,Parkinson's Disease and Movement Disorders Group of the Institut de Neurociències (Universitat de Barcelona), Barcelona, Catalonia, Spain
| | - Donina Obiang
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - Eduardo Tolosa
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - María-José Martí
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - Shalini Padmanabhan
- The Michael J. Fox Foundation for Parkinson's Research, Grand Central Station, New York, New York, USA
| | - Cristina Malagelada
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain.,Department of Biomedicine, Faculty of Medicine, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Mario Ezquerra
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - Rubén Fernández-Santiago
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain.,Histology Unit, Department of Biomedicine, Faculty of Medicine, Universitat de Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
534
|
Takahashi S, Mashima K. Neuroprotection and Disease Modification by Astrocytes and Microglia in Parkinson Disease. Antioxidants (Basel) 2022; 11:antiox11010170. [PMID: 35052674 PMCID: PMC8773262 DOI: 10.3390/antiox11010170] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/03/2022] [Accepted: 01/13/2022] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress and neuroinflammation are common bases for disease onset and progression in many neurodegenerative diseases. In Parkinson disease, which is characterized by the degeneration of dopaminergic neurons resulting in dopamine depletion, the pathogenesis differs between hereditary and solitary disease forms and is often unclear. In addition to the pathogenicity of alpha-synuclein as a pathological disease marker, the involvement of dopamine itself and its interactions with glial cells (astrocyte or microglia) have attracted attention. Pacemaking activity, which is a hallmark of dopaminergic neurons, is essential for the homeostatic maintenance of adequate dopamine concentrations in the synaptic cleft, but it imposes a burden on mitochondrial oxidative glucose metabolism, leading to reactive oxygen species production. Astrocytes provide endogenous neuroprotection to the brain by producing and releasing antioxidants in response to oxidative stress. Additionally, the protective function of astrocytes can be modified by microglia. Some types of microglia themselves are thought to exacerbate Parkinson disease by releasing pro-inflammatory factors (M1 microglia). Although these inflammatory microglia may further trigger the inflammatory conversion of astrocytes, microglia may induce astrocytic neuroprotective effects (A2 astrocytes) simultaneously. Interestingly, both astrocytes and microglia express dopamine receptors, which are upregulated in the presence of neuroinflammation. The anti-inflammatory effects of dopamine receptor stimulation are also attracting attention because the functions of astrocytes and microglia are greatly affected by both dopamine depletion and therapeutic dopamine replacement in Parkinson disease. In this review article, we will focus on the antioxidative and anti-inflammatory effects of astrocytes and their synergism with microglia and dopamine.
Collapse
Affiliation(s)
- Shinichi Takahashi
- Department of Neurology and Stroke, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka-shi 350-1298, Japan
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
- Correspondence: ; Tel.: +81-42-984-4111 (ext. 7412); Fax: +81-42-984-0664
| | - Kyoko Mashima
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
- Department of Neurology, Tokyo Saiseikai Central Hospital, 1-4-17 Mita, Minato-ku, Tokyo 108-0073, Japan
| |
Collapse
|
535
|
Zhang XL, Zhang XH, Yu X, Zheng LF, Feng XY, Liu CZ, Quan ZS, Zhang Y, Zhu JX. Enhanced Contractive Tension and Upregulated Muscarinic Receptor 2/3 in Colorectum Contribute to Constipation in 6-Hydroxydopamine-Induced Parkinson's Disease Rats. Front Aging Neurosci 2022; 13:770841. [PMID: 35002677 PMCID: PMC8733788 DOI: 10.3389/fnagi.2021.770841] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/02/2021] [Indexed: 11/30/2022] Open
Abstract
