51
|
Guo T, Guan X, Zeng Q, Xuan M, Gu Q, Xu X, Zhang M. Correlations between CSF proteins and spontaneous neuronal activity in Parkinson's disease. Neurosci Lett 2018; 673:61-66. [PMID: 29501577 DOI: 10.1016/j.neulet.2018.02.062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 02/27/2018] [Accepted: 02/27/2018] [Indexed: 12/21/2022]
Abstract
The relationship between cerebrospinal fluid (CSF) proteins and brain function in Parkinson's disease (PD) is not explained clearly. We investigated the correlations between CSF proteins and spontaneous neuronal activity in PD patients via fractional amplitude of low-frequency fluctuation (fALFF) using the Parkinson's Progression Markers Initiative database. Twenty-eight PD patients underwent resting-state functional magnetic resonance imaging in "off" status and lumbar puncture within a month. Correlation analyses between CSF proteins and fALFF value in whole brain as well as clinical assessment scores were performed. We found CSF total tau (t-tau) level was negatively correlated with fALFF in posterior cingulate gyrus. And fALFF in posterior cingulate gyrus was positively correlated with Hopkins Verbal Learning Test-Revised recognition discrimination index. Besides, alpha-synuclein (α-syn) level was correlated with fALFF in bilateral inferior frontal gyrus. This study provides evidence that CSF proteins may have a relationship with brain function related to cognitive status in PD patients.
Collapse
Affiliation(s)
- Tao Guo
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Guan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiaoling Zeng
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Min Xuan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Quanquan Gu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Xu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
52
|
Daniele S, Frosini D, Pietrobono D, Petrozzi L, Lo Gerfo A, Baldacci F, Fusi J, Giacomelli C, Siciliano G, Trincavelli ML, Franzoni F, Ceravolo R, Martini C, Bonuccelli U. α-Synuclein Heterocomplexes with β-Amyloid Are Increased in Red Blood Cells of Parkinson's Disease Patients and Correlate with Disease Severity. Front Mol Neurosci 2018; 11:53. [PMID: 29520218 PMCID: PMC5827358 DOI: 10.3389/fnmol.2018.00053] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/07/2018] [Indexed: 02/02/2023] Open
Abstract
Neurodegenerative disorders (NDs) are characterized by abnormal accumulation/misfolding of specific proteins, primarily α-synuclein (α-syn), β-amyloid1-42 (Aβ1-42) and tau, in both brain and peripheral tissues. In addition to oligomers, the role of the interactions of α-syn with Aβ or tau has gradually emerged. Nevertheless, despite intensive research, NDs have no accepted peripheral markers for biochemical diagnosis. In this respect, Red Blood Cells (RBCs) are emerging as a valid peripheral model for the study of aging-related pathologies. Herein, a small cohort (N = 28) of patients affected by Parkinson's disease (PD) and age-matched controls were enrolled to detect the content of α-syn (total and oligomeric), Aβ1-42 and tau (total and phosphorylated) in RBCs. Moreover, the presence of α-syn association with tau and Aβ1-42 was explored by co-immunoprecipitation/western blotting in the same cells, and quantitatively confirmed by immunoenzymatic assays. For the first time, PD patients were demonstrated to exhibit α-syn heterocomplexes with Aβ1-42 and tau in peripheral tissues; interestingly, α-syn-Aβ1-42 concentrations were increased in PD subjects with respect to healthy controls (HC), and directly correlated with disease severity and motor deficits. Moreover, total-α-syn levels were decreased in PD subjects and inversely related to their motor deficits. Finally, an increase of oligomeric-α-syn and phosphorylated-tau was observed in RBCs of the enrolled patients. The combination of three parameters (total-α-syn, phosphorylated-tau and α-syn-Aβ1-42 concentrations) provided the best fitting predictive index for discriminating PD patients from controls. Nevertheless further investigations should be required, overall, these data suggest α-syn hetero-aggregates in RBCs as a putative tool for the diagnosis of PD.
Collapse
Affiliation(s)
| | - Daniela Frosini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Lucia Petrozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Annalisa Lo Gerfo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Jonathan Fusi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Ferdinando Franzoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
53
|
Simuni T, Caspell-Garcia C, Coffey CS, Weintraub D, Mollenhauer B, Lasch S, Tanner CM, Jennings D, Kieburtz K, Chahine LM, Marek K. Baseline prevalence and longitudinal evolution of non-motor symptoms in early Parkinson's disease: the PPMI cohort. J Neurol Neurosurg Psychiatry 2018; 89:78-88. [PMID: 28986467 PMCID: PMC5732865 DOI: 10.1136/jnnp-2017-316213] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/18/2017] [Accepted: 08/23/2017] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To examine the baseline prevalence and longitudinal evolution in non-motor symptoms (NMS) in a prospective cohort of, at baseline, patients with de novo Parkinson's disease (PD) compared with healthy controls (HC). METHODS Parkinson's Progression Markers Initiative (PPMI) is a longitudinal, ongoing, controlled study of de novo PD participants and HC. NMS were rated using the Movement Disorder Society Unified Parkinson's Disease Rating Scale (MDS-UPDRS) Part I score and other validated NMS scales at baseline and after 2 years. Biological variables included cerebrospinal fluid (CSF) markers and dopamine transporter imaging. RESULTS 423 PD subjects and 196 HC were enrolled and followed for 2 years. MDS-UPDRS Part I total mean (SD) scores increased from baseline 5.6 (4.1) to 7.7 (5.0) at year 2 in PD subjects (p<0.001) versus from 2.9 (3.0) to 3.2 (3.0) in HC (p=0.38), with a significant difference between the groups (p<0.001). In the multivariate analysis, higher baseline NMS score was associated with female sex (p=0.008), higher baseline MDS-UPDRS Part II scores (p<0.001) and more severe motor phenotype (p=0.007). Longitudinal increase in NMS severity was associated with the older age (0.008) and lower CSF Aβ1-42 (0.005) at baseline. There was no association with the dose or class of dopaminergic therapy. CONCLUSIONS This study of NMS in early PD identified clinical and biological variables associated with both baseline burden and predictors of progression. The association of a greater longitudinal increase in NMS with lower baseline Aβ1-42 level is an important finding that will have to be replicated in other cohorts. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT01141023.
Collapse
Affiliation(s)
- Tanya Simuni
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | | - Daniel Weintraub
- University of Pennsylvania School of Medicine and the Parkinson’s Disease and Mental Illness Research, Education and Clinical Centers (PADRECC and MIRECC), Philadelphia Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Brit Mollenhauer
- Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel, University Medical Center Goettingen, Kassel, Germany
| | - Shirley Lasch
- Institute for Neurodegenerative Disorders, New Haven, Connecticut, USA
| | - Caroline M Tanner
- University of California San Francisco, San Francisco, California, USA
| | | | - Karl Kieburtz
- University of Rochester Medical Center, Rochester, New York, USA
| | - Lana M Chahine
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kenneth Marek
- Institute for Neurodegenerative Disorders, New Haven, Connecticut, USA
| |
Collapse
|
54
|
Goldman JG, Andrews H, Amara A, Naito A, Alcalay RN, Shaw LM, Taylor P, Xie T, Tuite P, Henchcliffe C, Hogarth P, Frank S, Saint‐Hilaire M, Frasier M, Arnedo V, Reimer AN, Sutherland M, Swanson‐Fischer C, Gwinn K, Kang UJ. Cerebrospinal fluid, plasma, and saliva in the BioFIND study: Relationships among biomarkers and Parkinson's disease Features. Mov Disord 2017; 33:282-288. [PMID: 29205509 PMCID: PMC5836918 DOI: 10.1002/mds.27232] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/08/2017] [Accepted: 10/16/2017] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE Examine relationships among neurodegenerative biomarkers and PD motor and nonmotor symptoms. BACKGROUND CSF alpha-synuclein is decreased in PD versus healthy controls, but whether plasma and saliva alpha-synuclein differentiate these groups is controversial. Correlations of alpha-synuclein among biofluids (CSF, plasma, saliva) or biomarkers (eg, beta-amyloid, tau [total, phosphorylated]) are not fully understood. The relationships of these biomarkers with PD clinical features remain unclear. METHODS BioFIND, a cross-sectional, observational study, examines clinical and biomarker characteristics in moderate-advanced PD and matched healthy controls. We compared alpha-synuclein concentrations across diagnosis, biofluids, and CSF biomarkers. Correlations of CSF biomarkers and MDS-UPDRS, motor phenotype, MoCA, and rapid eye movement sleep behavior disorder questionnaire scores in PD were examined. RESULTS CSF alpha-synuclein was lower in PD versus controls (P = .01), controlling for age, gender, and education. Plasma and saliva alpha-synuclein did not differ between PD and controls, and alpha-synuclein did not significantly correlate among biofluids. CSF beta-amyloid1-42 was lower in PD versus controls (P < .01), and correlated weakly with MoCA recall scores (r = 0.23, P = .02). CSF alpha-synuclein was lower in the postural instability/gait difficulty phenotype than other motor phenotypes (P < .01). No CSF biomarkers predicted or correlated with total motor or rapid eye movement sleep behavior disorder scores. CSF alpha-synuclein correlated with beta-amyloid1-42 , total-tau, and phosphorylated-tau (r = 0.41, 0.81, 0.43, respectively; Ps < .001). CONCLUSION Lower CSF alpha-synuclein is associated with diagnosis and motor phenotype in moderate-advanced PD. Plasma and saliva alpha-synuclein neither correlate with CSF alpha-synuclein, nor distinguish PD from controls. CSF beta-amyloid1-42 remains a potential biomarker for cognitive impairment in PD. © 2017 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jennifer G. Goldman
- Section of Parkinson Disease and Movement Disorders, Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Howard Andrews
- Division of Movement Disorders, Department of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
| | - Amy Amara
- Department of NeurologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Anna Naito
- The Michael J. Fox Foundation for Parkinson's ResearchNew YorkNew YorkUSA
| | - Roy N. Alcalay
- Division of Movement Disorders, Department of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
| | - Leslie M. Shaw
- Department of Pathology & Laboratory Medicine, Institute on Aging, Center for Neurodegenerative Disease ResearchUniversity of Pennsylvania School of MedicinePhiladelphiaPAUSA
| | | | - Tao Xie
- Parkinson Disease and Movement Disorder Program, Department of NeurologyUniversity of ChicagoChicagoIllinoisUSA
| | - Paul Tuite
- Department of NeurologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | | | - Penelope Hogarth
- Department of Molecular and Medical GeneticsOregon Health & Science UniversityPortlandOregonUSA
| | - Samuel Frank
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
| | | | - Mark Frasier
- The Michael J. Fox Foundation for Parkinson's ResearchNew YorkNew YorkUSA
| | - Vanessa Arnedo
- The Michael J. Fox Foundation for Parkinson's ResearchNew YorkNew YorkUSA
| | - Alyssa N. Reimer
- The Michael J. Fox Foundation for Parkinson's ResearchNew YorkNew YorkUSA
| | - Margaret Sutherland
- National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Christine Swanson‐Fischer
- National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Katrina Gwinn
- National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | | | - Un Jung Kang
- Division of Movement Disorders, Department of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
| |
Collapse
|
55
|
Tropea TF, Xie SX, Rick J, Chahine LM, Dahodwala N, Doshi J, Davatzikos C, Shaw LM, Van Deerlin V, Trojanowski JQ, Weintraub D, Chen-Plotkin AS. APOE, thought disorder, and SPARE-AD predict cognitive decline in established Parkinson's disease. Mov Disord 2017; 33:289-297. [PMID: 29168904 DOI: 10.1002/mds.27204] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/23/2017] [Accepted: 09/18/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND People with PD are at high risk of developing cognitive impairment and dementia. Cross-sectional studies have identified candidate biomarkers associated with cognitive decline. However, longitudinal studies on this topic are rarer, and few have investigated the use of biomarker panels encompassing multiple modalities. The objective of this study was to find baseline predictors of cognitive decline in longitudinally followed, nondemented Parkinson's disease patients. METHODS We performed a prospective cohort study of 100 PD patients with a median disease duration of 6.4 years. All participants were nondemented at baseline. We examined 16 baseline biomarkers from clinical, genetic, biochemical, and MRI-based imaging modalities for their association with longitudinal cognitive decline for up to 8 years. We investigated biomarkers individually, as well as in a multivariate linear mixed-effects model encompassing multimodal biomarkers, with change in the Mattis Dementia Rating Scale-2 over time as the primary outcome. Annual consensus process-derived cognitive diagnosis was used for Cox proportional hazards modeling of risk for cognitive decline. RESULTS In multivariate analysis, the presence of the APOE E4 allele, thought disorder, and an Alzheimer's disease pattern of brain atrophy (spatial pattern of abnormality for recognition of early Alzheimer's disease index) best predicted cognitive decline, with APOE E4 genotype exerting the greatest effect. The presence of the APOE E4 allele was associated with a 3.5 times higher risk of worsening cognitive diagnosis over time (HR, 3.53; 95% CI, 1.52-8.24; P < 0.05). The APOE genotype effect was not specific to any Mattis Dementia Rating Scale-2 domain. CONCLUSIONS Our results confirm the importance of Alzheimer's disease biomarkers as risk factors for cognitive decline in established Parkinson's disease. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Thomas F Tropea
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sharon X Xie
- Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jacqueline Rick
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lana M Chahine
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nabila Dahodwala
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jimit Doshi
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christos Davatzikos
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leslie M Shaw
- Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Vivianna Van Deerlin
- Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John Q Trojanowski
- Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel Weintraub
- Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Parkinson's Disease and Mental Illness Research, Education and Clinical Centers (PADRECC and MIRECC), Philadelphia Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
56
|
Cerebrospinal fluid levels of coenzyme Q10 are reduced in multiple system atrophy. Parkinsonism Relat Disord 2017; 46:16-23. [PMID: 29107645 DOI: 10.1016/j.parkreldis.2017.10.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 09/20/2017] [Accepted: 10/17/2017] [Indexed: 11/23/2022]
Abstract
INTRODUCTION The finding of mutations of the COQ2 gene and reduced coenzyme Q10 levels in the cerebellum in multiple system atrophy (MSA) suggest that coenzyme Q10 is relevant to MSA pathophysiology. Two recent studies have reported reduced coenzyme Q10 levels in plasma and serum (respectively) of MSA patients compared to Parkinson's disease and/or control subjects, but with largely overlapping values, limited comparison with other parkinsonisms, or dependence on cholesterol levels. We hypothesized that cerebrospinal fluid (CSF) is reliable to assess reductions in coenzyme Q10 as a candidate biomarker of MSA. METHODS In this preliminary cross-sectional study we assessed CSF coenzyme Q10 levels in 20 patients with MSA from the multicenter Catalan MSA Registry and of 15 PD patients, 10 patients with progressive supranuclear palsy (PSP), and 15 control subjects from the Movement Disorders Unit Biosample Collection of Hospital Clinic de Barcelona. A specific ELISA kit was used to determine CSF coenzyme Q10 levels. CSF coenzyme Q10 levels were compared in MSA vs. the other groups globally, pair-wise, and by binary logistic regression models adjusted for age, sex, disease severity, disease duration, and dopaminergic treatment. RESULTS CSF coenzyme Q10 levels were significantly lower in MSA than in other groups in global and pair-wise comparisons, as well as in multivariate regression models. Receiver operating characteristic curve analyses yielded significant areas under the curve for MSA vs. PD, PSP and controls. CONCLUSIONS These findings support coenzyme Q10 relevance in MSA. Low CSF coenzyme Q10 levels deserve further consideration as a biomarker of MSA.
