1
|
Cousins KAQ, Irwin DJ, Tropea TF, Rhodes E, Phillips J, Chen-Plotkin AS, Brumm MC, Coffey CS, Kang JH, Simuni T, Foroud TM, Toga AW, Tanner CM, Kieburtz KD, Mollenhauer B, Galasko D, Hutten S, Weintraub D, Siderowf AD, Marek K, Poston KL, Shaw LM. Evaluation of ATN PD Framework and Biofluid Markers to Predict Cognitive Decline in Early Parkinson Disease. Neurology 2024; 102:e208033. [PMID: 38306599 PMCID: PMC11383879 DOI: 10.1212/wnl.0000000000208033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 10/13/2023] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND AND OBJECTIVES In Parkinson disease (PD), Alzheimer disease (AD) copathology is common and clinically relevant. However, the longitudinal progression of AD CSF biomarkers-β-amyloid 1-42 (Aβ42), phosphorylated tau 181 (p-tau181), and total tau (t-tau)-in PD is poorly understood and may be distinct from clinical AD. Moreover, it is unclear whether CSF p-tau181 and serum neurofilament light (NfL) have added prognostic utility in PD, when combined with CSF Aβ42. First, we describe longitudinal trajectories of biofluid markers in PD. Second, we modified the AD β-amyloid/tau/neurodegeneration (ATN) framework for application in PD (ATNPD) using CSF Aβ42 (A), p-tau181 (T), and serum NfL (N) and tested ATNPD prediction of longitudinal cognitive decline in PD. METHODS Participants were selected from the Parkinson's Progression Markers Initiative cohort, clinically diagnosed with sporadic PD or as controls, and followed up annually for 5 years. Linear mixed-effects models (LMEMs) tested the interaction of diagnosis with longitudinal trajectories of analytes (log transformed, false discovery rate [FDR] corrected). In patients with PD, LMEMs tested how baseline ATNPD status (AD [A+T+N±] vs not) predicted clinical outcomes, including Montreal Cognitive Assessment (MoCA; rank transformed, FDR corrected). RESULTS Participants were 364 patients with PD and 168 controls, with comparable baseline mean (±SD) age (patients with PD = 62 ± 10 years; controls = 61 ± 11 years]; Mann-Whitney Wilcoxon: p = 0.4) and sex distribution (patients with PD = 231 male individuals [63%]; controls = 107 male individuals [64%]; χ2: p = 1). Patients with PD had overall lower CSF p-tau181 (β = -0.16, 95% CI -0.23 to -0.092, p = 2.2e-05) and t-tau than controls (β = -0.13, 95% CI -0.19 to -0.065, p = 4e-04), but not Aβ42 (p = 0.061) or NfL (p = 0.32). Over time, patients with PD had greater increases in serum NfL than controls (β = 0.035, 95% CI 0.022 to 0.048, p = 9.8e-07); slopes of patients with PD did not differ from those of controls for CSF Aβ42 (p = 0.18), p-tau181 (p = 1), or t-tau (p = 0.96). Using ATNPD, PD classified as A+T+N± (n = 32; 9%) had worse cognitive decline on global MoCA (β = -73, 95% CI -110 to -37, p = 0.00077) than all other ATNPD statuses including A+ alone (A+T-N-; n = 75; 21%). DISCUSSION In patients with early PD, CSF p-tau181 and t-tau were low compared with those in controls and did not increase over 5 years of follow-up. Our study shows that classification using modified ATNPD (incorporating CSF Aβ42, CSF p-tau181, and serum NfL) can identify biologically relevant subgroups of PD to improve prediction of cognitive decline in early PD.