Constipation and defecatory dysfunctions are frequent symptoms in patients with Parkinson’s disease (PD). The pathology of Lewy bodies in colonic and rectal cholinergic neurons suggests that cholinergic pathways are involved in colorectal dysmotility in PD. However, the underlying mechanism is unclear. The aim of the present study is to examine the effect of central dopaminergic denervation in rats, induced by injection 6-hydroxydopamine into the bilateral substania nigra (6-OHDA rats), on colorectal contractive activity, content of acetylcholine (ACh), vasoactive intestinal peptide (VIP) and expression of neural nitric oxide synthase (nNOS) and muscarinic receptor (MR). Strain gauge force transducers combined with electrical field stimulation (EFS), gut transit time, immunohistochemistry, ELISA, western blot and ultraperformance liquid chromatography tandem mass spectrometry were used in this study. The 6-OHDA rats exhibited outlet obstruction constipation characterized by prolonged transit time, enhanced contractive tension and fecal retention in colorectum. Pretreatment with tetrodotoxin significantly increased the colorectal motility. EFS-induced cholinergic contractions were diminished in the colorectum. Bethanechol chloride promoted colorectal motility in a dose-dependent manner, and much stronger reactivity of bethanechol chloride was observed in 6-OHDA rats. The ACh, VIP and protein expression of nNOS was decreased, but M2R and M3R were notably upregulated in colorectal muscularis externa. Moreover, the number of cholinergic neurons was reduced in sacral parasympathetic nucleus (SPN) of 6-OHDA rats. In conclusion, central nigrostriatal dopaminergic denervation is associated with decreased cholinergic neurons in SPN, decreased ACh, VIP content, and nNOS expression and upregulated M2R and M3R in colorectum, resulting in colorectal dysmotility, which contributes to outlet obstruction constipation. The study provides new insights into the mechanism of constipation and potential therapeutic targets for constipation in PD patients.
Collapse
Affiliation(s)
- Xiao-Li Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiao-Hui Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Artificial Liver Treatment Center, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xiao Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Li-Fei Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiao-Yan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chen-Zhe Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhu-Sheng Quan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yue Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jin-Xia Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
536
|
Grazia A, Wimmer M, Müller-Putz GR, Wriessnegger SC. Neural Suppression Elicited During Motor Imagery Following the Observation of Biological Motion From Point-Light Walker Stimuli. Front Hum Neurosci 2022; 15:788036. [PMID: 35069155 PMCID: PMC8779203 DOI: 10.3389/fnhum.2021.788036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/10/2021] [Indexed: 11/26/2022] Open
Abstract
Introduction: Advantageous effects of biological motion (BM) detection, a low-perceptual mechanism that allows the rapid recognition and understanding of spatiotemporal characteristics of movement via salient kinematics information, can be amplified when combined with motor imagery (MI), i.e., the mental simulation of motor acts. According to Jeannerod's neurostimulation theory, asynchronous firing and reduction of mu and beta rhythm oscillations, referred to as suppression over the sensorimotor area, are sensitive to both MI and action observation (AO) of BM. Yet, not many studies investigated the use of BM stimuli using combined AO-MI tasks. In this study, we assessed the neural response in the form of event-related synchronization and desynchronization (ERD/S) patterns following the observation of point-light-walkers and concordant MI, as compared to MI alone. Methods: Twenty right-handed healthy participants accomplished the experimental task by observing BM stimuli and subsequently performing the same movement using kinesthetic MI (walking, cycling, and jumping conditions). We recorded an electroencephalogram (EEG) with 32 channels and performed time-frequency analysis on alpha (8-13 Hz) and beta (18-24 Hz) frequency bands during the MI task. A two-way repeated-measures ANOVA was performed to test statistical significance among conditions and electrodes of interest. Results: The results revealed significant ERD/S patterns in the alpha frequency band between conditions and electrode positions. Post hoc comparisons showed significant differences between condition 1 (walking) and condition 3 (jumping) over the left primary motor cortex. For the beta band, a significantly less difference in ERD patterns (p < 0.01) was detected only between condition 3 (jumping) and condition 4 (reference). Discussion: Our results confirmed that the observation of BM combined with MI elicits a neural suppression, although just in the case of jumping. This is in line with previous findings of AO and MI (AOMI) eliciting a neural suppression for simulated whole-body movements. In the last years, increasing evidence started to support the integration of AOMI training as an adjuvant neurorehabilitation tool in Parkinson's disease (PD). Conclusion: We concluded that using BM stimuli in AOMI training could be promising, as it promotes attention to kinematic features and imitative motor learning.