Collapse
|
57
|
Polyakova TA, Arablinsky AV. [Neuroimaging and molecular biomarkers of dementia]. Zh Nevrol Psikhiatr Im S S Korsakova 2017; 117:16-22. [PMID: 28980608 DOI: 10.17116/jnevro20171176216-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Development of laboratory diagnosis and neuroimaging revealed a number of biomarkers for in vivo diagnosis of the most common forms of dementia (Alzheimer's disease, Lewy body dementia and vascular dementia). Currently, the highest diagnostic sensitivity and specificity of molecular biomarkers in the cerebrospinal fluid are detected for Alzheimer's disease. At the same time, the changes according to the magnetic resonance imaging are more prognostically significant for future cognitive decline than cerebrospinal fluid biomarkers. Cerebral microbleeds are an available adjuvant diagnostic marker, which increases the diagnostic value of leukoaraiosis that suggests the development of cerebral amyloid angiopathy or hypertensive microangiopathy, especially in cases of mixed pathology and severe cognitive deficits.
Collapse
Affiliation(s)
- T A Polyakova
- Russian Medical Academy for Continued Professional Education, Moscow, Russia
| | - A V Arablinsky
- Sechenov First Moscow Medical University, Moscow, Russia
| |
Collapse
|
58
|
Changes of cerebrospinal fluid Aβ 42, t-tau, and p-tau in Parkinson's disease patients with cognitive impairment relative to those with normal cognition: a meta-analysis. Neurol Sci 2017; 38:1953-1961. [PMID: 28808876 DOI: 10.1007/s10072-017-3088-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 08/02/2017] [Indexed: 01/11/2023]
Abstract
The cerebrospinal fluid (CSF) signature of reduced amyloid beta 1-42 (Aβ42), elevated total tau (t-tau), and phosphorylated tau181 (p-tau) is important for the early diagnosis of Alzheimer's disease (AD). Aβ42, t-tau, and p-tau have been reported in numerous studies to contribute to predicting cognitive impairment in Parkinson's disease (PDCI). However, no consistent conclusion can be drawn so far. Literatures regarding Aβ42, t-tau, and p-tau in CSF were systematically reviewed, and a meta-analysis was thus performed to evaluate the changes of these biomarkers in PDCI patients, including PD with mild cognitive impairment (PDMCI) and PD dementia (PDD) patients, relative to PD with normal cognition (PDNC) patients. Databases of "PubMed," "EBSCO," and "Springer" were retrieved for articles concerning Aβ42, t-tau, and p-tau in PDCI patients relative to those in PDNC patients published from January 1, 2000 to February 1, 2017. The following keywords were set, namely, "dementia" or "cognitive impairment" or "mild cognitive impairment" and "cerebrospinal fluid" and "Parkinson*." Sixteen articles comprising 590 PDCI patients and 1182 PDNC patients were included. The results showed that CSF Aβ42 level in PDCI cohort was lower than that in PDNC cohort (pooled Std.MD = -0.44, 95% CI [-0.61, -0.26], p < 0.00001). Reduced Aβ42 (pooled Std.MD = -0.60, 95% CI [-0.75, -0.45], p < 0.00001) as well as elevated t-tau (pooled Std.MD = 0.21, 95% CI [0.06, 0.35], p = 0.006) and p-tau (pooled Std.MD = 0.36, 95% CI [0.02, 0.69], p = 0.04) could be observed in PDD cohort compared with PDNC cohort. Therefore, amyloid pathology and tauopathy may participate in the development of PDD, which is similar to AD.
Collapse
|
59
|
LaBelle DR, Walsh RR, Banks SJ. Latent Cognitive Phenotypes in De Novo Parkinson's Disease: A Person-Centered Approach. J Int Neuropsychol Soc 2017; 23:551-563. [PMID: 28651678 PMCID: PMC6435330 DOI: 10.1017/s1355617717000406] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVES Cognitive impairment is an important aspect of Parkinson's disease (PD), but there is considerable heterogeneity in its presentation. This investigation aims to identify and characterize latent cognitive phenotypes in early PD. METHODS Latent class analysis, a data-driven, person-centered, cluster analysis was performed on cognitive data from the Parkinson's Progressive Markers Initiative baseline visit. This analytic method facilitates identification of naturally occurring endophenotypes. Resulting classes were compared across biomarker, symptom, and demographic data. RESULTS Six cognitive phenotypes were identified. Three demonstrated consistent performance across indicators, representing poor ("Weak-Overall"), average ("Typical-Overall"), and strong ("Strong-Overall") cognition. The remaining classes demonstrated unique patterns of cognition, characterized by "Strong-Memory," "Weak-Visuospatial," and "Amnestic" profiles. The Amnestic class evidenced greater tremor severity and anosmia, but was unassociated with biomarkers linked with Alzheimer's disease. The Weak-Overall class was older and reported more non-motor features associated with cognitive decline, including anxiety, depression, autonomic dysfunction, anosmia, and REM sleep behaviors. The Strong-Overall class was younger, more female, and reported less dysautonomia and anosmia. Classes were unrelated to disease duration, functional independence, or available biomarkers. CONCLUSIONS Latent cognitive phenotypes with focal patterns of impairment were observed in recently diagnosed individuals with PD. Cognitive profiles were found to be independent of traditional biomarkers and motoric indices of disease progression. Only globally impaired class was associated with previously reported indicators of cognitive decline, suggesting this group may drive the effects reported in studies using variable-based analysis. Longitudinal and neuroanatomical characterization of classes will yield further insight into the evolution of cognitive change in the disease. (JINS, 2017, 23, 551-563).
Collapse
Affiliation(s)
- Denise R LaBelle
- Neurology Department Cleveland Clinic Lou Ruvo Center for Brain Health,Las Vegas,Nevada
| | - Ryan R Walsh
- Neurology Department Cleveland Clinic Lou Ruvo Center for Brain Health,Las Vegas,Nevada
| | - Sarah J Banks
- Neurology Department Cleveland Clinic Lou Ruvo Center for Brain Health,Las Vegas,Nevada
| |
Collapse
|
60
|
Chiasserini D, Biscetti L, Eusebi P, Salvadori N, Frattini G, Simoni S, De Roeck N, Tambasco N, Stoops E, Vanderstichele H, Engelborghs S, Mollenhauer B, Calabresi P, Parnetti L. Differential role of CSF fatty acid binding protein 3, α-synuclein, and Alzheimer's disease core biomarkers in Lewy body disorders and Alzheimer's dementia. ALZHEIMERS RESEARCH & THERAPY 2017; 9:52. [PMID: 28750675 PMCID: PMC5532764 DOI: 10.1186/s13195-017-0276-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/05/2017] [Indexed: 02/08/2023]
Abstract
Background Neurodegenerative disorders such as Alzheimer’s disease (AD), Parkinson’s disease with dementia (PDD), and dementia with Lewy bodies (DLB) share clinical and molecular features. Cerebrospinal fluid (CSF) biomarkers may help the characterization of these diseases, improving the differential diagnosis. We evaluated the diagnostic performance of five CSF biomarkers across a well-characterized cohort of patients diagnosed with AD, DLB, PDD, and Parkinson’s disease (PD). Methods A total of 208 patients were enrolled in 3 European centers. The diagnostic groups (AD, n = 48; DLB, n = 40; PDD, n = 20; PD, n = 54) were compared with cognitively healthy neurological control subjects (patients with other neurological diseases [OND], n = 46). CSF levels of fatty acid binding protein 3, heart type (FABP3), α-synuclein (α-syn), amyloid-β peptide 1–42, total tau (t-tau), and phosphorylated tau 181 (p-tau) were assessed with immunoassays. Univariate and multivariate statistical analyses were applied to calculate the diagnostic value of the biomarkers as well as their association with clinical scores. Results FABP3 levels were significantly increased in patients with AD and DLB compared with those with PD and OND (p < 0.001). CSF t-tau, p-tau, and α-syn were significantly higher in patients with AD than in patients with PDD, DLB, PD, and OND. Combination of FABP3 with p-tau showed high accuracy for the differential diagnosis between AD and DLB (AUC 0.92), whereas patients with AD were separated from those with PDD using a combination of p-tau, FABP3, and α-syn (AUC 0.96). CSF FABP3 was inversely associated with Mini Mental State Examination score in the whole cohort (r = −0.42, p < 0.001). Conclusions The combination of CSF biomarkers linked to different aspects of neurodegeneration, such as FABP3, α-syn, and AD biomarkers, improves the biochemical characterization of AD and Lewy body disorders. Electronic supplementary material The online version of this article (doi:10.1186/s13195-017-0276-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Davide Chiasserini
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy. .,Oncoproteomics Laboratory, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| | - Leonardo Biscetti
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Paolo Eusebi
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Nicola Salvadori
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Giulia Frattini
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Simone Simoni
- Neurology Clinic, University Hospital S. Maria della Misericordia - University of Perugia, Perugia, Italy
| | - Naomi De Roeck
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Nicola Tambasco
- Neurology Clinic, University Hospital S. Maria della Misericordia - University of Perugia, Perugia, Italy
| | - Erik Stoops
- ADx NeuroSciences, Technologiepark 4, 9052, Gent, Belgium
| | | | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Brit Mollenhauer
- Department of Neurosurgery and Institute of Neuropathology, University Medicine Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.,Paracelsus-Elena Klinik, Klinikstrasse 16, 34128, Kassel, Germany
| | - Paolo Calabresi
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy.,Neurology Clinic, University Hospital S. Maria della Misericordia - University of Perugia, Perugia, Italy.,IRRCS S. Lucia Foundation, Rome, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy.,Neurology Clinic, University Hospital S. Maria della Misericordia - University of Perugia, Perugia, Italy
| |
Collapse
|
61
|
Farotti L, Paciotti S, Tasegian A, Eusebi P, Parnetti L. Discovery, validation and optimization of cerebrospinal fluid biomarkers for use in Parkinson's disease. Expert Rev Mol Diagn 2017; 17:771-780. [PMID: 28604235 DOI: 10.1080/14737159.2017.1341312] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Parkinson's disease (PD) is a complex and phenotypically heterogeneous neurodegenerative disease, for which the diagnosis is mainly based on clinical parameters (even if neuroimaging plays a role in diagnostic assessment); as a consequence, misdiagnosis is common, especially in early stages. Thus, there is an urgent need of having available biomarkers in order to achieve an early and accurate diagnosis. Since molecular changes in the brain are reliably and timely reflected in cerebrospinal fluid (CSF), CSF represents an ideal source for biomarkers of different pathophysiological processes characterizing the disease since its early phases. Areas covered: The aim of this review is to provide an update on the role, development and validation of most studied CSF biomarkers showing a role in the diagnosis and/or prognosis of PD. Oligomeric alpha-synuclein, DJ-1, lysosomal enzymes (namely, glucocerebrosidase) show consistent evidence as potential diagnostic biomarkers of PD. Neurofilament light chain may also have a significant role in differentiating PD from other parkinsonisms. Amyloid beta peptide 1-42 has consistently shown a prognostic value in terms of development of cognitive impairment and dementia in PD patients. Expert commentary: CSF biomarkers represent a very promising approach to early and differential diagnosis of PD. The biomarkers available so far need preanalytical and analytical validation in order to have these CSF biomarkers ready for clinical use.