Collapse
Affiliation(s)
- Katheryn A Q Cousins
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - David J Irwin
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Thomas F Tropea
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Emma Rhodes
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Jeffrey Phillips
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Alice S Chen-Plotkin
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Michael C Brumm
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Christopher S Coffey
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Ju Hee Kang
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Tanya Simuni
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Tatiana M Foroud
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Arthur W Toga
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Caroline M Tanner
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Karl D Kieburtz
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Brit Mollenhauer
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Douglas Galasko
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Samantha Hutten
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Daniel Weintraub
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Andrew D Siderowf
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Kenneth Marek
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Kathleen L Poston
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Leslie M Shaw
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| |
Collapse
|
2
|
Zagkos L, Dib MJ, Cronjé HT, Elliott P, Dehghan A, Tzoulaki I, Gill D, Daghlas I. Cerebrospinal Fluid C1-Esterase Inhibitor and Tie-1 Levels Affect Cognitive Performance: Evidence from Proteome-Wide Mendelian Randomization. Genes (Basel) 2024; 15:71. [PMID: 38254961 PMCID: PMC10815381 DOI: 10.3390/genes15010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/29/2023] [Accepted: 01/01/2024] [Indexed: 01/24/2024] Open
Abstract
OBJECTIVE The association of cerebrospinal fluid (CSF) protein levels with cognitive function in the general population remains largely unexplored. We performed Mendelian randomization (MR) analyses to query which CSF proteins may have potential causal effects on cognitive performance. METHODS AND ANALYSIS Genetic associations with CSF proteins were obtained from a genome-wide association study conducted in up to 835 European-ancestry individuals and for cognitive performance from a meta-analysis of GWAS including 257,841 European-ancestry individuals. We performed Mendelian randomization (MR) analyses to test the effect of randomly allocated variation in 154 genetically predicted CSF protein levels on cognitive performance. Findings were validated by performing colocalization analyses and considering cognition-related phenotypes. RESULTS Genetically predicted C1-esterase inhibitor levels in the CSF were associated with a better cognitive performance (SD units of cognitive performance per 1 log-relative fluorescence unit (RFU): 0.23, 95% confidence interval: 0.12 to 0.35, p = 7.91 × 10-5), while tyrosine-protein kinase receptor Tie-1 (sTie-1) levels were associated with a worse cognitive performance (-0.43, -0.62 to -0.23, p = 2.08 × 10-5). These findings were supported by colocalization analyses and by concordant effects on distinct cognition-related and brain-volume measures. CONCLUSIONS Human genetics supports a role for the C1-esterase inhibitor and sTie-1 in cognitive performance.
Collapse
Affiliation(s)
- Loukas Zagkos
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London SW7 2BX, UK; (P.E.); (A.D.); (I.T.); (D.G.)
| | - Marie-Joe Dib
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Héléne T. Cronjé
- Department of Public Health, Section of Epidemiology, University of Copenhagen, 1165 Copenhagen, Denmark;
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London SW7 2BX, UK; (P.E.); (A.D.); (I.T.); (D.G.)
- UK Dementia Research Institute at Imperial College London, Hammersmith Hospital, London W1T 7NF, UK
- Medical Research Council Centre for Environment and Health, School of Public Health, Imperial College London, London SW7 2AZ, UK
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London SW7 2BX, UK; (P.E.); (A.D.); (I.T.); (D.G.)
- UK Dementia Research Institute at Imperial College London, Hammersmith Hospital, London W1T 7NF, UK
- Medical Research Council Centre for Environment and Health, School of Public Health, Imperial College London, London SW7 2AZ, UK
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London SW7 2BX, UK; (P.E.); (A.D.); (I.T.); (D.G.)
- UK Dementia Research Institute at Imperial College London, Hammersmith Hospital, London W1T 7NF, UK
- Medical Research Council Centre for Environment and Health, School of Public Health, Imperial College London, London SW7 2AZ, UK
- Centre for Systems Biology, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London SW7 2BX, UK; (P.E.); (A.D.); (I.T.); (D.G.)
| | - Iyas Daghlas
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA;
| |
Collapse
|
3
|
Pilotto A, Zanusso G, Antelmi E, Okuzumi A, Zatti C, Lupini A, Bongianni M, Padovani A, Hattori N. Biofluid Markers and Tissue Biopsies Analyses for the Prodromal and Earliest Phase of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2024; 14:S333-S344. [PMID: 39331105 PMCID: PMC11494635 DOI: 10.3233/jpd-240007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/09/2024] [Indexed: 09/28/2024]
Abstract
The recent development of new methods to detect misfolded α-synuclein (αSyn) aggregates in biofluids and tissue biopsies in the earliest Parkinson's disease (PD) phases is dramatically challenging the biological definition of PD. The αSyn seed amplification methods in cerebrospinal fluid (CSF) showed high sensitivity and specificity for early diagnosis of PD and Lewy bodies disorders. Several studies in isolated REM sleep behavior disorders and other at-risk populations also demonstrated a high prevalence of CSF αSyn positivity and its potential value in predicting the phenoconversion to clinically manifested diseases. Growing evidence exists for αSyn aggregates in olfactory mucosa, skin, and other tissues in subjects with PD or at-risk subjects. DOPA decarboxylase and numerous other candidates have been additionally proposed for either diagnostic or prognostic purposes in earliest PD phases. The newly described αSyn detection in blood, through its quantification in neuronally-derived exosome vesicles, represents a technical challenge that could open a new scenario for the biological diagnosis of PD. Despite this growing evidence in the field, most of method of αSyn detection and markers still need to be validated in ongoing longitudinal studies through an accurate assessment of different prodromal disease subtypes and scenarios before being definitively implemented in clinical settings.