Collapse
Affiliation(s)
- Alice Grazia
- Deutsches Zentrum für Neurodegenerative Erkrankungen, Rostock-Greifswald, Rostock, Germany
- Department of General Psychology, University of Padova, Padua, Italy
| | - Michael Wimmer
- Institute of Neural Engineering, Graz University of Technology, Graz, Austria
| | - Gernot R. Müller-Putz
- Institute of Neural Engineering, Graz University of Technology, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Selina C. Wriessnegger
- Institute of Neural Engineering, Graz University of Technology, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| |
Collapse
|
537
|
Xu H, Liu YY, Li LS, Liu YS. Sirtuins at the Crossroads between Mitochondrial Quality Control and Neurodegenerative Diseases: Structure, Regulation, Modifications, and Modulators. Aging Dis 2022; 14:794-824. [PMID: 37191431 DOI: 10.14336/ad.2022.1123] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/23/2022] [Indexed: 04/03/2023] Open
Abstract
Sirtuins (SIRT1-SIRT7), a family of nicotinamide adenine dinucleotide (NAD+)-dependent enzymes, are key regulators of life span and metabolism. In addition to acting as deacetylates, some sirtuins have the properties of deacylase, decrotonylase, adenosine diphosphate (ADP)-ribosyltransferase, lipoamidase, desuccinylase, demalonylase, deglutarylase, and demyristolyase. Mitochondrial dysfunction occurs early on and acts causally in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Sirtuins are implicated in the regulation of mitochondrial quality control, which is highly associated with the pathogenesis of neurodegenerative diseases. There is growing evidence indicating that sirtuins are promising and well-documented molecular targets for the treatment of mitochondrial dysfunction and neurodegenerative disorders by regulating mitochondrial quality control, including mitochondrial biogenesis, mitophagy, mitochondrial fission/fusion dynamics, and mitochondrial unfolded protein responses (mtUPR). Therefore, elucidation of the molecular etiology of sirtuin-mediated mitochondrial quality control points to new prospects for the treatment of neurodegenerative diseases. However, the mechanisms underlying sirtuin-mediated mitochondrial quality control remain obscure. In this review, we update and summarize the current understanding of the structure, function, and regulation of sirtuins with an emphasis on the cumulative and putative effects of sirtuins on mitochondrial biology and neurodegenerative diseases, particularly their roles in mitochondrial quality control. In addition, we outline the potential therapeutic applications for neurodegenerative diseases of targeting sirtuin-mediated mitochondrial quality control through exercise training, calorie restriction, and sirtuin modulators in neurodegenerative diseases.
Collapse
|
538
|
Role of Phytoconstituents as PPAR Agonists: Implications for Neurodegenerative Disorders. Biomedicines 2021; 9:biomedicines9121914. [PMID: 34944727 PMCID: PMC8698906 DOI: 10.3390/biomedicines9121914] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/09/2021] [Accepted: 12/11/2021] [Indexed: 12/16/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPAR-γ, PPAR-α, and PPAR-β/δ) are ligand-dependent nuclear receptors that play a critical role in the regulation of hundreds of genes through their activation. Their expression and targeted activation play an important role in the treatment of a variety of diseases, including neurodegenerative, cardiovascular, diabetes, and cancer. In recent years, several reviews have been published describing the therapeutic potential of PPAR agonists (natural or synthetic) in the disorders listed above; however, no comprehensive report defining the role of naturally derived phytoconstituents as PPAR agonists targeting neurodegenerative diseases has been published. This review will focus on the role of phytoconstituents as PPAR agonists and the relevant preclinical studies and mechanistic insights into their neuroprotective effects. Exemplary research includes flavonoids, fatty acids, cannabinoids, curcumin, genistein, capsaicin, and piperine, all of which have been shown to be PPAR agonists either directly or indirectly. Additionally, a few studies have demonstrated the use of clinical samples in in vitro investigations. The role of the fruit fly Drosophila melanogaster as a potential model for studying neurodegenerative diseases has also been highlighted.