Collapse
Affiliation(s)
- Lucia Farotti
- a Clinica Neurologica, Laboratorio di Neurochimica Clinica , Universita degli Studi di Perugia , Perugia , Italy
| | - Silvia Paciotti
- a Clinica Neurologica, Laboratorio di Neurochimica Clinica , Universita degli Studi di Perugia , Perugia , Italy
| | - Anna Tasegian
- a Clinica Neurologica, Laboratorio di Neurochimica Clinica , Universita degli Studi di Perugia , Perugia , Italy
| | - Paolo Eusebi
- a Clinica Neurologica, Laboratorio di Neurochimica Clinica , Universita degli Studi di Perugia , Perugia , Italy
| | - Lucilla Parnetti
- a Clinica Neurologica, Laboratorio di Neurochimica Clinica , Universita degli Studi di Perugia , Perugia , Italy
| |
Collapse
|
62
|
Delgado-Alvarado M, Gago B, Gorostidi A, Jiménez-Urbieta H, Dacosta-Aguayo R, Navalpotro-Gómez I, Ruiz-Martínez J, Bergareche A, Martí-Massó JF, Martínez-Lage P, Izagirre A, Rodríguez-Oroz MC. Tau/α-synuclein ratio and inflammatory proteins in Parkinson's disease: An exploratory study. Mov Disord 2017; 32:1066-1073. [DOI: 10.1002/mds.27001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/01/2017] [Accepted: 03/03/2017] [Indexed: 01/13/2023] Open
Affiliation(s)
- Manuel Delgado-Alvarado
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute; San Sebastián Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
| | - Belén Gago
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute; San Sebastián Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
| | - Ana Gorostidi
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute; San Sebastián Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
- Genomics Platform, Biodonostia Research Institute; San Sebastián Spain
| | - Haritz Jiménez-Urbieta
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute; San Sebastián Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
| | - Rosalía Dacosta-Aguayo
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute; San Sebastián Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
| | - Irene Navalpotro-Gómez
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute; San Sebastián Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
| | - Javier Ruiz-Martínez
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute; San Sebastián Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
- Movement Disorders Unit, Department of Neurology, University Hospital Donostia; San Sebastián Spain
| | - Alberto Bergareche
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute; San Sebastián Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
- Movement Disorders Unit, Department of Neurology, University Hospital Donostia; San Sebastián Spain
| | - José F. Martí-Massó
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute; San Sebastián Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
- Movement Disorders Unit, Department of Neurology, University Hospital Donostia; San Sebastián Spain
| | | | - Andrea Izagirre
- Department of Neurology, CITA-Alzheimer Foundation; San Sebastián Spain
| | - María C. Rodríguez-Oroz
- Neurodegenerative Disorders Area, Biodonostia Health Research Institute; San Sebastián Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
- Movement Disorders Unit, Department of Neurology, University Hospital Donostia; San Sebastián Spain
- Ikerbasque (Basque Foundation for Science); Bilbao Spain
- Basque Center on Cognition Brain and Language (BCBL); San Sebastián Spain
| |
Collapse
|
63
|
Johar I, Mollenhauer B, Aarsland D. Cerebrospinal Fluid Biomarkers of Cognitive Decline in Parkinson's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 132:275-294. [PMID: 28554411 DOI: 10.1016/bs.irn.2016.12.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Among the nonmotor symptoms in Parkinson's disease (PD), cognitive impairment is one of the most common and devastating. Over recent years, mild cognitive impairment (MCI) has become a recognized feature of PD (PD-MCI). The underlying mechanisms which influence onset, rate of decline, and conversion to dementia (PDD) are largely unknown. Adding to this uncertainty is the heterogeneity of cognitive domains affected. Currently there are no disease-modifying treatments that can slow or reverse this process. Identification of biomarkers that can predict rate and risk of cognitive decline is therefore an unmet need. Cerebrospinal fluid (CSF) is an ideal biomarker candidate as its constituents reflect the metabolic processes underlying the functioning of brain parenchyma. The pathological hallmark of PD is the presence of aggregated α-synuclein (α-Syn) in intracellular Lewy inclusions. In addition, there is concomitant Alzheimer's disease (AD) pathology. In AD, decreased CSF β-amyloid 1-42 (Aβ42) and increased CSF tau levels are predictive of future cognitive decline, setting a precedent for such studies to be carried out in PD. CSF studies in PD have focused on the classical AD biomarkers and α-Syn. Longitudinal studies indicate that low levels of CSF Aβ42 are predictive of cognitive decline; however, results for tau and α-Syn were not consistent. This chapter summarizes recent findings of CSF biomarker studies and cognitive dysfunction in PD.
Collapse
Affiliation(s)
- Iskandar Johar
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany; University Medical Center, Göttingen, Germany
| | - Dag Aarsland
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| |
Collapse
|
64
|
Cho YE, Lee MH, Song BJ. Neuronal Cell Death and Degeneration through Increased Nitroxidative Stress and Tau Phosphorylation in HIV-1 Transgenic Rats. PLoS One 2017; 12:e0169945. [PMID: 28107387 PMCID: PMC5249108 DOI: 10.1371/journal.pone.0169945] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/27/2016] [Indexed: 12/16/2022] Open
Abstract
The underlying mechanisms for increased neurodegeneration and neurocognitive deficits in HIV-infected people are unclear. Therefore, this study was aimed to investigate the mechanisms of increased neurodegeneration in 5-month old male HIV-1 Transgenic (Tg) rats compared to the age- and gender-matched wild-type (WT) by evaluating histological changes and biochemical parameters of the key proteins involved in the cell death signaling and apoptosis. Histological and immunohistochemical analyses revealed decreased neuronal cells with elevated astrogliosis in HIV-1 Tg rats compared to WT. Mechanistic studies revealed that increased levels of nitroxidative stress marker proteins such as NADPH-oxidase, cytochrome P450-2E1 (CYP2E1), inducible nitric oxide synthase (iNOS), the stress-activated mitogen-activated protein kinases such as JNK and p38K, activated cell-cycle dependent CDK5, hypoxia-inducible protein-1α, nitrated proteins, hyperphosphorylated tau, and amyloid plaques in HIV-Tg rats were consistently observed in HIV-1 Tg rats. Confocal microscopy and cell viability analyses showed that treatment with an antioxidant N-acetylcysteine or a specific inhibitor of iNOS 1400W significantly prevented the increased apoptosis of neuro-2A cells by HIV-1 Tat or gp120 protein, demonstrating the causal role of HIV-1 mediated nitroxidative stress and protein nitration in promoting neuronal cell death. Immunoprecipitation and immunoblot analysis confirmed nitration of Hsp90, evaluated as an example of nitrated proteins, suggesting possible involvement of nitrated proteins in neuronal damage. Further, activated p-JNK directly binds tau and phosphorylates multiple amino acids, suggesting an important role of p-JNK in tau hyperphosphorylation and tauopathy. These changes were accompanied with elevated levels of many apoptosis-related proteins Bax and cleaved (activated) caspase-3 as well as proinflammatory cytokines including TNF-α, IL-6 and MCP-1. Collectively, these results indicate that raised nitroxidative stress accompanied by elevated inflammation, cell death signaling pathway including activated p-JNK, C-terminal C99 amyloid fragment formation and tau hyperphosphorylation are responsible for increased apoptosis of neuronal cells and neurodegeneration in 5-month old HIV-Tg rats.
Collapse
Affiliation(s)
- Young-Eun Cho
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, United States of America
| | - Myoung-Hwa Lee
- Office of the Clinical Director, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States of America
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
65
|
Brennan L, Devlin KM, Xie SX, Mechanic-Hamilton D, Tran B, Hurtig HH, Chen-Plotkin A, Chahine LM, Morley JF, Duda JE, Roalf DR, Dahodwala N, Rick J, Trojanowski JQ, Moberg PJ, Weintraub D. Neuropsychological Subgroups in Non-Demented Parkinson's Disease: A Latent Class Analysis. JOURNAL OF PARKINSON'S DISEASE 2017; 7:385-395. [PMID: 28387684 PMCID: PMC5548408 DOI: 10.3233/jpd-171081] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Methods to detect early cognitive decline and account for heterogeneity of deficits in Parkinson's disease (PD) are needed. Quantitative methods such as latent class analysis (LCA) offer an objective approach to delineate discrete phenotypes of impairment. OBJECTIVE To identify discrete neurocognitive phenotypes in PD patients without dementia. METHODS LCA was applied to a battery of 8 neuropsychological measures to identify cognitive subtypes in a cohort of 199 non-demented PD patients. Two measures were analyzed from each of four domains: executive functioning, memory, visuospatial abilities, and language. Additional analyses compared groups on clinical characteristics and cognitive diagnosis. RESULTS LCA identified 3 distinct groups of PD patients: an intact cognition group (54.8%), an amnestic group (32.2%), and a mixed impairment group with dysexecutive, visuospatial and lexical retrieval deficits (13.1%). The two impairment groups had significantly lower instrumental activities of daily living ratings and greater motor symptoms than the intact group. Of those diagnosed as cognitively normal according to MDS criteria, LCA classified 23.2% patients as amnestic and 9.9% as mixed cognitive impairment. CONCLUSIONS Non-demented PD patients exhibit distinct neuropsychological profiles. One-third of patients with LCA-determined impairment were diagnosed as cognitively intact by expert consensus, indicating that classification using a statistical algorithm may improve detection of initial and subtle cognitive decline. This study also demonstrates that memory impairment is common in non-demented PD even when cognitive impairment is not clinically apparent. This study has implications for predicting eventual emergence of significant cognitive decline, and treatment trials for cognitive dysfunction in PD.
Collapse
Affiliation(s)
- Laura Brennan
- Department of Neurology, Thomas Jefferson University Hospital, Philadelphia, PA
| | | | - Sharon X. Xie
- Department of Biostatistics and Epidemiology, University of Pennsylvania School of Medicine; Philadelphia, PA
| | - Dawn Mechanic-Hamilton
- Department of Psychiatry, University of Pennsylvania School of Medicine; Philadelphia, PA
| | - Baochan Tran
- Department of Psychology, Widener University
- Department of Neurology, University of Pennsylvania School of Medicine; Philadelphia, PA
| | - Howard H. Hurtig
- Department of Neurology, University of Pennsylvania School of Medicine; Philadelphia, PA
| | - Alice Chen-Plotkin
- Department of Neurology, University of Pennsylvania School of Medicine; Philadelphia, PA
| | - Lama M. Chahine
- Department of Neurology, University of Pennsylvania School of Medicine; Philadelphia, PA
| | - James F. Morley
- Department of Neurology, University of Pennsylvania School of Medicine; Philadelphia, PA
- Parkinson's Disease Research, Education, and Clinical Center, Philadelphia Veterans Affairs Medical Center; Philadelphia, PA
| | - John E. Duda
- Department of Neurology, University of Pennsylvania School of Medicine; Philadelphia, PA
- Parkinson's Disease Research, Education, and Clinical Center, Philadelphia Veterans Affairs Medical Center; Philadelphia, PA
| | - David R. Roalf
- Department of Psychiatry, University of Pennsylvania School of Medicine; Philadelphia, PA
| | - Nabila Dahodwala
- Department of Neurology, University of Pennsylvania School of Medicine; Philadelphia, PA
| | - Jacqueline Rick
- Department of Neurology, University of Pennsylvania School of Medicine; Philadelphia, PA
| | - John Q. Trojanowski
- Department of Pathology, and Laboratory Medicine, University of Pennsylvania School of Medicine; Philadelphia, PA
| | - Paul J. Moberg
- Department of Psychiatry, University of Pennsylvania School of Medicine; Philadelphia, PA
| | - Daniel Weintraub
- Department of Psychiatry, University of Pennsylvania School of Medicine; Philadelphia, PA
- Department of Neurology, University of Pennsylvania School of Medicine; Philadelphia, PA
- Parkinson's Disease Research, Education, and Clinical Center, Philadelphia Veterans Affairs Medical Center; Philadelphia, PA
- Mental Illness Research, Education, and Clinical Center, Philadelphia Veterans Affairs Medical Center; Philadelphia, PA
| |
Collapse
|
66
|
Biomarkers of Nonmotor Symptoms in Parkinson's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 133:259-289. [DOI: 10.1016/bs.irn.2017.05.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
67
|
Modreanu R, Cerquera SC, Martí MJ, Ríos J, Sánchez-Gómez A, Cámara A, Fernández M, Compta Y. Cross-sectional and longitudinal associations of motor fluctuations and non-motor predominance with cerebrospinal τ and Aβ as well as dementia-risk in Parkinson's disease. J Neurol Sci 2016; 373:223-229. [PMID: 28131192 DOI: 10.1016/j.jns.2016.12.064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 12/19/2016] [Accepted: 12/28/2016] [Indexed: 10/20/2022]
Abstract
Experimental, neuropathological and cerebrospinal fluid (CSF) studies support τ and amyloid-β (Aβ) relevance in Parkinson's disease (PD) related dementia. Lesser motor fluctuations (MFs) and non-motor features have also been related to PD-dementia. Yet, little is known about the association of MFs and non-motor symptoms with CSF τ and Aβ in PD. We hypothesized that lesser MFs and non-motor predominance are related to these CSF markers and dementia-risk in PD. We studied 58 PD patients (dementia at baseline, n=21; dementia at 18-months, n=35) in whom CSF Aβ and τ had been determined with ELISA techniques. MFs and a number of non-motor symptoms (apathy, anxiety, irritability, depression, visual hallucinations, spatial disorientation, memory complaints) over disease course were dichotomized as absent-mild vs. moderate-severe by retrospective clinical chart review blind to CSF findings. Non-motor predominance was defined as ≥3 non-motor symptoms (after the cohort-median of non-motor symptoms per patient) with ≥2 being moderate-severe and ≥1 having been present from onset, with all these being more disabling overall than motor features. Cross-sectionally, CSF biomarkers were non-parametrically compared according to dichotomized MFs and non-motor predominance. Longitudinally, dementia was the outcome (dependent variable), CSF markers, MFs and non-motor predominance were the predictors (independent variables), and potential modifiers as age, sex, and memory complaints were the covariates in binary regression models. Absent-mild MFs were associated with higher CSF τ markers and shorter time-to-dementia, while non-motor predominance and decreasing CSF Aβ independently increased longitudinal dementia-risk. In summary, absent-mild MFs, non-motor predominance and CSF τ and Aβ might define endophenotypes related to the timing or risk of dementia in PD.