Collapse
Affiliation(s)
- Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili of Brescia, Brescia, Italy
| | - Gianluigi Zanusso
- Neurology Unit, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Elena Antelmi
- Neurology Unit, Parkinson Disease and Movement Disorders Division, Department of Engineering and Medicine of Innovation, University of Verona, Verona, Italy
| | - Ayami Okuzumi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Cinzia Zatti
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
| | - Alessandro Lupini
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
| | - Matilde Bongianni
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili of Brescia, Brescia, Italy
- Brain Health Center, University of Brescia, Brescia, Italy
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Neurodegenerative Disorders Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, Japan
| |
Collapse
|
4
|
Zenuni H, Bovenzi R, Bissacco J, Grillo P, Simonetta C, Mascioli D, Pieri M, Bernardini S, Sancesario GM, Stefani A, Mercuri NB, Schirinzi T. Clinical and neurochemical correlates of the APOE genotype in early-stage Parkinson's disease. Neurobiol Aging 2023; 131:24-28. [PMID: 37572524 DOI: 10.1016/j.neurobiolaging.2023.07.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 08/14/2023]
Abstract
Emerging evidence indicates that apolipoprotein E (APOE) genotype may influence Parkinson's disease (PD) course, although clinical and neurochemical correlates have not been completely established. This study aimed to determine the associations of APOE genotypes (ε4 vs. non-ε4) with cerebrospinal fluid (CSF) neurodegeneration biomarkers and clinical parameters in early-stage PD patients. One hundred and seventy-five PD patients and 89 non-neurodegenerative controls grouped in APOE-ε4 carriers (28 PD; 12 controls) and non-APOE-ε4 carriers (147 PD; 78 controls) were enrolled. CSF levels of amyloid-β-42, amyloid-β-40, total and 181-phosphorylated tau, and clinical scores were compared among groups adjusting for main covariates. APOE genotypes prevalence was similar in PD and controls. PD APOE-ε4 carriers had lower amyloid-β-42 CSF levels than PD non-APOE-ε4 carriers and controls, independently from age. PD APOE-ε4 carriers also had higher total and "item 5" (attention and memory) non-motor symptoms scale scores than PD non-APOE-ε4 carriers, independently from confounding factors. APOE-ε4 genotype might thus account for a more vulnerable PD subtype characterized by prominent amyloidopathy and a greater burden of non-motor symptoms in the early disease stages. DATA AVAILABILITY: Data are available upon reasonable request.
Collapse
Affiliation(s)
- Henri Zenuni
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Roberta Bovenzi
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Jacopo Bissacco
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Piergiorgio Grillo
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Clara Simonetta
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Davide Mascioli
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Massimo Pieri
- Clinical Biochemistry Unit, Department of Experimental Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Segio Bernardini
- Clinical Biochemistry Unit, Department of Experimental Medicine, University of Roma Tor Vergata, Rome, Italy
| | | | - Alessandro Stefani
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Tommaso Schirinzi
- Unit of Neurology, Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy.
| |
Collapse
|
5
|
Rastogi S, Rani K, Rai S, Singh R, Bharti PS, Sharma V, Sahu J, Kapoor V, Vishwakarma P, Garg S, Gholap SL, Inampudi KK, Modi GP, Rani N, Tripathi M, Srivastava A, Rajan R, Nikolajeff F, Kumar S. Fluorescence-tagged salivary small extracellular vesicles as a nanotool in early diagnosis of Parkinson's disease. BMC Med 2023; 21:335. [PMID: 37667227 PMCID: PMC10478478 DOI: 10.1186/s12916-023-03031-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Parkinson's disease is generally asymptomatic at earlier stages. At an early stage, there is an extensive progression in the neuropathological hallmarks, although, at this stage, diagnosis is not possible with currently available diagnostic methods. Therefore, the pressing need is for susceptibility risk biomarkers that can aid in better diagnosis and therapeutics as well can objectively serve to measure the endpoint of disease progression. The role of small extracellular vesicles (sEV) in the progression of neurodegenerative diseases could be potent in playing a revolutionary role in biomarker discovery. METHODS In our study, the salivary sEV were efficiently isolated by chemical precipitation combined with ultrafiltration from subjects (PD = 70, healthy controls = 26, and prodromal PD = 08), followed by antibody-based validation with CD63, CD9, GAPDH, Flotillin-1, and L1CAM. Morphological characterization of the isolated sEV through transmission electron microscopy. The quantification of sEV was achieved by fluorescence (lipid-binding dye-labeled) nanoparticle tracking analysis and antibody-based (CD63 Alexa fluor 488 tagged sEV) nanoparticle tracking analysis. The total alpha-synuclein (α-synTotal) in salivary sEVs cargo was quantified by ELISA. The disease severity staging confirmation for n = 18 clinically diagnosed Parkinson's disease patients was done by 99mTc-TRODAT-single-photon emission computed tomography. RESULTS We observed a significant increase in total sEVs concentration in PD patients than in the healthy control (HC), where fluorescence lipid-binding dye-tagged sEV were observed to be higher in PD (p = 0.0001) than in the HC using NTA with a sensitivity of 94.34%. In the prodromal PD cases, the fluorescence lipid-binding dye-tagged sEV concentration was found to be higher (p = 0.008) than in HC. This result was validated through anti-CD63 tagged sEV (p = 0.0006) with similar sensitivity of 94.12%. We further validated our findings with the ELISA based on α-synTotal concentration in sEV, where it was observed to be higher in PD (p = 0.004) with a sensitivity of 88.24%. The caudate binding ratios in 99mTc-TRODAT-SPECT represent a positive correlation with sEV concentration (r = 0.8117 with p = 0.0112). CONCLUSIONS In this study, for the first time, we have found that the fluorescence-tagged sEV has the potential to screen the progression of disease with clinically acceptable sensitivity and can be a potent early detection method for PD.