Collapse
|
539
|
Li X, Fan X, Yang H, Liu Y. Review of Metabolomics-Based Biomarker Research for Parkinson's Disease. Mol Neurobiol 2021; 59:1041-1057. [PMID: 34826053 DOI: 10.1007/s12035-021-02657-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/17/2021] [Indexed: 01/12/2023]
Abstract
Parkinson's disease (PD), as the second most common neurodegenerative disease, is seriously affecting the life quality of the elderly. However, there is still a lack of efficient medical methods to diagnosis PD before apparent symptoms occur. In recent years, clinical biomarkers including genetic, imaging, and tissue markers have exhibited remarkable benefits in assisting PD diagnoses. Due to the advantages of high-throughput detection of metabolites and almost non-invasive sample collection, metabolomics research of PD is widely used for diagnostic biomarker discovery. However, there are also a few shortages for those identified biomarkers, such as the scarcity of verifications regarding the sensitivity and specificity. Thus, reviewing the research progress of PD biomarkers based on metabolomics techniques is of great significance for developing PD diagnosis. To comprehensively clarify the progress of current metabolic biomarker studies in PD, we reviewed 20 research articles regarding the discovery and validation of biomarkers for PD diagnosis from three mainstream academic databases (NIH PubMed, ISI Web of Science, and Elsevier ScienceDirect). By analyzing those materials, we summarized the metabolic biomarkers identified by those metabolomics studies and discussed the potential approaches used for biomarker verifications. In conclusion, this review provides a comprehensive and updated overview of PD metabolomics research in the past two decades and particularly discusses the validation of disease biomarkers. We hope those discussions might provide inspiration for PD biomarker discovery and verification in the future.
Collapse
Affiliation(s)
- Xin Li
- School of Pharmaceutical Sciences, Liaoning University, No. 66 Chongshan Middle Road, Huanggu District, Liaoning Province, 110036, Shenyang, People's Republic of China
| | - Xiaoying Fan
- School of Pharmaceutical Sciences, Liaoning University, No. 66 Chongshan Middle Road, Huanggu District, Liaoning Province, 110036, Shenyang, People's Republic of China
| | - Hongtian Yang
- School of Pharmaceutical Sciences, Liaoning University, No. 66 Chongshan Middle Road, Huanggu District, Liaoning Province, 110036, Shenyang, People's Republic of China
| | - Yufeng Liu
- School of Pharmaceutical Sciences, Liaoning University, No. 66 Chongshan Middle Road, Huanggu District, Liaoning Province, 110036, Shenyang, People's Republic of China. .,Natural Products Pharmaceutical Engineering Technology Research Center of Liaoning Province, Shenyang, 110036, People's Republic of China.
| |
Collapse
|
540
|
Giovannini D, Andreola F, Spitalieri P, Krasnowska EK, Colini Baldeschi A, Rossi S, Sangiuolo F, Cozzolino M, Serafino A. Natriuretic peptides are neuroprotective on in vitro models of PD and promote dopaminergic differentiation of hiPSCs-derived neurons via the Wnt/β-catenin signaling. Cell Death Discov 2021; 7:330. [PMID: 34725335 PMCID: PMC8560781 DOI: 10.1038/s41420-021-00723-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 12/16/2022] Open
Abstract
Over the last 20 years, the efforts to develop new therapies for Parkinson's disease (PD) have focused not only on the improvement of symptomatic therapy for motor and non-motor symptoms but also on the discovering of the potential causes of PD, in order to develop disease-modifying treatments. The emerging role of dysregulation of the Wnt/β-catenin signaling in the onset and progression of PD, as well as of other neurodegenerative diseases (NDs), renders the targeting of this signaling an attractive therapeutic opportunity for curing this brain disorder. The natriuretic peptides (NPs) atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and C-type natriuretic peptide (CNP), are cardiac and vascular-derived hormones also widely expressed in mammalian CNS, where they seem to participate in numerous brain functions including neural development/differentiation and neuroprotection. We recently demonstrated that ANP affects the Wnt/β-catenin pathway possibly through a Frizzled receptor-mediated mechanism and that it acts as a neuroprotective agent in in vitro models of PD by upregulating this signaling. Here we provide further evidence supporting the therapeutic potential of this class of natriuretic hormones. Specifically, we demonstrate that all the three natriuretic peptides are neuroprotective for SHSY5Y cells and primary cultures of DA neurons from mouse brain, subjected to neurotoxin insult with 6-hydroxydopamine (6-OHDA) for mimicking the neurodegeneration of PD, and these effects are associated with the activation of the Wnt/β-catenin pathway. Moreover, ANP, BNP, CNP are able to improve and accelerate the dopaminergic differentiation and maturation of hiPSCs-derived neural population obtained from two differed healthy donors, concomitantly affecting the canonical Wnt signaling. Our results support the relevance of exogenous ANP, BNP, and CNP as attractive molecules for both neuroprotection and neurorepair in PD, and more in general, in NDs for which aberrant Wnt signaling seems to be the leading pathogenetic mechanism.