Collapse
Affiliation(s)
- Raluca Modreanu
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, ICN, Hospital Clínic, IDIBAPS, CIBERNED, University of Barcelona, Barcelona, Catalonia, Spain; Parkinson's Disease and Movement Disorders Unit, Neurology Service, Segeberger Kliniken, Bad Segeberg, Germany
| | - Sonia Catalina Cerquera
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, ICN, Hospital Clínic, IDIBAPS, CIBERNED, University of Barcelona, Barcelona, Catalonia, Spain; Neurology Unit, Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - María José Martí
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, ICN, Hospital Clínic, IDIBAPS, CIBERNED, University of Barcelona, Barcelona, Catalonia, Spain
| | - José Ríos
- Statistics and Methodologic Support Unit, Unitat d'Avaluació, Suport i Prevenció (UASP), Hospital Clínic, IDIBAPS, Barcelona, Catalonia, Spain
| | - Almudena Sánchez-Gómez
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, ICN, Hospital Clínic, IDIBAPS, CIBERNED, University of Barcelona, Barcelona, Catalonia, Spain
| | - Ana Cámara
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, ICN, Hospital Clínic, IDIBAPS, CIBERNED, University of Barcelona, Barcelona, Catalonia, Spain
| | - Manel Fernández
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, ICN, Hospital Clínic, IDIBAPS, CIBERNED, University of Barcelona, Barcelona, Catalonia, Spain
| | - Yaroslau Compta
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, ICN, Hospital Clínic, IDIBAPS, CIBERNED, University of Barcelona, Barcelona, Catalonia, Spain.
| |
Collapse
|
68
|
Buongiorno M, Antonelli F, Compta Y, Fernandez Y, Pavia J, Lomeña F, Ríos J, Ramírez I, García JR, Soler M, Cámara A, Fernández M, Basora M, Salazar F, Sanchez-Etayo G, Valldeoriola F, Barrio JR, Marti MJ. Cross-Sectional and Longitudinal Cognitive Correlates of FDDNP PET and CSF Amyloid-β and Tau in Parkinson’s Disease1. J Alzheimers Dis 2016; 55:1261-1272. [DOI: 10.3233/jad-160698] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Mariateresa Buongiorno
- Parkinson’s Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic, Spain Instituto de Investigaciones Biomédicas August Pi i Sunyer IDIBAPS, Barcelona, Catalonia, Spain CIBER
| | - Francesca Antonelli
- Parkinson’s Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic, Spain Instituto de Investigaciones Biomédicas August Pi i Sunyer IDIBAPS, Barcelona, Catalonia, Spain CIBER
| | - Yaroslau Compta
- Parkinson’s Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic, Spain Instituto de Investigaciones Biomédicas August Pi i Sunyer IDIBAPS, Barcelona, Catalonia, Spain CIBER
| | | | - Javier Pavia
- Nuclear Medicine Department, Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona, Catalonia, Spain
- Biomedical Research Networking Centre on Bioengineering, Biomaterials and. Nanomedicine (CIBER-BBN), Barcelona, Catalonia, Spain
| | - Francisco Lomeña
- Nuclear Medicine Department, Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona, Catalonia, Spain
- Biomedical Research Networking Centre for Mental Health (CIBERSAM), Barcelona, Catalonia, Spain
| | - José Ríos
- Medical Statistics Core Facility, IDIBAPS, (Hospital Clinic), Barcelona, Spain. Biostatistics Unit, Faculty of Medicine, Universitat Autònoma de Barcelona. Catalonia, Spain
| | | | | | - Marina Soler
- CETIR Nuclear Medicine Esplugues de Llobregat, Catalonia, Spain
| | - Ana Cámara
- Parkinson’s Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic, Spain Instituto de Investigaciones Biomédicas August Pi i Sunyer IDIBAPS, Barcelona, Catalonia, Spain CIBER
| | - Manel Fernández
- Parkinson’s Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic, Spain Instituto de Investigaciones Biomédicas August Pi i Sunyer IDIBAPS, Barcelona, Catalonia, Spain CIBER
| | - Misericòrdia Basora
- Anaesthesiology Service, Hospital Clínic, IDIBAPS, Barcelona, Catalonia, Spain
| | - Fàtima Salazar
- Anaesthesiology Service, Hospital Clínic, IDIBAPS, Barcelona, Catalonia, Spain
| | | | - Francesc Valldeoriola
- Parkinson’s Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic, Spain Instituto de Investigaciones Biomédicas August Pi i Sunyer IDIBAPS, Barcelona, Catalonia, Spain CIBER
| | - Jorge Raúl Barrio
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California, USA
| | - Maria Jose Marti
- Parkinson’s Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic, Spain Instituto de Investigaciones Biomédicas August Pi i Sunyer IDIBAPS, Barcelona, Catalonia, Spain CIBER
| |
Collapse
|
69
|
Naturally Occurring Autoantibodies against Tau Protein Are Reduced in Parkinson's Disease Dementia. PLoS One 2016; 11:e0164953. [PMID: 27802290 PMCID: PMC5089716 DOI: 10.1371/journal.pone.0164953] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/04/2016] [Indexed: 11/23/2022] Open
Abstract
Background and Objective Altered levels of naturally occurring autoantibodies (nAbs) against disease-associated neuronal proteins have been reported for neurodegenerative diseases, such as Alzheimer's (AD) and Parkinson's disease (PD). Recent histopathologic studies suggest a contribution of both Lewy body- and AD-related pathology to Parkinson's disease dementia (PDD). Therefore, we explored nAbs against alpha-synuclein (αS), tau and β-amyloid (Aβ) in PDD compared to cognitively normal PD patients. Materials and Methods We established three different ELISAs to quantify the nAbs-tau, nAbs-αS, and nAbs-Aβ levels and avidity towards their specific antigen in serum samples of 18 non-demented (PDND) and 18 demented PD patients (PDD), which were taken from an ongoing multi-center cohort study (DEMPARK/LANDSCAPE). Results PDD patients had significantly decreased nAbs-tau serum levels compared to PDND patients (p = 0.007), whereas the serum titers of nAbs-αS and nAbs-Aβ were unchanged. For all three nAbs, no significant differences in avidity were found between PDD and PDND cohorts. However, within both patient groups, nAbs-tau showed lowest avidity to their antigen, followed by nAbs-αS, and nAbs-Aβ. Though, due to a high interassay coefficient of variability and the exclusion of many samples below the limit of detection, conclusions for nAbs-Aβ are only conditionally possible. Conclusion We detected a significantly decreased nAbs-tau serum level in PDD patients, indicating a potential linkage between nAbs-tau serum titer and cognitive deficits in PD. Thus, further investigation in larger samples is justified to confirm our findings.
Collapse
|
70
|
Gao R, Zhang G, Chen X, Yang A, Smith G, Wong DF, Zhou Y. CSF Biomarkers and Its Associations with 18F-AV133 Cerebral VMAT2 Binding in Parkinson's Disease-A Preliminary Report. PLoS One 2016; 11:e0164762. [PMID: 27764160 PMCID: PMC5072678 DOI: 10.1371/journal.pone.0164762] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 09/30/2016] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Cerebrospinal fluid (CSF) biomarkers, such as α-synuclein (α-syn), amyloid beta peptide 1-42 (Aβ1-42), phosphorylated tau (181P) (p-tau), and total tau (t-tau), have long been associated with the development of Parkinson disease (PD) and other neurodegenerative diseases. In this investigation, we reported the assessment of CSF biomarkers and their correlations with vesicular monoamine transporter 2 (VMAT2) bindings measured with 18F-9-fluoropropyl-(+)-dihydrotetrabenazine (18F-AV133) that is being developed as a biomarker for PD. We test the hypothesis that monoaminergic degeneration was correlated with CSF biomarker levels in untreated PD patients. METHODS The available online data from the Parkinson's Progression Markers Initiative study (PPMI) project were collected and analyzed, which include demographic information, clinical evaluations, CSF biomarkers (α-syn, Aβ1-42, p-tau, and t-tau), 18F-AV133 brain PET, and T1 weighted MRIs. Region of interest (ROI) and voxel-wise Pearson correlation between standardized uptake value ratio (SUVR) and CSF biomarkers were calculated. RESULTS Our major findings are: 1) Compared with controls, CSF α-syn and tau levels decreased significantly in PD; 2) α-syn was closely correlated with Aβ1-42 and tau in PD, especially in early-onset patients; and 3) hypothesis-driven ROI analysis found a significant negative correlation between CSF Aβ1-42 levels and VMAT2 densities in post cingulate, left caudate, left anterior putamen, and left ventral striatum in PDs. CSF t-tau and p-tau levels were significantly negatively related to VMAT2 SUVRs in substantia nigra and left ventral striatum, respectively. Voxel-wise analysis showed that left caudate, parahippocampal gyrus, insula and temporal lobe were negatively correlated with Aβ1-42. In addition, superior frontal gyrus and transverse temporal gyrus were negatively correlated with CSF p-tau levels. CONCLUSION These results suggest that monoaminergic degeneration in PD is correlated with CSF biomarkers associated with cognitive impairment in neurodegenerative diseases including Alzheimer's disease. The association between loss of dopamine synaptic function and pathologic protein accumulations in PD indicates an important role of CSF biomarkers in PD development.
Collapse
Affiliation(s)
- Rui Gao
- Department of Nuclear Medicine, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi 710061, China
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States of America
| | - Guangjian Zhang
- Department of Surgery, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xueqi Chen
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States of America
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Aimin Yang
- Department of Nuclear Medicine, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Gwenn Smith
- Division of Geriatric Psychiatry and Neuropsychiatry, Johns Hopkins Bayview Medical Center, Baltimore, Maryland 21287, United States of America
| | - Dean F. Wong
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States of America
- Department of Psychiatry, Johns Hopkins University, Baltimore, Maryland 21205, United States of America
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21205, United States of America
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21205, United States of America
| | - Yun Zhou
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States of America
| |
Collapse
|
71
|
McMillan CT, Wolk DA. Presence of cerebral amyloid modulates phenotype and pattern of neurodegeneration in early Parkinson's disease. J Neurol Neurosurg Psychiatry 2016; 87:1112-22. [PMID: 27288043 PMCID: PMC5154297 DOI: 10.1136/jnnp-2015-312690] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 05/11/2016] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To evaluate the frequency of cerebral amyloid in early Parkinson's disease (ePD) and provide a multimodal assessment of the influence of cerebral amyloid on disease phenotype. METHODS We performed a multicentre cohort study of the Parkinson's Progression Markers Initiative (PPMI), including 369 drug-naïve patients with ePD and 174 healthy controls (HC). Cerebrospinal fluid (CSF) amyloid-β levels were transformed using the linear regression procedure. A cut-off of >198 pg/mL was used to define amyloid-negative (PD-) and amyloid-positive (PD+) subgroups. Grey matter (GM) density and hippocampal volume from the MRI was measured using Advanced Normalisation Tools (ANTs). We compared demographic, genetic, CSF, behavioural, functional and imaging modalities across ePD- and ePD+ groups. RESULTS We observed that 16.5% of ePD have CSF evidence of amyloidosis. PD+ was significantly older than PD-, had a higher frequency of APOE e4 alleles and all CSF measures (total-tau, phosphorylated-tau and α-synuclein) were reduced. PD+ had reduced cognitive performance relative to PD- on Symbol-Digit Matching, Verbal Category Fluency and Delayed Recall tests. Imaging analysis in a subset of individuals (PD+ =43; PD- =241) revealed overlapping GM atrophy relative to HC in medial temporal, frontal and brainstem structures. Direct comparisons revealed PD+ GM reductions predominantly located in the frontal cortex while PD- had GM reductions in subcortical structures. These observations remain when controlling for age and APOE e4 allele status. CONCLUSIONS Cerebral amyloid in ePD yields a unique phenotype across all measured modalities that is consistent with a synergistic interaction between α-synuclein and amyloid pathology. Amyloid status should be considered when screening these individuals for trials involving disease-modifying agents.