Collapse
Affiliation(s)
- Simran Rastogi
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Komal Rani
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
- Department of Pathology & Laboratory Medicine, All India Institute of Medical Sciences Bibinagar, Hyderabad, 508126, India
| | - Sanskriti Rai
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Rishabh Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Prahalad Singh Bharti
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Vaibhav Sharma
- Department of Health, Education, and Technology, Luleå University of Technology, 97187, Luleå, Sweden
| | - Jyoti Sahu
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Vrinda Kapoor
- School of Interdisciplinary Research, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Poorvi Vishwakarma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sumit Garg
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | | | | | - Gyan Prakash Modi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology BHU, Varanasi, 221005, India
| | - Neerja Rani
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Madhavi Tripathi
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Achal Srivastava
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Roopa Rajan
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Fredrik Nikolajeff
- Department of Health, Education, and Technology, Luleå University of Technology, 97187, Luleå, Sweden
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India.
- Department of Health, Education, and Technology, Luleå University of Technology, 97187, Luleå, Sweden.
| |
Collapse
|
6
|
Barba L, Abu-Rumeileh S, Halbgebauer S, Bellomo G, Paolini Paoletti F, Gaetani L, Oeckl P, Steinacker P, Massa F, Parnetti L, Otto M. CSF Synaptic Biomarkers in AT(N)-Based Subgroups of Lewy Body Disease. Neurology 2023; 101:e50-e62. [PMID: 37188538 PMCID: PMC10351307 DOI: 10.1212/wnl.0000000000207371] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/17/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Patients with Lewy body disease (LBD) often show a co-occurring Alzheimer disease (AD) pathology. CSF biomarkers allow the detection in vivo of AD-related pathologic hallmarks included in the amyloid-tau-neurodegeneration (AT(N)) classification system. Here, we aimed to investigate whether CSF biomarkers of synaptic and neuroaxonal damage are correlated with the presence of AD copathology in LBD and can be useful to differentiate patients with LBD with different AT(N) profiles. METHODS We retrospectively measured CSF levels of AD core biomarkers (Aβ42/40 ratio, phosphorylated tau protein, and total tau protein) and of synaptic (β-synuclein, α-synuclein, synaptosomal-associated protein 25 [SNAP-25], and neurogranin) and neuroaxonal proteins (neurofilament light chain [NfL]) in 28 cognitively unimpaired participants with nondegenerative neurologic conditions and 161 participants with a diagnosis of either LBD or AD (at both mild cognitive impairment, AD-MCI, and dementia stages, AD-dem). We compared CSF biomarker levels in clinical and AT(N)-based subgroups. RESULTS CSF β-synuclein, α-synuclein, SNAP-25, neurogranin, and NfL levels did not differ between LBD (n = 101, age 67.2 ± 7.8 years, 27.7% females) and controls (age 64.8 ± 8.6 years, 39.3% females) and were increased in AD (AD-MCI: n = 30, AD-dem: n = 30, age 72.3 ± 6.0 years, 63.3% females) compared with both groups (p < 0.001 for all comparisons). In LBD, we found increased levels of synaptic and neuroaxonal degeneration biomarkers in patients with A+T+ (LBD/A+T+) than with A-T- profiles (LBD/A-T-) (p < 0.01 for all), and β-synuclein showed the highest discriminative accuracy between the 2 groups (area under the curve 0.938, 95% CI 0.884-0.991). CSF β-synuclein (p = 0.0021), α-synuclein (p = 0.0099), and SNAP-25 concentrations (p = 0.013) were also higher in LBD/A+T+ than in LBD/A+T- cases, which had synaptic biomarker levels within the normal range. CSF α-synuclein was significantly decreased only in patients with LBD with T- profiles compared with controls (p = 0.0448). Moreover, LBD/A+T+ and AD cases did not differ in any biomarker level. DISCUSSION LBD/A+T+ and AD cases showed significantly increased CSF levels of synaptic and neuroaxonal biomarkers compared with LBD/A-T- and control subjects. Patients with LBD and AT(N)-based AD copathology showed, thus, a distinct signature of synaptic dysfunction from other LBD cases. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that CSF levels of β-synuclein, α-synuclein, SNAP-25, neurogranin, and NfL are higher in patients with AD than in patients with LBD.