Collapse
Affiliation(s)
- Daniela Giovannini
- Institute of Translational Pharmacology-National Research Council of Italy, Rome, Italy
| | - Federica Andreola
- Institute of Translational Pharmacology-National Research Council of Italy, Rome, Italy
| | - Paola Spitalieri
- Department of Biomedicine and Prevention, Genetic Medicine Unit, University of Rome "Tor Vergata", Rome, Italy
| | | | | | - Simona Rossi
- Institute of Translational Pharmacology-National Research Council of Italy, Rome, Italy
| | - Federica Sangiuolo
- Department of Biomedicine and Prevention, Genetic Medicine Unit, University of Rome "Tor Vergata", Rome, Italy
| | - Mauro Cozzolino
- Institute of Translational Pharmacology-National Research Council of Italy, Rome, Italy
| | - Annalucia Serafino
- Institute of Translational Pharmacology-National Research Council of Italy, Rome, Italy.
| |
Collapse
|
541
|
Rossi M, Perez-Lloret S, Merello M. How much time is needed in clinical practice to reach a diagnosis of clinically established Parkinson's disease? Parkinsonism Relat Disord 2021; 92:53-58. [PMID: 34695656 DOI: 10.1016/j.parkreldis.2021.10.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/08/2021] [Accepted: 10/16/2021] [Indexed: 12/26/2022]
Abstract
INTRODUCTION The implementation of accepted clinical diagnostic criteria has improved the accuracy of a clinical diagnosis of Parkinson's disease (PD). Time frames of 3-10 years have been empirically proposed to reach a diagnosis of clinically established PD. METHODS We explored the time to a Final Clinical Diagnosis (FCD) and the factors that predict faster diagnoses in patients presenting with parkinsonism and/or tremor between 2009 and 2015 at our tertiary center. All patients underwent a standardized workout process to reach a FCD, which included an acute levodopa challenge (LDC) after the first visit. RESULTS Among the 326 patients included, 215 (66%) received a FCD within the first six months after the LDC. A FCD was reached in 95% and 100% of patients in 33 and 108 months, respectively. PD was the FCD in 196 patients (60.1%). The FCD was reached faster in patients with a positive response to levodopa and when the FCD was PD. CONCLUSION The time needed to reach a final diagnosis in the clinical setting was 2.75 years in 95% of patients presenting initially with parkinsonism and/or tremor. Patients with positive responses to levodopa at the LDC, benefited from shorter delays until the FCD.
Collapse
Affiliation(s)
- Malco Rossi
- Movement Disorders Section, Neuroscience Department, Fleni, Buenos Aires, Argentina; National Research Council (CONICET), Buenos Aires, Argentina.