Collapse
Affiliation(s)
- Corey T McMillan
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
72
|
Skogseth RE, Bronnick K, Pereira JB, Mollenhauer B, Weintraub D, Fladby T, Aarsland D. Associations between Cerebrospinal Fluid Biomarkers and Cognition in Early Untreated Parkinson's Disease. JOURNAL OF PARKINSONS DISEASE 2016; 5:783-92. [PMID: 26599300 DOI: 10.3233/jpd-150682] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Mild cognitive impairment and dementia are common, clinically important features of Parkinson's disease (PD). The underlying disease pathology is heterogeneous and not yet well characterized. Biomarkers for cognitive impairment in PD could aid in diagnostic and prognostic evaluation and in the development of new cognitive enhancing treatments. OBJECTIVE To examine the relationship between CSF markers and cognition in a large, multicenter, cohort study of early, untreated PD, and compare marker concentrations between PD patients with and without MCI and healthy, age-matched controls. METHODS 414 early, untreated PD (34% with mild cognitive impairment) and 189 healthy, cognitively intact controls with baseline neuropsychological testing and CSF abeta42, t-tau, p-tau181 and α-synuclein results were included. Multiple linear regression models were constructed with a composite cognition factor, or memory-, or visuospatial- or executive-attention domains as dependent variables, and CSF markers, demographic characteristics and MDS-UPDRS III score as predictors. RESULTS Lower α-synuclein was associated with reduced performance on the executive-attention domain and the composite cognition factor in the whole PD-group. Abeta42 was significantly decreased in PD with mild cognitive impairment compared with controls after adjusting for covariates, while values in PD without MCI were identical to healthy controls. CONCLUSIONS The association between reduced CSF α-synuclein concentrations and cognition suggests that α-synuclein pathology contributes to early cognitive impairment in PD, in particular to executive-attentional dysfunction. Longitudinal analyses are needed to determine if this and other CSF biomarkers in early Parkinson's disease are associated with the risk of future cognitive decline and dementia.
Collapse
Affiliation(s)
- Ragnhild E Skogseth
- Haraldsplass Deaconess Hospital, Kavli Research Center for Geriatrics and Dementia, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Norway
| | - Kolbjorn Bronnick
- TIPS -Regional Center for Clinical Research in Psychosis, Stavanger University Hospital, Norway.,Faculty of Social Sciences, University of Stavanger, Norway
| | - Joana B Pereira
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute H1, Division of Clinical Geriatrics, Sweden
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik and Department of Neuropathology, University Medical Center Göttingen, Germany
| | - Daniel Weintraub
- Department of Psychiatry and Neurology, Perelman School of Medicine at the University of Pennsylvania; Philadelphia VA Medical Center, Philadelphia, PA, USA
| | - Tormod Fladby
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, Campus Ahus, University of Oslo, Norway
| | - Dag Aarsland
- Center forAlzheimer's Disease Research, Department of Neurobiology, CareSciences and Society, Karolinska Institutet H1, Division of Neurogeriatrics, Sweden.,Center for Age-Related Medicine Department of Psychiatry, Stavanger University Hospital, Norway
| |
Collapse
|
73
|
Compta Y, Buongiorno M, Bargalló N, Valldeoriola F, Muñoz E, Tolosa E, Ríos J, Cámara A, Fernández M, Martí MJ. White matter hyperintensities, cerebrospinal amyloid-β and dementia in Parkinson's disease. J Neurol Sci 2016; 367:284-90. [PMID: 27423605 DOI: 10.1016/j.jns.2016.06.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/24/2016] [Accepted: 06/03/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Yaroslau Compta
- Parkinson disease & Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències (ICN), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Mariateresa Buongiorno
- Parkinson disease & Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències (ICN), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Núria Bargalló
- Magnetic Resonance Unit, Neurorradiology Section, Centre de Diagnòstic per la Imatge (CDI), IDIBAPS, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Francesc Valldeoriola
- Parkinson disease & Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències (ICN), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Esteban Muñoz
- Parkinson disease & Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències (ICN), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Eduardo Tolosa
- Parkinson disease & Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències (ICN), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - José Ríos
- Statistics and Methodologic Support Unit, Unitat d'Avaluació, Suport i Prevenció (UASP), Hospital Clínic, IDIBAPS, Barcelona, Catalonia, Spain
| | - Ana Cámara
- Parkinson disease & Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències (ICN), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Manel Fernández
- Parkinson disease & Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències (ICN), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Maria J Martí
- Parkinson disease & Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències (ICN), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain.
| |
Collapse
|
74
|
Hall S, Surova Y, Öhrfelt A, Blennow K, Zetterberg H, Hansson O. Longitudinal Measurements of Cerebrospinal Fluid Biomarkers in Parkinson's Disease. Mov Disord 2016; 31:898-905. [PMID: 26878815 PMCID: PMC5067556 DOI: 10.1002/mds.26578] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 12/28/2015] [Accepted: 01/25/2016] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE The purpose of this study was to investigate whether cerebrospinal fluid (CSF) levels of tau, phosphorylated tau, β-amyloid42 , α-synuclein, neurofilament light, and YKL-40 change over time and if changes correlate with motor progression and/or cognitive decline in patients with PD and controls. METHODS We included 63 patients with PD (nondemented) and 21 neurologically healthy controls from the prospective and longitudinal Swedish BioFINDER study, all of whom had clinical assessments and lumbar punctures at baseline and after 2 years. RESULTS CSF tau levels correlated strongly with α-synuclein. The levels of CSF α-synuclein, tau, phosphorylated tau, neurofilament light, and YKL-40, but not β-amyloid42 , increased in CSF over 2 years in PD. No changes were seen in the control group. Studying patients with a short disease duration ( ≤ 5 years) and patients with a long disease duration ( > 5 years) separately, α-synuclein and tau only increased in the PD group with long disease duration. In the PD group, an increase in phosphorylated tau over 2 years correlated with faster motor progression and faster cognitive decline. An increase in YKL-40 over 2 years correlated with faster cognitive decline. CONCLUSION CSF biomarkers reflecting Lewy body pathology and neurodegeneration (α-synuclein), neuronal degeneration (tau, phosphorylated tau, and neurofilament light), and inflammation (YKL-40) increase significantly over 2 years in PD. CSF levels of α-synuclein and tau correlate and remain stable in the early symptomatic phase of PD but increase in the later phase. We hypothesize that CSF α-synuclein levels might increase as a result of more intense neurodegeneration in PD with long disease duration. © 2016 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sara Hall
- Department of NeurologySkåne University HospitalMalmöSweden
- Department of Clinical SciencesLund UniversityMalmöSweden
| | - Yulia Surova
- Department of NeurologySkåne University HospitalMalmöSweden
- Department of Clinical SciencesLund UniversityMalmöSweden
| | - Annika Öhrfelt
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of GothenburgGothenburg and MölndalSweden
| | | | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of GothenburgGothenburg and MölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of GothenburgGothenburg and MölndalSweden
- University College London Institute of NeurologyLondonUnited Kingdom
| | - Oskar Hansson
- Department of Clinical SciencesLund UniversityMalmöSweden
- Memory ClinicSkåne University HospitalMalmöSweden
| |
Collapse
|
75
|
Kang JH. Cerebrospinal Fluid Amyloid β1-42, Tau, and Alpha-Synuclein Predict the Heterogeneous Progression of Cognitive Dysfunction in Parkinson's Disease. J Mov Disord 2016; 9:89-96. [PMID: 27240810 PMCID: PMC4886208 DOI: 10.14802/jmd.16017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 04/20/2016] [Indexed: 01/13/2023] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease with heterogeneous pathological and clinical features. Cognitive dysfunction, a frequent non-motor complication, is a risk factor for poor prognosis and shows inter-individual variation in its progression. Of the clinical studies performed to identify biomarkers of PD progression, the Parkinson’s Progression Markers Initiative (PPMI) study is the largest study that enrolled drug-naïve and very early stage PD patients. The baseline characteristics of the PPMI cohort were recently published. The diagnostic utility of cerebrospinal fluid (CSF) biomarkers, including alpha-synuclein (α-syn), total tau, phosphorylated tau at Thr181, and amyloid β1-42, was not satisfactory. However, the baseline data on CSF biomarkers in the PPMI study suggested that the measurement of the CSF biomarkers enables the prediction of future cognitive decline in PD patients, which was consistent with previous studies. To prove the hypothesis that the interaction between Alzheimer’s pathology and α-syn pathology is important to the progression of cognitive dysfunction in PD, longitudinal observational studies must be followed. In this review, the neuropathological nature of heterogeneous cognitive decline in PD is briefly discussed, followed by a summarized interpretation of baseline CSF biomarkers derived from the data in the PPMI study. The combination of clinical, biochemical, genetic and imaging biomarkers of PD constitutes a feasible strategy to predict the heterogeneous progression of PD.