Collapse
Affiliation(s)
- Lorenzo Barba
- From the Department of Neurology (L.B., S.A.-R., P.S., M.O.), Martin-Luther-University of Halle-Wittenberg, Germany; Section of Neurology (L.B., G.B., F.P.P., L.G., L.P.), Department of Medicine and Surgery, University of Perugia, Italy; Department of Neurology (S.H., P.O., M.O.), Ulm University, Germany; German Center for Neurodegenerative Disorders Ulm (DZNE e.V.) (P.O.); and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (F.M.), University of Genoa, Italy.
| | - Samir Abu-Rumeileh
- From the Department of Neurology (L.B., S.A.-R., P.S., M.O.), Martin-Luther-University of Halle-Wittenberg, Germany; Section of Neurology (L.B., G.B., F.P.P., L.G., L.P.), Department of Medicine and Surgery, University of Perugia, Italy; Department of Neurology (S.H., P.O., M.O.), Ulm University, Germany; German Center for Neurodegenerative Disorders Ulm (DZNE e.V.) (P.O.); and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (F.M.), University of Genoa, Italy
| | - Steffen Halbgebauer
- From the Department of Neurology (L.B., S.A.-R., P.S., M.O.), Martin-Luther-University of Halle-Wittenberg, Germany; Section of Neurology (L.B., G.B., F.P.P., L.G., L.P.), Department of Medicine and Surgery, University of Perugia, Italy; Department of Neurology (S.H., P.O., M.O.), Ulm University, Germany; German Center for Neurodegenerative Disorders Ulm (DZNE e.V.) (P.O.); and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (F.M.), University of Genoa, Italy
| | - Giovanni Bellomo
- From the Department of Neurology (L.B., S.A.-R., P.S., M.O.), Martin-Luther-University of Halle-Wittenberg, Germany; Section of Neurology (L.B., G.B., F.P.P., L.G., L.P.), Department of Medicine and Surgery, University of Perugia, Italy; Department of Neurology (S.H., P.O., M.O.), Ulm University, Germany; German Center for Neurodegenerative Disorders Ulm (DZNE e.V.) (P.O.); and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (F.M.), University of Genoa, Italy
| | - Federico Paolini Paoletti
- From the Department of Neurology (L.B., S.A.-R., P.S., M.O.), Martin-Luther-University of Halle-Wittenberg, Germany; Section of Neurology (L.B., G.B., F.P.P., L.G., L.P.), Department of Medicine and Surgery, University of Perugia, Italy; Department of Neurology (S.H., P.O., M.O.), Ulm University, Germany; German Center for Neurodegenerative Disorders Ulm (DZNE e.V.) (P.O.); and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (F.M.), University of Genoa, Italy
| | - Lorenzo Gaetani
- From the Department of Neurology (L.B., S.A.-R., P.S., M.O.), Martin-Luther-University of Halle-Wittenberg, Germany; Section of Neurology (L.B., G.B., F.P.P., L.G., L.P.), Department of Medicine and Surgery, University of Perugia, Italy; Department of Neurology (S.H., P.O., M.O.), Ulm University, Germany; German Center for Neurodegenerative Disorders Ulm (DZNE e.V.) (P.O.); and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (F.M.), University of Genoa, Italy
| | - Patrick Oeckl
- From the Department of Neurology (L.B., S.A.-R., P.S., M.O.), Martin-Luther-University of Halle-Wittenberg, Germany; Section of Neurology (L.B., G.B., F.P.P., L.G., L.P.), Department of Medicine and Surgery, University of Perugia, Italy; Department of Neurology (S.H., P.O., M.O.), Ulm University, Germany; German Center for Neurodegenerative Disorders Ulm (DZNE e.V.) (P.O.); and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (F.M.), University of Genoa, Italy
| | - Petra Steinacker
- From the Department of Neurology (L.B., S.A.-R., P.S., M.O.), Martin-Luther-University of Halle-Wittenberg, Germany; Section of Neurology (L.B., G.B., F.P.P., L.G., L.P.), Department of Medicine and Surgery, University of Perugia, Italy; Department of Neurology (S.H., P.O., M.O.), Ulm University, Germany; German Center for Neurodegenerative Disorders Ulm (DZNE e.V.) (P.O.); and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (F.M.), University of Genoa, Italy
| | - Federico Massa
- From the Department of Neurology (L.B., S.A.-R., P.S., M.O.), Martin-Luther-University of Halle-Wittenberg, Germany; Section of Neurology (L.B., G.B., F.P.P., L.G., L.P.), Department of Medicine and Surgery, University of Perugia, Italy; Department of Neurology (S.H., P.O., M.O.), Ulm University, Germany; German Center for Neurodegenerative Disorders Ulm (DZNE e.V.) (P.O.); and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (F.M.), University of Genoa, Italy
| | - Lucilla Parnetti
- From the Department of Neurology (L.B., S.A.-R., P.S., M.O.), Martin-Luther-University of Halle-Wittenberg, Germany; Section of Neurology (L.B., G.B., F.P.P., L.G., L.P.), Department of Medicine and Surgery, University of Perugia, Italy; Department of Neurology (S.H., P.O., M.O.), Ulm University, Germany; German Center for Neurodegenerative Disorders Ulm (DZNE e.V.) (P.O.); and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (F.M.), University of Genoa, Italy
| | - Markus Otto
- From the Department of Neurology (L.B., S.A.-R., P.S., M.O.), Martin-Luther-University of Halle-Wittenberg, Germany; Section of Neurology (L.B., G.B., F.P.P., L.G., L.P.), Department of Medicine and Surgery, University of Perugia, Italy; Department of Neurology (S.H., P.O., M.O.), Ulm University, Germany; German Center for Neurodegenerative Disorders Ulm (DZNE e.V.) (P.O.); and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (F.M.), University of Genoa, Italy.