| | - Santiago Perez-Lloret
- National Research Council (CONICET), Buenos Aires, Argentina; Faculty of Medicine, Pontifical Catholic University of Argentina, Buenos Aires, Argentina; Universidad Abierta Interamericana, Centro de Altos Estudios en Ciencias Humanas y de la Salud (UAI-CAECIHS), Buenos Aires, Argentina; Department of Physiology, Faculty of Medicine, University of Buenos Aires, Argentina
| | - Marcelo Merello
- Movement Disorders Section, Neuroscience Department, Fleni, Buenos Aires, Argentina; National Research Council (CONICET), Buenos Aires, Argentina; Faculty of Medicine, Pontifical Catholic University of Argentina, Buenos Aires, Argentina
| |
Collapse
|
542
|
Palermo G, Giannoni S, Bellini G, Siciliano G, Ceravolo R. Dopamine Transporter Imaging, Current Status of a Potential Biomarker: A Comprehensive Review. Int J Mol Sci 2021; 22:11234. [PMID: 34681899 PMCID: PMC8538800 DOI: 10.3390/ijms222011234] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
A major goal of current clinical research in Parkinson's disease (PD) is the validation and standardization of biomarkers enabling early diagnosis, predicting outcomes, understanding PD pathophysiology, and demonstrating target engagement in clinical trials. Molecular imaging with specific dopamine-related tracers offers a practical indirect imaging biomarker of PD, serving as a powerful tool to assess the status of presynaptic nigrostriatal terminals. In this review we provide an update on the dopamine transporter (DAT) imaging in PD and translate recent findings to potentially valuable clinical practice applications. The role of DAT imaging as diagnostic, preclinical and predictive biomarker is discussed, especially in view of recent evidence questioning the incontrovertible correlation between striatal DAT binding and nigral cell or axon counts.
Collapse
Affiliation(s)
- Giovanni Palermo
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.P.); (S.G.); (G.B.); (G.S.)
| | - Sara Giannoni
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.P.); (S.G.); (G.B.); (G.S.)
- Unit of Neurology, San Giuseppe Hospital, 50053 Empoli, Italy
| | - Gabriele Bellini
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.P.); (S.G.); (G.B.); (G.S.)
| | - Gabriele Siciliano
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.P.); (S.G.); (G.B.); (G.S.)
| | - Roberto Ceravolo
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.P.); (S.G.); (G.B.); (G.S.)
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson’s Disease and Movement Disorders, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
543
|
Itokazu Y, Fuchigami T, Morgan JC, Yu RK. Intranasal infusion of GD3 and GM1 gangliosides downregulates alpha-synuclein and controls tyrosine hydroxylase gene in a PD model mouse. Mol Ther 2021; 29:3059-3071. [PMID: 34111562 DOI: 10.1016/j.ymthe.2021.06.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/20/2021] [Accepted: 06/03/2021] [Indexed: 12/17/2022] Open
Abstract
Parkinson's disease (PD) is characterized by Lewy bodies (composed predominantly of alpha-synuclein [aSyn]) and loss of pigmented midbrain dopaminergic neurons comprising the nigrostriatal pathway. Most PD patients show significant deficiency of gangliosides, including GM1, in the brain, and GM1 ganglioside appears to keep dopaminergic neurons functioning properly. Thus, supplementation of GM1 could potentially provide some rescuing effects. In this study, we demonstrate that intranasal infusion of GD3 and GM1 gangliosides reduces intracellular aSyn levels. GM1 also significantly enhances expression of tyrosine hydroxylase (TH) in the substantia nigra pars compacta of the A53T aSyn overexpressing mouse, following restored nuclear expression of nuclear receptor related 1 (Nurr1, also known as NR4A2), an essential transcription factor for differentiation, maturation, and maintenance of midbrain dopaminergic neurons. GM1 induces epigenetic activation of the TH gene, including augmentation of acetylated histones and recruitment of Nurr1 to the TH promoter region. Our data indicate that intranasal administration of gangliosides could reduce neurotoxic proteins and restore functional neurons via modulating chromatin status by nuclear gangliosides.