Collapse
Affiliation(s)
- Ju-Hee Kang
- Department of Pharmacology and Medicinal Toxicology Research Center, Hypoxia-related Disease Research Center, Inha University School of Medicine, Incheon, Korea
| |
Collapse
|
76
|
Delgado-Alvarado M, Gago B, Navalpotro-Gomez I, Jiménez-Urbieta H, Rodriguez-Oroz MC. Biomarkers for dementia and mild cognitive impairment in Parkinson's disease. Mov Disord 2016; 31:861-81. [PMID: 27193487 DOI: 10.1002/mds.26662] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 04/15/2016] [Accepted: 04/18/2016] [Indexed: 12/27/2022] Open
Abstract
Cognitive decline is one of the most frequent and disabling nonmotor features of Parkinson's disease. Around 30% of patients with Parkinson's disease experience mild cognitive impairment, a well-established risk factor for the development of dementia. However, mild cognitive impairment in patients with Parkinson's disease is a heterogeneous entity that involves different types and extents of cognitive deficits. Because it is not currently known which type of mild cognitive impairment confers a higher risk of progression to dementia, it would be useful to define biomarkers that could identify these patients to better study disease progression and possible interventions. In this sense, the identification among patients with Parkinson's disease and mild cognitive impairment of biomarkers associated with dementia would allow the early detection of this process. This review summarizes studies from the past 25 years that have assessed the potential biomarkers of dementia and mild cognitive impairment in Parkinson's disease patients. Despite the potential importance, no biomarker has as yet been validated. However, features such as low levels of epidermal and insulin-like growth factors or uric acid in plasma/serum and of Aß in CSF, reduction of cerebral cholinergic innervation and metabolism measured by PET mainly in posterior areas, and hippocampal atrophy in MRI might be indicative of distinct deficits with a distinct risk of dementia in subgroups of patients. Longitudinal studies combining the existing techniques and new approaches are needed to identify patients at higher risk of dementia. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Manuel Delgado-Alvarado
- Biodonostia Health Research Institute, San Sebastián, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Belén Gago
- Biodonostia Health Research Institute, San Sebastián, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Irene Navalpotro-Gomez
- Biodonostia Health Research Institute, San Sebastián, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Haritz Jiménez-Urbieta
- Biodonostia Health Research Institute, San Sebastián, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - María C Rodriguez-Oroz
- Biodonostia Health Research Institute, San Sebastián, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Neurology Department, University Hospital Donostia, San Sebastián, Spain.,Ikerbasque (Basque Foundation for Science), Bilbao, Spain.,Basque Center on Cognition, Brain and Language (BCBL), San Sebastián, Spain.,Physiology Department, Medical School University of Navarra, Pamplona, Spain
| |
Collapse
|
77
|
Blennow K, Biscetti L, Eusebi P, Parnetti L. Cerebrospinal fluid biomarkers in Alzheimer's and Parkinson's diseases-From pathophysiology to clinical practice. Mov Disord 2016; 31:836-47. [DOI: 10.1002/mds.26656] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/22/2016] [Accepted: 03/25/2016] [Indexed: 01/05/2023] Open
Affiliation(s)
- Kaj Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg; Mölndal Campus Mölndal Sweden
| | - Leonardo Biscetti
- Section of Neurology, Department of Medicine, Center for Memory Disturbances, University of Perugia; Sant'Andrea delle Fratte Perugia Italy
| | - Paolo Eusebi
- Section of Neurology, Department of Medicine, Center for Memory Disturbances, University of Perugia; Sant'Andrea delle Fratte Perugia Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine, Center for Memory Disturbances, University of Perugia; Sant'Andrea delle Fratte Perugia Italy
| |
Collapse
|
78
|
Vilas D, Shaw LM, Taylor P, Berg D, Brockmann K, Aasly J, Marras C, Pont-Sunyer C, Ríos J, Marek K, Tolosa E. Cerebrospinal fluid biomarkers and clinical features in leucine-rich repeat kinase 2 (LRRK2) mutation carriers. Mov Disord 2016; 31:906-14. [DOI: 10.1002/mds.26591] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 12/22/2022] Open
Affiliation(s)
- Dolores Vilas
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, Institut de Neurociències Hospital Clínic, University of Barcelona; Catalonia Spain
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine; Perelman School of Medicine, University of Pennsylvania; Philadelphia Pennsylvania USA
| | | | - Daniela Berg
- Department of Neurodegenerative Diseases and Hertie Institute for Clinical Brain Research; University of Tübingen; Tübingen Germany
| | - Kathrin Brockmann
- Department of Neurodegenerative Diseases and Hertie Institute for Clinical Brain Research; University of Tübingen; Tübingen Germany
| | - Jan Aasly
- Department of Neuroscience; Norwegian University of Science and Technology; Trondheim Norway
- Department of Neurology; St. Olav's Hospital; Trondheim Norway
| | - Connie Marras
- Morton and Gloria Shulman Movement Disorders Centre and the Edmond J Safra Program in Parkinson's Disease, Toronto Western Hospital; Toronto Ontario Canada
| | - Claustre Pont-Sunyer
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, Institut de Neurociències Hospital Clínic, University of Barcelona; Catalonia Spain
| | - José Ríos
- Biostatistics and Data Management Core Facility, IDIBAPS (Hospital Clinic); Barcelona Spain
- Biostatistics Unit, Faculty of Medicine, Universitat Autònoma de Barcelona; Catalonia Spain
| | - Ken Marek
- Institute for Neurodegenerative Disorders; New Haven Connecticut USA
| | - Eduardo Tolosa
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, Institut de Neurociències Hospital Clínic, University of Barcelona; Catalonia Spain
- Centro en Red para la Investigación de Enfermedades Neurodegenerativas CIBERNED; Spain
| |
Collapse
|
79
|
Fullard ME, Tran B, Xie SX, Toledo JB, Scordia C, Linder C, Purri R, Weintraub D, Duda JE, Chahine LM, Morley JF. Olfactory impairment predicts cognitive decline in early Parkinson's disease. Parkinsonism Relat Disord 2016; 25:45-51. [PMID: 26923521 DOI: 10.1016/j.parkreldis.2016.02.013] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/30/2016] [Accepted: 02/14/2016] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To evaluate the association between baseline olfaction and both cross-sectional and longitudinal cognitive assessments, motor symptoms, non-motor symptoms (NMS), and CSF biomarkers in early Parkinson's disease (PD). METHODS Parkinson's Progression Marker's Initiative (PPMI) participants underwent baseline olfactory testing with the University of Pennsylvania Smell Identification Test (UPSIT). Serial assessments included measures of motor symptoms, NMS, neuropsychological assessment, and CSF biomarkers. Up to three years follow-up data were included. RESULTS At baseline, worse olfaction (lowest tertile) was associated with more severe NMS, including anxiety and autonomic symptoms. Those in the lowest olfactory tertile were more likely to report cognitive impairment (37.4%) compared to those in the middle (24.4%) and highest olfactory tertiles (14.2%, p < 0.001). Aβ1-42 was significantly lower, and tau/Aβ1-42 ratio was higher in those with worse olfaction. In longitudinal analyses, lower UPSIT score was associated with greater decline in MoCA score (β = 0.02 [0.01, 0.03], p = 0.001) over time, as were composite measures of UPSIT score and either Aβ1-42 or tau/Aβ1-42 ratio. In a Cox proportional hazards model, a composite measure of olfaction and Aβ1-42 was a significant predictor of conversion from normal cognition to mild cognitive impairment (MCI; i.e., MoCA < 26), with subjects most impaired on both measures being 87% more likely to develop incident MCI (HR = 1.87 [1.16, 3.01], p = 0.01). CONCLUSIONS Worse baseline olfaction is associated with long-term cognitive decline. The addition of AD CSF biomarkers to olfactory testing may increase the likelihood of identifying those at highest risk for cognitive decline and progression to MCI.
Collapse
Affiliation(s)
- Michelle E Fullard
- Parkinson's Disease Research, Education, and Clinical Center (PADRECC), Philadelphia VA Medical Center, 3900 Woodland Ave, Philadelphia, PA 19104, USA
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Baochan Tran
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Sharon X Xie
- Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Jon B Toledo
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Christi Scordia
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Carly Linder
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Rachael Purri
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Daniel Weintraub
- Parkinson's Disease Research, Education, and Clinical Center (PADRECC), Philadelphia VA Medical Center, 3900 Woodland Ave, Philadelphia, PA 19104, USA
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - John E Duda
- Parkinson's Disease Research, Education, and Clinical Center (PADRECC), Philadelphia VA Medical Center, 3900 Woodland Ave, Philadelphia, PA 19104, USA
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Lama M Chahine
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - James F Morley
- Parkinson's Disease Research, Education, and Clinical Center (PADRECC), Philadelphia VA Medical Center, 3900 Woodland Ave, Philadelphia, PA 19104, USA
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| |
Collapse
|
80
|
Mollenhauer B, Parnetti L, Rektorova I, Kramberger MG, Pikkarainen M, Schulz-Schaeffer WJ, Aarsland D, Svenningsson P, Farotti L, Verbeek MM, Schlossmacher MG. Biological confounders for the values of cerebrospinal fluid proteins in Parkinson's disease and related disorders. J Neurochem 2016; 139 Suppl 1:290-317. [DOI: 10.1111/jnc.13390] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 09/11/2015] [Accepted: 09/21/2015] [Indexed: 12/26/2022]
Affiliation(s)
- Brit Mollenhauer
- Paracelsus-Elena-Klinik; Kassel Germany
- University Medical Center (Department of Neuropathology); Georg-August University Goettingen; Goettingen Germany
| | - Lucilla Parnetti
- Centro Disturbi della Memoria- Unità Valutativa Alzheimer; Clinica Neurologica; Università di Perugia; Perugia Italy
| | - Irena Rektorova
- Applied Neuroscience Group; CEITEC MU; Masaryk University; Brno Czech Republic
| | - Milica G. Kramberger
- Department of Neurology; University Medical Center Ljubljana; Ljubljana Slovenia
- Division for Neurogeriatrics; Department of NVS; Karolinska Institutet; Center for Alzheimer Research; Stockholm Sweden
- Centre for Age-Related Medicine; Stavanger University Hospital; Stavanger Norway
| | - Maria Pikkarainen
- Institute of Clinical Medicine / Neurology; University of Eastern Finland; Kuopio Finland
| | - Walter J. Schulz-Schaeffer
- University Medical Center (Department of Neuropathology); Georg-August University Goettingen; Goettingen Germany
| | - Dag Aarsland
- Division for Neurogeriatrics; Department of NVS; Karolinska Institutet; Center for Alzheimer Research; Stockholm Sweden
- Centre for Age-Related Medicine; Stavanger University Hospital; Stavanger Norway
| | - Per Svenningsson
- Department for Clinical Neuroscience; Karolinska Institute; Stockholm Sweden
| | - Lucia Farotti
- Centro Disturbi della Memoria- Unità Valutativa Alzheimer; Clinica Neurologica; Università di Perugia; Perugia Italy
| | - Marcel M. Verbeek
- Department of Neurology; Department of Laboratory Medicine; Donders Institute for Brain, Cognition and Behaviour; Radboud University Medical Centre; Nijmegen The Netherlands
| | - Michael G. Schlossmacher
- Program in Neuroscience and Division of Neurology; The Ottawa Hospital; University of Ottawa Brain & Mind Research Institute; Ottawa Ontario Canada
| |
Collapse
|
81
|
Zuo LJ, Yu SY, Wang F, Hu Y, Piao YS, Du Y, Lian TH, Wang RD, Yu QJ, Wang YJ, Wang XM, Chan P, Chen SD, Wang Y, Zhang W. Parkinson's Disease with Fatigue: Clinical Characteristics and Potential Mechanisms Relevant to α-Synuclein Oligomer. J Clin Neurol 2016; 12:172-80. [PMID: 26869370 PMCID: PMC4828563 DOI: 10.3988/jcn.2016.12.2.172] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/01/2015] [Accepted: 10/02/2015] [Indexed: 12/31/2022] Open
Abstract
Background and Purpose The aim of this study was to identify the clinical characteristics and potential mechanisms relevant to pathological proteins in Parkinson's disease (PD) patients who experience fatigue. Methods PD patients (n=102) were evaluated using a fatigue severity scale and scales for motor and nonmotor symptoms. The levels of three pathological proteins—α-synuclein oligomer, β-amyloid (Aβ)1-42, and tau—were measured in 102 cerebrospinal fluid (CSF) samples from these PD patients. Linear regression analyses were performed between fatigue score and the CSF levels of the above-listed pathological proteins in PD patients. Results The frequency of fatigue in the PD patients was 62.75%. The fatigue group had worse motor symptoms and anxiety, depression, and autonomic dysfunction. The CSF level of α-synuclein oligomer was higher and that of Aβ1-42 was lower in the fatigue group than in the non-fatigue group. In multiple linear regression analyses, fatigue severity was significantly and positively correlated with the α-synuclein oligomer level in the CSF of PD patients, after adjusting for confounders. Conclusions PD patients experience a high frequency of fatigue. PD patients with fatigue have worse motor and part nonmotor symptoms. Fatigue in PD patients is associated with an increased α-synuclein oligomer level in the CSF.
Collapse
Affiliation(s)
- Li Jun Zuo
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shu Yang Yu
- Department of Geriatrics, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fang Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yang Hu
- Department of Geriatrics, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ying Shan Piao
- Department of Geriatrics, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yang Du
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Teng Hong Lian
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Rui Dan Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qiu Jin Yu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ya Jie Wang
- Core Laboratory for Clinical Medical Research, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiao Min Wang
- Department of Physiology, Capital Medical University, Beijing, China
| | - Piu Chan
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China.,Department of Neurobiology, Beijing Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Sheng Di Chen
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yongjun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Wei Zhang
- Department of Geriatrics, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Beijing Key Laboratory on Parkinson's Disease, Beijing, China.
| |
Collapse
|
82
|
Leaver K, Poston KL. Do CSF Biomarkers Predict Progression to Cognitive Impairment in Parkinson's disease patients? A Systematic Review. Neuropsychol Rev 2015; 25:411-23. [PMID: 26626621 PMCID: PMC5152566 DOI: 10.1007/s11065-015-9307-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 11/09/2015] [Indexed: 12/01/2022]
Abstract
Many patients with Parkinson's disease (PD) will develop cognitive impairment. Cross-sectional studies have shown that certain protein levels are altered in the cerebrospinal fluid (CSF) of PD patients with dementia and are thought to represent potential biomarkers of underlying pathogenesis. Recent studies suggest that CSF biomarker levels may be predictive of future risk of cognitive decline in non-demented PD patients. However, the strength of this evidence and difference between specific CSF biomarkers is not well delineated. We therefore performed a systematic review to assess if levels of specific CSF protein biomarkers are predictive of progression to cognitive impairment. Nine articles were identified that met inclusion criteria for the review. Findings from the review suggest a convergence of evidence that a low baseline Aβ42 in the CSF of non-demented PD patients predicts development of cognitive impairment over time. Conversely, there is limited evidence that CSF levels of tau, either total tau or phosphorylated tau, is a useful predictive biomarker. There are mixed results for other CSF biomarkers such as α-synuclein, Neurofilament light chain, and Heart fatty acid-binding protein. Overall the results of this review show that certain CSF biomarkers have better predictive ability to identify PD patients who are at risk for developing cognitive impairment. Given the interest in developing disease-modifying therapies, identifying this group will be important for clinical trials as initiation of therapy prior to the onset of cognitive decline is likely to be more efficacious.
Collapse
Affiliation(s)
- Katherine Leaver
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Kathleen L Poston
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
- Department of Neurosurgery, Stanford University, Stanford, CA, USA.
| |
Collapse
|
83
|
Cavaleri F. Review of Amyotrophic Lateral Sclerosis, Parkinson’s and Alzheimer’s diseases helps further define pathology of the novel paradigm for Alzheimer’s with heavy metals as primary disease cause. Med Hypotheses 2015; 85:779-90. [DOI: 10.1016/j.mehy.2015.10.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/25/2015] [Accepted: 10/11/2015] [Indexed: 01/07/2023]
|
84
|
Goldman JG, Aggarwal NT, Schroeder CD. Mild cognitive impairment: an update in Parkinson's disease and lessons learned from Alzheimer's disease. Neurodegener Dis Manag 2015; 5:425-43. [PMID: 26517759 DOI: 10.2217/nmt.15.34] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Cognitive dysfunction is an important focus of research in Parkinson's disease (PD) and Alzheimer's disease (AD). While the concept of amnestic mild cognitive impairment (MCI) as a prodrome to AD has been recognized for many years, the construct of MCI in PD is a relative newcomer with recent development of diagnostic criteria, biomarker research programs and treatment trials. Controversies and challenges, however, regarding PD-MCI's definition, application, heterogeneity and different trajectories have arisen. This review will highlight current research advances and challenges in PD-MCI. Furthermore, lessons from the AD field, which has witnessed an evolution in MCI/AD definitions, relevant advances in biomarker research and development of disease-modifying and targeted therapeutic trials will be discussed.