| |
Collapse
|
7
|
Ezzat K, Sturchio A, Espay AJ. The shift to a proteinopenia paradigm in neurodegeneration. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:23-32. [PMID: 36803814 DOI: 10.1016/b978-0-323-85555-6.00001-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
The toxic proteinopathy paradigm has defined neurodegenerative disorders for over a century. This gain-of-function (GOF) framework posited that proteins become toxic when turned into amyloids (pathology), predicting that lowering its levels would translate into clinical benefits. Genetic observations used to support a GOF framework are equally compatible with a loss-of-function (LOF) framework, as the soluble pool of proteins rendered unstable by these mutations (e.g., APP in Alzheimer's disease, SNCA in Parkinson's disease) aggregate, becoming depleted. In this review, we highlight misconceptions that have prevented LOF from gaining currency. Some of these misconceptions include no phenotype in knock-out animals (there is neurodegenerative phenotype in knock-out animals) and high levels of proteins in patients (patients have lower levels of the proteins involved in neurodegeneration than healthy age-matched controls). We also expose the internal contradictions within the GOF framework, namely that (1) pathology can have both pathogenic and protective roles; (2) the neuropathology gold standard for diagnosis can be present in normal individuals and absent in those affected; (3) oligomers are the toxic species even if they are ephemeral and decrease over time. We therefore advocate for a paradigm shift from proteinopathy (GOF) to proteinopenia (LOF) based on the universal depletion of soluble functional proteins in neurodegenerative diseases (low amyloid-β 42 in Alzheimer's disease, low α-synuclein in Parkinson's disease, and low tau in progressive supranuclear palsy) and supported by the confluence of biologic, thermodynamic, and evolutionary principles with proteins having evolved to perform a function, not to become toxic, and where protein depletion is consequential. Such shift to a Proteinopenia paradigm is necessary to examining the safety and efficacy of protein replacement strategies instead of perpetuating a therapeutic paradigm with further antiprotein permutations.
Collapse
Affiliation(s)
- Kariem Ezzat
- Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Andrea Sturchio
- Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet, Stockholm, Sweden; James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | - Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
8
|
Nabizadeh F, Pirahesh K, Ramezannezhad E. Longitudinal striatal dopamine transporter binding and cerebrospinal fluid alpha-synuclein, amyloid beta, total tau, and phosphorylated tau in Parkinson's disease. Neurol Sci 2023; 44:573-585. [PMID: 36227385 DOI: 10.1007/s10072-022-06440-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/02/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Previous studies investigated CSF levels of α-synuclein (α-syn), amyloid-β (Aβ1-42), total tau (t-tau), and phosphorylated tau (p-tau) with clinical progression of Parkinson's disease (PD). However, there is limited data on the association between CSF biomarkers and dopamine uptake status in PD. AIM In the current study, we aim to investigate the longitudinal association between striatal dopaminergic neuronal loss assessed by dopamine active transporter single photon emission computerized tomography (DaTSCAN) imaging with CSF α-syn, t-tau, p-tau, and Aβ1-42. METHODS A total of 413 early-stage PD patients and 187 healthy controls (HCs) from the PPMI. Striatal binding ratios (SBRs) of DaTSCAN images in caudate and putamen nuclei were calculated. We investigated the cross-sectional and longitudinal association between CSF biomarkers and dopamine uptake using partial correlation models adjusted for the effect of age, sex, and years of education over 24 months of follow-up. RESULTS The level of CSF α-syn, Aβ1-42, t-tau, and p-tau was significantly higher in HCs compared to PD groups at any time point. We found that higher CSF α-syn was associated with a higher SBR score in the left caudate at baseline (P = 0.038) and after 12 months (P = 0.012) in PD patients. Moreover, SBR scores in the left caudate and CSF Aβ1-42 were positively correlated at baseline (P = 0.021), 12 months (P = 0.006), and 24 months (P = 0.014) in patients with PD. Our findings demonstrated that change in CSF Aβ1-42 was positively correlated with change in SBR score in the left caudate after 24 months in the PD group (P = 0.043). CONCLUSION We found that cross-sectional levels of α-syn and Aβ1-42 could reflect the degree of dopaminergic neuron loss in the left caudate nucleus. Interestingly, longitudinal changes in CSF Aβ1-42 could predict the severity of left caudal dopaminergic neuron loss throughout the disease. This suggested that Aβ pathology might precede dopaminergic loss in striatal nuclei in this case left caudate and subsequently cognitive impairment in PD patients, although future studies are needed to confirm our results and expand the understanding of the pathophysiology of cognitive dysfunction in PD.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- Neuroscience Research Group (NRG), Universal Scientific Education and Research Network (USERN), Tehran, Iran. .,School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Kasra Pirahesh