Collapse
Affiliation(s)
- Yutaka Itokazu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Takahiro Fuchigami
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - John C Morgan
- Movement Disorders Program, Parkinson's Foundation Center of Excellence, Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Robert K Yu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
544
|
Xing F, Marsili L, Truong DD. Parkinsonism in viral, paraneoplastic, and autoimmune diseases. J Neurol Sci 2021; 433:120014. [PMID: 34629181 DOI: 10.1016/j.jns.2021.120014] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022]
Abstract
Secondary parkinsonism, namely parkinsonism due to causes other than idiopathic neurodegeneration, may have multiple etiologies. Common secondary etiologies of parkinsonism such as drug-induced or vascular etiologies are well documented. Other secondary causes of parkinsonism such as infectious (mainly viral and prion-like diseases), autoimmune (systemic/drug-induced) and paraneoplastic etiologies are rare but are a topic of increasing interest. Older examples from the existing literature demonstrate the intricacies of viral infection from the last pandemic of the 20th century on the development of hypokinetic symptoms experienced in post-encephalitic patients. Viral and prion-like infections are only part of a complex interplay between the body's immune response and aberrant cell cycle perturbations leading to malignancy. In addition to the classic systemic autoimmune diseases (mainly systemic lupus erythematosus - SLE, and Sjögren syndrome), there have been new developments in the context of the current COVID-19 pandemic as well as more prominent use of immunotherapies such as immune checkpoint inhibitors in the treatment of solid tumors. Both of these developments have deepened our understanding of the underlying pathophysiologic process. Increased awareness and understanding of these rarer etiologies of parkinsonism is crucial to the modern diagnostic evaluation of a patient with parkinsonian symptoms as the potential treatment options may differ from the conventional levodopa-based therapeutic regimen of idiopathic Parkinson's disease. This review article aims to give an up-to-date review of the current literature on parkinsonian symptoms, their pathogenesis, diagnostic methods, and available treatment options. Many potential future directions in the field of parkinsonian conditions remain to be explored. This article is part of the Special Issue "Parkinsonism across the spectrum of movement disorders and beyond" edited by Joseph Jankovic, Daniel D. Truong and Matteo Bologna.
Collapse
Affiliation(s)
- Frank Xing
- Truong Neuroscience Institute, Orange Coast Memorial Medical Center, Fountain Valley, CA, USA
| | - Luca Marsili
- Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, USA
| | - Daniel D Truong
- Truong Neuroscience Institute, Orange Coast Memorial Medical Center, Fountain Valley, CA, USA; Department of Neurosciences, UC Riverside, Riverside, CA, USA.
| |
Collapse
|
545
|
Bologna M, Truong D, Jankovic J. The etiopathogenetic and pathophysiological spectrum of parkinsonism. J Neurol Sci 2021; 433:120012. [PMID: 34642022 DOI: 10.1016/j.jns.2021.120012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/05/2021] [Accepted: 09/29/2021] [Indexed: 12/12/2022]
Abstract
Parkinsonism is a syndrome characterized by bradykinesia, rigidity, and tremor. Parkinsonism is a common manifestation of Parkinson's disease and other neurodegenerative diseases referred to as atypical parkinsonism. However, a growing body of clinical and scientific evidence indicates that parkinsonism may be part of the phenomenological spectrum of various neurological conditions to a greater degree than expected by chance. These include neurodegenerative conditions not traditionally classified as movement disorders, e.g., dementia and motor neuron diseases. In addition, parkinsonism may characterize a wide range of central nervous system diseases, e.g., autoimmune diseases, infectious diseases, cerebrospinal fluid disorders (e.g., normal pressure hydrocephalus), cerebrovascular diseases, and other conditions. Several pathophysiological mechanisms have been identified in Parkinson's disease and atypical parkinsonism. Conversely, it is not entirely clear to what extent the same mechanisms and key brain areas are also involved in parkinsonism due to a broader etiopathogenetic spectrum. We aimed to provide a comprehensive and up-to-date overview of the various etiopathogenetic and pathophysiological mechanisms of parkinsonism in a wide spectrum of neurological conditions, with a particular focus on the role of the basal ganglia involvement. The paper also highlights potential implications in the diagnostic approach and therapeutic management of patients. This article is part of the Special Issue "Parkinsonism across the spectrum of movement disorders and beyond" edited by Joseph Jankovic, Daniel D. Truong and Matteo Bologna.