Collapse
Affiliation(s)
- Jennifer G Goldman
- Rush University Medical Center, Department of Neurological Sciences, Section of Parkinson Disease & Movement Disorders, 1725 W. Harrison Street, Suite 755, Chicago, IL 60612, USA
| | - Neelum T Aggarwal
- Rush University Medical Center, Department of Neurological Sciences & Rush Alzheimer's Disease Center, 600 South Paulina, Suite 1038, Chicago, IL 60612, USA
| | - Cynthia D Schroeder
- Rush University Medical Center, Department of Neurological Sciences, 1735 W. Harrison Street, Suite 306, Chicago, IL 60612, USA
| |
Collapse
|
85
|
Sancesario GM, Bernardini S. How many biomarkers to discriminate neurodegenerative dementia? Crit Rev Clin Lab Sci 2015; 52:314-26. [PMID: 26292074 DOI: 10.3109/10408363.2015.1051658] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A number of cerebrospinal fluid (CSF) biomarkers are currently used for the diagnosis of dementia. Opposite changes in the level of amyloid-β(1-42) versus total tau and phosphorylated-tau181 in the CSF reflect the specific pathology of Alzheimer's disease (AD) in the brain. This panel of biomarkers has proven to be effective to differentiate AD from controls and from the major types of neurodegenerative dementia, and to evaluate the progression from mild cognitive impairment to AD. In the absence of specific biomarkers reflecting the pathologies of the other most common forms of dementia, such as Lewy Body disease, Frontotemporal lobar degeneration, Creutzfeldt-Jakob disease, etc., the evaluation of biomarkers of AD pathology is used, attempting to exclude rather than to confirm AD. Other biomarkers included in the common clinical practice do not clearly relate to the underlying pathology: progranulin (PGRN) is a selective marker of frontotemporal dementia with mutations in the PGRN gene; the 14-3-3 protein is a highly sensitive and specific marker for Creutzfeldt-Jakob disease, but has to be used carefully in differentiating rapid progressive dementia; and α-synuclein is an emerging candidate biomarker of the different forms of synucleinopathy. This review summarizes several biomarkers of neurodegenerative dementia validated based on the neuropathological processes occurring in brain tissue. Notwithstanding the paucity of pathologically validated biomarkers and their high analytical variability, the combinations of these biomarkers may well represent a key and more precise analytical and diagnostic tool in the complex plethora of degenerative dementia.
Collapse
Affiliation(s)
- Giulia M Sancesario
- a Department of Clinical and Behavioural Neurology , Santa Lucia Foundation, IRCCS , Rome , Italy and
| | - Sergio Bernardini
- b Department of Experimental Medicine and Surgery , Tor Vergata University of Rome , Rome , Italy
| |
Collapse
|
86
|
Börger M, Funke S, Bähr M, Grus F, Lingor P. Biomarker sources for Parkinson's disease: Time to shed tears? ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.baga.2015.05.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
87
|
Stav AL, Aarsland D, Johansen KK, Hessen E, Auning E, Fladby T. Amyloid-β and α-synuclein cerebrospinal fluid biomarkers and cognition in early Parkinson's disease. Parkinsonism Relat Disord 2015; 21:758-64. [DOI: 10.1016/j.parkreldis.2015.04.027] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/14/2015] [Accepted: 04/22/2015] [Indexed: 01/06/2023]
|
88
|
Simuni T, Caspell-Garcia C, Coffey C, Chahine LM, Lasch S, Oertel WH, Mayer G, Högl B, Postuma R, Videnovic A, Amara AW, Marek K. Correlates of excessive daytime sleepiness in de novo Parkinson's disease: A case control study. Mov Disord 2015; 30:1371-81. [PMID: 26095202 DOI: 10.1002/mds.26248] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 03/10/2015] [Accepted: 03/19/2015] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVE This study was undertaken to determine the frequency and correlates of excessive daytime sleepiness in de novo, untreated Parkinson's disease (PD) patients compared with the matched healthy controls. METHODS Data were obtained from the Parkinson's Progression Markers Initiative, an international study of de novo, untreated PD patients and healthy controls. At baseline, participants were assessed with a wide range of motor and nonmotor scales, including the Movement Disorder Society Unified Parkinson's Disease Rating Scale (MDS-UPDRS). Excessive daytime sleepiness was assessed based on the Epworth Sleepiness scale (ESS), with a cutoff of 10. RESULTS Four hundred twenty-three PD subjects and 196 healthy controls were recruited into the study. Mean ESS (min, max) score was 5.8 (0, 20) for the PD subjects and 5.6 (0, 19) for healthy controls (P = 0.54). Sixty-six (15.6%) PD subjects and 24 (12%) healthy controls had ESS of at least 10 (P = 0.28). No difference was seen in demographic characteristics, age of onset, disease duration, PD subtype, cognitive status, or utilization of sedatives between the PD sleepiness-positive versus the negative group. The sleepiness-positive group had higher MDS-UPDRS Part I and II but not III scores, and higher depression and autonomic dysfunction scores. Sleepiness was associated with a marginal reduction of A-beta (P = 0.05) but not alpha-synuclein spinal fluid levels in PD. CONCLUSIONS This largest case control study demonstrates no difference in prevalence of excessive sleepiness in subjects with de novo untreated PD compared with healthy controls. The only clinical correlates of sleepiness were mood and autonomic dysfunction. Ongoing longitudinal analyses will be essential to further examine clinical and biological correlates of sleepiness in PD and specifically the role of dopaminergic therapy.
Collapse
Affiliation(s)
- Tanya Simuni
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | | - Lama M Chahine
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shirley Lasch
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| | | | - Geert Mayer
- Hephata-Klinik, Hephata Hessisches Diakoniezentrum e. V
| | - Birgit Högl
- Innsbruck Medical University, Innsbruck, Austria
| | | | | | | | - Ken Marek
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| | | |
Collapse
|
89
|
Kansal K, Irwin DJ. The use of cerebrospinal fluid and neuropathologic studies in neuropsychiatry practice and research. Psychiatr Clin North Am 2015; 38:309-22. [PMID: 25998118 PMCID: PMC4443852 DOI: 10.1016/j.psc.2015.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gold standard for diagnosis of neurodegenerative diseases (ie, Alzheimer disease, frontotemporal dementia, Parkinson disease, dementia with Lewy bodies, amyotrophic lateral sclerosis) is neuropathologic examination at autopsy. As such, laboratory studies play a central role in antemortem diagnosis of these conditions and their differentiation from the neuroinflammatory, infectious, toxic, and other nondegenerative etiologies (eg, rapidly progressive dementias) that are encountered in neuropsychiatric practice. This article summarizes the use of cerebrospinal fluid (CSF) laboratory studies in the diagnostic evaluation of dementia syndromes and emerging CSF biomarkers specific for underlying neuropathology in neurodegenerative disease research.
Collapse
Affiliation(s)
- Kalyani Kansal
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - David J. Irwin
- Department of Neurology, University of Pennsylvania Perelman School of medicine, Philadelphia, PA USA
| |
Collapse
|
90
|
Lin CH, Wu RM. Biomarkers of cognitive decline in Parkinson's disease. Parkinsonism Relat Disord 2015; 21:431-43. [PMID: 25737398 DOI: 10.1016/j.parkreldis.2015.02.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 02/10/2015] [Accepted: 02/12/2015] [Indexed: 10/24/2022]
Abstract
Cognitive impairment is a frequent and devastating non-motor symptom of Parkinson's disease (PD). Impaired cognition has a major impact on either quality of life or mortality in patients with PD. Notably, the rate of cognitive decline and pattern of early cognitive deficits in PD are highly variable between individuals. Given that the underlying mechanisms of cognitive decline or dementia associated with PD remain unclear, there is currently no mechanism-based treatment available. Identification of biological markers, including neuroimaging, biofluids and common genetic variants, that account for the heterogeneity of PD related cognitive decline could provide important insights into the pathological processes that underlie cognitive impairment in PD. These combined biomarker approaches will enable early diagnosis and provide indicators of cognitive progression in PD patients. This review summarizes recent advances in the development of biomarkers for cognitive impairments in PD.
Collapse
Affiliation(s)
- Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ruey-Meei Wu
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| |
Collapse
|
91
|
Cavaleri F. Paradigm shift redefining molecular, metabolic and structural events in Alzheimer's disease involves a proposed contribution by transition metals. Defined lengthy preclinical stage provides new hope to circumvent advancement of disease- and age-related neurodegeneration. Med Hypotheses 2015; 84:460-9. [PMID: 25691377 DOI: 10.1016/j.mehy.2015.01.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 01/30/2015] [Indexed: 12/28/2022]
Abstract
It is estimated that 5.5 Million North Americans suffer from varying degrees of Alzheimer's disease (AD) and by the year 2050 it may be one in 85 people globally (100 Million). It will be shown that heavy metal toxicity plays a significant role in sporadic AD. Although current literature speaks to involvement of metal ions (via Fenton reaction), studies and reviewers have yet to link cellular events including known structural changes such as amyloid plaque development to this metal toxicity the way it is proposed here. Contrary to the current AD model which positions BACE1 (β-secretase) as an aberrant or AD-advancing enzyme, it is proposed herein that the neuron's protective counteraction to this metal toxicity is, in fact, a justified increase in BACE1 activity and amyloid precursor protein (APP) processing to yield more secreted APP (sAPP) and β-amyloid peptide in response to metal toxicity. This new perspective which justifies a functional role for APP, BACE1 enzyme activity and the peptide products from this activity may at first appear to be counterintuitive. Compelling evidence, however, is presented and a mechanism is shown herein that validate BACE1 recruitment and the resulting β-amyloid protein as strategic countermeasures serving the cell effectively against neuro-impeding disease. It is proposed that β-amyloid peptide chelates and sequesters free heavy metals in the extracellular medium to aggregate as amyloid plaque while unchelated β-amyloid migrates across the cell membrane to chelate intracellular free divalent metals. The sequestered intracellular metal is subsequently chaperoned as a metallo-peptide to cross the plasma membrane and aggregate as amyloid plaques extracellularly. The BACE1 countermeasure is not genetic or metabolic aberration; and this novel conclusion demonstrates that it must not be inhibited as currently targeted. APP, BACE1, β-amyloid peptide, and sAPP play positive roles against the preclinical oxidative load that predates AD symptoms for as long as 20 years. A healthy neuron may tolerate free metal toxicity, such as iron in the case of injury-induced amyloid, for as long as twenty years due to this very BACE1 activity. In later stages, the uncontrolled metals and ROS are compounded by other factors which together overcome this BACE1/β-amyloid protein countermeasure. This results in a sudden increase in IL-1 leading to Tau's hyperphosphorylation as cited and eventually to Tau dissociation from the microtubule cytoskeleton interrupting cell trafficking. At this later stage of AD the β-amyloid protein which once served as a vehicle to escort toxic metals to the extracellular medium and a trap to form a relatively benign extraneuronal disposal site is no longer translocated due to interruption of trafficking and now accumulates intracellularly facilitating hyper-oxidative ROS levels and contributes to irreversible neuron apoptosis.
Collapse
Affiliation(s)
- Franco Cavaleri
- Brain Research Center, UBC Hospital, 2211 Wesbrook Mall, Vancouver, BC V6T 2B5, Canada.
| |
Collapse
|
92
|
CSF tau and tau/Aβ42 predict cognitive decline in Parkinson's disease. Parkinsonism Relat Disord 2015; 21:271-6. [PMID: 25596881 DOI: 10.1016/j.parkreldis.2014.12.027] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 11/06/2014] [Accepted: 12/28/2014] [Indexed: 01/24/2023]
Abstract
INTRODUCTION A substantial proportion of patients with Parkinson's disease (PD) have concomitant cognitive dysfunction. Identification of biomarker profiles that predict which PD patients have a greater likelihood for progression of cognitive symptoms is pressingly needed for future treatment and prevention approaches. METHODS Subjects were drawn from the Deprenyl and Tocopherol Antioxidative Therapy of Parkinsonism (DATATOP) study, a large clinical trial that enrolled initially untreated PD patients. For the current study, Phase One encompassed trial baseline until just prior to levodopa administration (n = 403), and Phase Two spanned the initiation of levodopa treatment until the end of cognitive follow-up (n = 305). Correlations and linear mixed models were performed to determine cross-sectional and longitudinal associations between baseline amyloid β1-42 (Aβ42), total tau (t-tau), and phosphorylated tau (p-tau) in cerebrospinal fluid (CSF) and measures of memory and executive function. Analyses also considered APOE genotype and tremor- vs. rigidity-dominant phenotype. RESULTS No association was found between baseline CSF biomarkers and cognitive test performance during Phase One. However, once levodopa treatment was initiated, higher p-tau and p-tau/Aβ42 predicted subsequent decline on cognitive tasks involving both memory and executive functions. The interactions between biomarkers and cognition decline did not appear to be influenced by levodopa dosage, APOE genotype or motor phenotype. CONCLUSIONS The current study has, for the first time, demonstrated the possible involvement of tau species, whose gene (MAPT) has been consistently linked to the risk of PD by genome-wide association studies, in the progression of cognitive symptoms in PD.