- School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | | |
Collapse
|
9
|
Carneiro P, Loureiro JA, Delerue-Matos C, Morais S, Pereira MDC. Nanostructured label–free electrochemical immunosensor for detection of a Parkinson's disease biomarker. Talanta 2023; 252:123838. [DOI: 10.1016/j.talanta.2022.123838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 10/15/2022]
|
10
|
Luo J, Wu H, Li J, Xian W, Li W, Locascio JJ, Pei Z, Liu G. Joint Modeling Study Identifies Blood-Based Transcripts Link to Cognitive Decline in Parkinson's Disease. Mov Disord 2022; 37:2386-2395. [PMID: 36087011 DOI: 10.1002/mds.29213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Cognitive decline in Parkinson's disease (PD) is prevalent, insidious, and burdensome during the progression of the disease. OBJECTIVES We aimed to find transcriptome-wide biomarkers in blood to predict cognitive decline and identify patients at high risk with cognitive impairment in PD. METHODS We carried out joint modeling analysis to characterize transcriptome-wide longitudinal gene expression and its association with the progression of mild cognitive impairment (MCI) in PD patients. The average time-dependent area under the curves (AUCs) were used for evaluating the accuracy of the significant joint models. A cognitive survival score (CogSs) derived from joint model was leveraged to predict the occurrence of MCI. All predicting models were built in a discovery cohort with 272 patients and replicated in an independent cohort with 177 patients. RESULTS We identified five longitudinal varied expression of transcripts that were significantly associated with MCI progression in patients with PD. The most significant transcript IGLC1 joint model accurately predicted the progression of MCI in PD patients in the discovery and replication cohorts (average time-dependent AUCs >0.82). The CogSs derived from the optimal IGLC1 joint model had a high accuracy at early study stage in both cohorts (AUC ≥0.91). CONCLUSIONS Our transcriptome-wide joint modeling analysis uncovered five blood-based transcripts related to cognitive decline in PD. The joint models will serve as a useful resource for clinicians and researchers to screen PD patients with high risk of development of cognitive impairment and pave the path for Parkinson's personalized medicine. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Junfeng Luo
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Hao Wu
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Jinxia Li
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Wenbiao Xian
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weimin Li
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Joseph J Locascio
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Zhong Pei
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | |
Collapse
|
11
|
Kluge A, Bunk J, Schaeffer E, Drobny A, Xiang W, Knacke H, Bub S, Lückstädt W, Arnold P, Lucius R, Berg D, Zunke F. OUP accepted manuscript. Brain 2022; 145:3058-3071. [PMID: 35722765 DOI: 10.1093/brain/awac115] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/12/2022] [Accepted: 03/13/2022] [Indexed: 11/13/2022] Open
Abstract
To date, no reliable clinically applicable biomarker has been established for Parkinson's disease. Our results indicate that a long anticipated blood test for Parkinson's disease may be realized. Following the isolation of neuron-derived extracellular vesicles of Parkinson's disease patients and non-Parkinson's disease individuals, immunoblot analyses were performed to detect extracellular vesicle-derived α-synuclein. Pathological α-synuclein forms derived from neuronal extracellular vesicles could be detected under native conditions and were significantly increased in all individuals with Parkinson's disease and clearly distinguished disease from the non-disease state. By performing an α-synuclein seeding assay these soluble conformers could be amplified and seeding of pathological protein folding was demonstrated. Amplified α-synuclein conformers exhibited β-sheet-rich structures and a fibrillary appearance. Our study demonstrates that the detection of pathological α-synuclein conformers from neuron-derived extracellular vesicles from blood plasma samples has the potential to evolve into a blood-biomarker of Parkinson's disease that is still lacking so far. Moreover, the distribution of seeding-competent α-synuclein within blood exosomes sheds a new light of pathological disease mechanisms in neurodegenerative disorders.