Collapse
Affiliation(s)
- Matteo Bologna
- Department of Human Neurosciences, Sapienza University of Rome, Italy; IRCCS Neuromed, Pozzilli, IS, Italy.
| | - Daniel Truong
- Truong Neuroscience Institute, Orange Coast Memorial Medical Center, Fountain Valley, CA, USA; Department of Neurosciences, UC Riverside, Riverside, CA, USA
| | - Joseph Jankovic
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
546
|
Genkwanin suppresses MPP +-induced cytotoxicity by inhibiting TLR4/MyD88/NLRP3 inflammasome pathway in a cellular model of Parkinson's disease. Neurotoxicology 2021; 87:62-69. [PMID: 34481870 DOI: 10.1016/j.neuro.2021.08.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/09/2021] [Accepted: 08/31/2021] [Indexed: 11/22/2022]
Abstract
Parkinson's disease (PD) is a complicated multifactorial neurodegenerative disorder. Oxidative stress, neuroinflammatory response, and activation of apoptosis have been proposed to be tightly involved in the pathogenesis of PD. Genkwanin is a typical bioactive non-glycosylated flavonoid with anti-inflammatory and anti-oxidant activities. However, the effect of genkwanin on PD remains unclear. Cell viability, lactate dehydrogenase (LDH) release, caspase-3/7 activity, and apoptosis was evaluated by MTT, LDH release assay, caspase-3/7 activity assay, and TUNEL assay, respectively. The secretion of prostaglandin E2 (PGE2), tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 were measured by respective commercial ELISA kits. The mRNA expression of TNF-α, IL-1β, and IL-6 was detected by qRT-PCR. The protein levels of cycloxygenase-2 (COX-2), toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), and NOD-like receptor (NLR) protein: 3 (NLRP3) were determined by western blot analysis. Genkwanin at concentrations less than 40 μM had no impact on cell viability and LDH release. Genkwanin suppressed MPP+-induced neuroinflammation in SH-SY5Y cells. MPP+ treatment inhibited cell viability, increased LDH release, apoptosis, and ROS generation, and reduced superoxide dismutase (SOD) activity in SH-SY5Y cells, which were abolished by genkwanin treatment. Genkwanin suppressed MPP+-induced activation of TLR4/MyD88/NLRP3 inflammasome pathway in SH-SY5Y cells. TLR4 overexpression weakened the anti-inflammatory and anti-neurotoxicity of genkwanin in SH-SY5Y cells. In conclusion, genkwanin attenuated neuroinflammation and neurotoxicity by inhibiting TLR4/MyD88/NLRP3 inflammasome pathway in MPP+-induced cellular model of PD.
Collapse
|
547
|
Bluhm A, Schrempel S, von Hörsten S, Schulze A, Roßner S. Proteolytic α-Synuclein Cleavage in Health and Disease. Int J Mol Sci 2021; 22:5450. [PMID: 34064208 PMCID: PMC8196865 DOI: 10.3390/ijms22115450] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
In Parkinson's disease, aggregates of α-synuclein within Lewy bodies and Lewy neurites represent neuropathological hallmarks. However, the cellular and molecular mechanisms triggering oligomeric and fibrillary α-synuclein aggregation are not fully understood. Recent evidence indicates that oxidative stress induced by metal ions and post-translational modifications such as phosphorylation, ubiquitination, nitration, glycation, and SUMOylation affect α-synuclein conformation along with its aggregation propensity and neurotoxic profiles. In addition, proteolytic cleavage of α-synuclein by specific proteases results in the formation of a broad spectrum of fragments with consecutively altered and not fully understood physiological and/or pathological properties. In the present review, we summarize the current knowledge on proteolytical α-synuclein cleavage by neurosin, calpain-1, cathepsin D, and matrix metalloproteinase-3 in health and disease. We also shed light on the contribution of the same enzymes to proteolytical processing of pathogenic proteins in Alzheimer's disease and report potential cross-disease mechanisms of pathogenic protein aggregation.
Collapse
Affiliation(s)
- Alexandra Bluhm
- Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany; (A.B.); (S.S.)
| | - Sarah Schrempel
- Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany; (A.B.); (S.S.)
| | - Stephan von Hörsten
- Department for Experimental Therapy, University Clinics Erlangen and Preclinical Experimental Center, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Anja Schulze
- Department of Molecular Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120 Halle/Saale, Germany;
| | - Steffen Roßner
- Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany; (A.B.); (S.S.)
| |
Collapse
|