Collapse
|
93
|
Buddhala C, Campbell MC, Perlmutter JS, Kotzbauer PT. Correlation between decreased CSF α-synuclein and Aβ₁₋₄₂ in Parkinson disease. Neurobiol Aging 2015; 36:476-84. [PMID: 25212463 PMCID: PMC4268043 DOI: 10.1016/j.neurobiolaging.2014.07.043] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 07/14/2014] [Accepted: 07/30/2014] [Indexed: 02/07/2023]
Abstract
Accumulation of misfolded α-synuclein (α-syn) protein in Lewy bodies and neurites is the cardinal pathologic feature of Parkinson disease (PD), but abnormal deposition of other proteins may also play a role. Cerebrospinal fluid (CSF) levels of proteins known to accumulate in PD may provide insight into disease-associated changes in protein metabolism and their relationship to disease progression. We measured CSF α-syn, amyloid β₁₋₄₂ (Aβ₁₋₄₂), and tau from 77 nondemented PD and 30 control participants. CSF α-syn and Aβ₁₋₄₂ were significantly lower in PD compared with controls. In contrast with increased CSF tau in Alzheimer disease, CSF tau did not significantly differ between PD and controls. CSF protein levels did not significantly correlate with ratings of motor function or performance on neuropsychological testing. As expected, CSF Aβ₁₋₄₂ inversely correlated with [(11)C]-Pittsburgh compound B (PiB) mean cortical binding potential, with PiB(+) PD participants having lower CSF Aβ₁₋₄₂ compared with PiB(-) PD participants. Furthermore, CSF α-syn positively correlated with Aβ₁₋₄₂ in PD participants but not in controls, suggesting a pathophysiologic connection between the metabolisms of these proteins in PD.
Collapse
Affiliation(s)
- Chandana Buddhala
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA; Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO, USA
| | - Meghan C Campbell
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA; Department of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Joel S Perlmutter
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO, USA; Department of Radiology, Washington University School of Medicine, St Louis, MO, USA; Department of Anatomy and Neurobiology, Washington University School of Medicine, St Louis, MO, USA; Program in Occupational Therapy, Washington University School of Medicine, St Louis, MO, USA; Program in Physical Therapy, Washington University School of Medicine, St Louis, MO, USA
| | - Paul T Kotzbauer
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA; Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
94
|
Jiménez-Jiménez FJ, Alonso-Navarro H, García-Martín E, Agúndez JAG. Cerebrospinal fluid biochemical studies in patients with Parkinson's disease: toward a potential search for biomarkers for this disease. Front Cell Neurosci 2014; 8:369. [PMID: 25426023 PMCID: PMC4227512 DOI: 10.3389/fncel.2014.00369] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/20/2014] [Indexed: 12/14/2022] Open
Abstract
The blood-brain barrier supplies brain tissues with nutrients and filters certain compounds from the brain back to the bloodstream. In several neurodegenerative diseases, including Parkinson's disease (PD), there are disruptions of the blood-brain barrier. Cerebrospinal fluid (CSF) has been widely investigated in PD and in other parkinsonian syndromes with the aim of establishing useful biomarkers for an accurate differential diagnosis among these syndromes. This review article summarizes the studies reported on CSF levels of many potential biomarkers of PD. The most consistent findings are: (a) the possible role of CSF urate on the progression of the disease; (b) the possible relations of CSF total tau and phosphotau protein with the progression of PD and with the preservation of cognitive function in PD patients; (c) the possible value of CSF beta-amyloid 1-42 as a useful marker of further cognitive decline in PD patients, and (d) the potential usefulness of CSF neurofilament (NFL) protein levels in the differential diagnosis between PD and other parkinsonian syndromes. Future multicentric, longitudinal, prospective studies with long-term follow-up and neuropathological confirmation would be useful in establishing appropriate biomarkers for PD.
Collapse
Affiliation(s)
| | | | - Elena García-Martín
- Department of Biochemistry and Molecular Biology, University of ExtremaduraCáceres, Spain
- AMGenomicsCáceres, Spain
| | - José A. G. Agúndez
- AMGenomicsCáceres, Spain
- Department of Pharmacology, University of ExtremaduraCáceres, Spain
| |
Collapse
|
95
|
Compta Y, Valente T, Saura J, Segura B, Iranzo Á, Serradell M, Junqué C, Tolosa E, Valldeoriola F, Muñoz E, Santamaria J, Cámara A, Fernández M, Fortea J, Buongiorno M, Molinuevo JL, Bargalló N, Martí MJ. Correlates of cerebrospinal fluid levels of oligomeric- and total-α-synuclein in premotor, motor and dementia stages of Parkinson’s disease. J Neurol 2014; 262:294-306. [DOI: 10.1007/s00415-014-7560-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 10/10/2014] [Accepted: 10/24/2014] [Indexed: 11/28/2022]
|
96
|
Abstract
Neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and frontotemporal dementia have several important features in common. They are progressive, they affect a relatively inaccessible organ, and we have no disease-modifying therapies for them. For these brain-based diseases, current diagnosis and evaluation of disease severity rely almost entirely on clinical examination, which may be only a rough approximation of disease state. Thus, the development of biomarkers-objective, relatively easily measured, and precise indicators of pathogenic processes-could improve patient care and accelerate therapeutic discovery. Yet existing, rigorously tested neurodegenerative disease biomarkers are few, and even fewer biomarkers have translated into clinical use. To find new biomarkers for these diseases, an unbiased, high-throughput screening approach may be needed. In this review, I will describe the potential utility of such an approach to biomarker discovery, using Parkinson's disease as a case example.
Collapse
Affiliation(s)
- Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, 3 West Gates, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
97
|
Schade S, Mollenhauer B. Biomarkers in biological fluids for dementia with Lewy bodies. ALZHEIMERS RESEARCH & THERAPY 2014; 6:72. [PMID: 25478030 PMCID: PMC4255553 DOI: 10.1186/s13195-014-0072-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dementia with Lewy bodies (DLB) has become the second most common neurodegenerative dementia due to demographic ageing. Differential diagnosis is still troublesome especially in early stages of the disease, since there is a great clinical and neuropathological overlap primarily with Alzheimer's disease and Parkinson's disease. Therefore, more specific biomarkers, not only for scientific reasons but also for clinical therapeutic decision-making, are urgently needed. In this review, we summarize the knowledge on fluid biomarkers for DLB, derived predominantly from cerebrospinal fluid. We discuss the value of well-defined markers (β-amyloid, (phosphorylated) tau, α-synuclein) as well as some promising 'upcoming' substances, which still have to be further evaluated.
Collapse
Affiliation(s)
- Sebastian Schade
- Paracelsus-Elena-Klinik, Klinikstraße 16, Kassel, D-34128, Germany ; Department of Clinical Neurophysiology, University Medical Center, Georg-August University, Robert-Koch Straße 40, Göttingen, 37075, Germany
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Klinikstraße 16, Kassel, D-34128, Germany ; Department of Neurosurgery, University Medical Center, Georg-August University, Robert-Koch Straße 40, Göttingen, 37075, Germany ; Department of Neuropathology, University Medical Center, Georg-August University, Robert-Koch Straße 40, Göttingen, 37075, Germany
| |
Collapse
|
98
|
Magdalinou N, Lees AJ, Zetterberg H. Cerebrospinal fluid biomarkers in parkinsonian conditions: an update and future directions. J Neurol Neurosurg Psychiatry 2014; 85:1065-75. [PMID: 24691581 PMCID: PMC4173749 DOI: 10.1136/jnnp-2013-307539] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Parkinsonian diseases comprise a heterogeneous group of neurodegenerative disorders, which show significant clinical and pathological overlap. Accurate diagnosis still largely relies on clinical acumen; pathological diagnosis remains the gold standard. There is an urgent need for biomarkers to diagnose parkinsonian disorders, particularly in the early stages when diagnosis is most difficult. In this review, several of the most promising cerebrospinal fluid candidate markers will be discussed. Their strengths and limitations will be considered together with future developments in the field.
Collapse
Affiliation(s)
- Nadia Magdalinou
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Andrew J Lees
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Henrik Zetterberg
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| |
Collapse
|
99
|
Vranová HP, Hényková E, Kaiserová M, Menšíková K, Vaštík M, Mareš J, Hluštík P, Zapletalová J, Strnad M, Stejskal D, Kaňovský P. Tau protein, beta-amyloid1–42 and clusterin CSF levels in the differential diagnosis of Parkinsonian syndrome with dementia. J Neurol Sci 2014; 343:120-4. [DOI: 10.1016/j.jns.2014.05.052] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 05/20/2014] [Accepted: 05/22/2014] [Indexed: 10/25/2022]
|
100
|
Parnetti L, Farotti L, Eusebi P, Chiasserini D, De Carlo C, Giannandrea D, Salvadori N, Lisetti V, Tambasco N, Rossi A, Majbour NK, El-Agnaf O, Calabresi P. Differential role of CSF alpha-synuclein species, tau, and Aβ42 in Parkinson's Disease. Front Aging Neurosci 2014; 6:53. [PMID: 24744728 PMCID: PMC3978246 DOI: 10.3389/fnagi.2014.00053] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/05/2014] [Indexed: 11/21/2022] Open
Abstract
There is a great interest in developing cerebrospinal fluid (CSF) biomarkers for diagnosis and prognosis of Parkinson's disease (PD). CSF alpha synuclein (α-syn) species, namely total and oligomeric α-syn (t-α-syn and o-α-syn), have shown to be of help for PD diagnosis. Preliminary evidences show that the combination of CSF t-α-syn and classical Alzheimer's disease (AD) biomarkers—β-amyloid 1–42 (Aβ42), total tau (t-tau), phosphorylated tau (p-tau)—differentiate PD patients from controls, and that reduced levels of Aβ42 represent a predictive factor for development of cognitive deterioration in PD. In this prospective study carried out in 44 PD patients and 25 neurological controls we wanted to verify whether the combination of CSF α-synuclein species—t-α-syn and o-α-syn—and classical AD biomarkers may help in differentiating PD from neurological controls, and if these biomarkers may predict cognitive decline. The median of follow-up duration was 3 years (range: 2–6 years). Mini Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA) were used for monitoring cognitive changes along time, being administered once a year. Oligo/total α-syn ratio (o/t-α-syn ratio) confirmed its diagnostic value, significantly contributing to the discrimination of PD from neurological controls. A greater diagnostic accuracy was reached when combining o/t-α-syn and Aβ42/tau ratios (Sens = 0.70, Spec = 0.84, AUC = 0.82; PPV = 0.89, NPV = 0.62, LR+ = 4.40, DOR = 12.52). Low CSF Aβ42 level was associated with a higher rate of MMSE and MoCA decline, confirming its role as independent predictive factor for cognitive decline in PD. None of the other biomarkers assessed (t-tau, p-tau, t-α-syn and o-α-syn) showed to have prognostic value. We conclude that combination of CSF o/t-α-syn and Aβ42/tau ratios improve the diagnostic accuracy of PD. PD patients showing low CSF Aβ42 levels at baseline are more prone to develop cognitive decline.
Collapse
Affiliation(s)
- Lucilla Parnetti
- Clinica Neurologica, Università degli Studi di Perugia Perugia, Italy
| | - Lucia Farotti
- Clinica Neurologica, Università degli Studi di Perugia Perugia, Italy
| | - Paolo Eusebi
- Clinica Neurologica, Università degli Studi di Perugia Perugia, Italy ; Dipartimento di Epidemiologia, Regione Umbria Perugia, Italy
| | - Davide Chiasserini
- Dipartimento di Scienze Farmaceutiche, Sezione di Biochimica, Università degli Studi di Perugia Perugia, Italy
| | - Claudia De Carlo
- Clinica Neurologica, Università degli Studi di Perugia Perugia, Italy
| | - David Giannandrea
- Clinica Neurologica, Università degli Studi di Perugia Perugia, Italy
| | - Nicola Salvadori
- Clinica Neurologica, Università degli Studi di Perugia Perugia, Italy
| | - Viviana Lisetti
- Clinica Neurologica, Università degli Studi di Perugia Perugia, Italy
| | - Nicola Tambasco
- Clinica Neurologica, Università degli Studi di Perugia Perugia, Italy
| | - Aroldo Rossi
- Clinica Neurologica, Università degli Studi di Perugia Perugia, Italy
| | - Nour K Majbour
- Department of Biochemistry, Faculty of Medicine and Health Sciences, United Arab Emirates University Al Ain, United Arab Emirates
| | - Omar El-Agnaf
- Department of Biochemistry, Faculty of Medicine and Health Sciences, United Arab Emirates University Al Ain, United Arab Emirates ; Faculty of Medicine, King Abdulaziz University Jeddah, Saudi Arabia
| | - Paolo Calabresi
- Clinica Neurologica, Università degli Studi di Perugia Perugia, Italy ; IRCCS Fondazione S. Lucia Roma, Italy
| |
Collapse
|