Collapse
Affiliation(s)
- Annika Kluge
- Department of Neurology, University Hospital Kiel, 24105 Kiel, Germany
| | - Josina Bunk
- Institute of Biochemistry, Christian-Albrecht-University Kiel, 24118 Kiel, Germany
| | - Eva Schaeffer
- Department of Neurology, University Hospital Kiel, 24105 Kiel, Germany
| | - Alice Drobny
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Henrike Knacke
- Department of Neurology, University Hospital Kiel, 24105 Kiel, Germany
| | - Simon Bub
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Wiebke Lückstädt
- Institute of Anatomy, Christian-Albrecht-University Kiel, 24118 Kiel, Germany
| | - Philipp Arnold
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Ralph Lucius
- Institute of Anatomy, Christian-Albrecht-University Kiel, 24118 Kiel, Germany
| | - Daniela Berg
- Department of Neurology, University Hospital Kiel, 24105 Kiel, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
12
|
Chiu SY, Bowers D, Armstrong MJ. Lewy Body Dementias: Controversies and Drug Development. Neurotherapeutics 2022; 19:55-67. [PMID: 34859379 PMCID: PMC9130410 DOI: 10.1007/s13311-021-01161-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2021] [Indexed: 01/03/2023] Open
Abstract
Lewy body dementia (LBD) is one of the most common neurodegenerative dementias. Clinical trials for symptomatic and disease-modifying therapies in LBD remain a national research priority, but there are many challenges in both past and active drug developments in LBD. This review highlights the controversies in picking the appropriate populations, interventions, target selections, and outcome measures, which are all critical components of clinical trial implementation in LBD. The heterogeneity of LBD neuropathology and clinical presentations, limited understanding of core features such as cognitive fluctuations, and lack of validated LBD-specific outcome measures and biomarkers represent some of the major challenges in LBD trials.
Collapse
Affiliation(s)
- Shannon Y Chiu
- Department of Neurology, University of Florida, PO Box 100268, Gainesville, FL, 32611, USA.
| | - Dawn Bowers
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, 32603, USA
| | - Melissa J Armstrong
- Department of Neurology, University of Florida, PO Box 100268, Gainesville, FL, 32611, USA
| |
Collapse
|
13
|
Enache D, Pereira JB, Jelic V, Winblad B, Nilsson P, Aarsland D, Bereczki E. Increased Cerebrospinal Fluid Concentration of ZnT3 Is Associated with Cognitive Impairment in Alzheimer's Disease. J Alzheimers Dis 2021; 77:1143-1155. [PMID: 32925049 DOI: 10.3233/jad-200498] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cognitive deficits arising in the course of Alzheimer's disease (AD), dementia with Lewy bodies (DLB), and Parkinson's disease with dementia (PDD) are directly linked to synaptic loss. Postmortem studies suggest that zinc transporter protein 3 (ZnT3), AMPA glutamate receptor 3 (GluA3), and Dynamin1 are associated with cognitive decline in AD and Lewy body dementia patients. OBJECTIVE We aimed to evaluate the diagnostic value of ZnT3, GluA3, and Dynamin 1 in the cerebrospinal fluid (CSF) of patients with dementia due to AD, DLB, and PDD compared to cognitively normal subjective cognitive decline (SCD) patients in a retrospective study. In addition, we assessed the relationship between synaptic markers and age, sex, cognitive impairment, and depressive symptoms as well as CSF amyloid, phosphorylated tau (p-tau), and total tau (T-tau). METHODS Commercially available ELISA immunoassay was used to measure the levels of proteins in a total of 97 CSF samples from AD (N = 24), PDD (N = 18), DLB (N = 27), and SCD (N = 28) patients. Cognitive impairment was assessed using the Mini-Mental State Examination (MMSE). RESULTS We found a significant increase in the concentrations of ZnT3, GluA3, and Dynamin1 in AD (p = 0.002) and of ZnT3 and Dynamin 1 in DLB (p = 0.001, p = 0.002) when compared to SCD patients. Changes in ZnT3 concentrations correlated with MMSE scores in AD (p = 0.011), and with depressive symptoms in SCD (p = 0.041). CONCLUSION We found alteration of CSF levels of synaptic proteins in AD, PDD, and DLB. Our results reveal distinct changes in CSF concentrations of ZnT3 that could reflect cognitive impairment in AD with implications for future prognostic and diagnostic marker development.
Collapse
Affiliation(s)
- Daniela Enache
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Joana B Pereira
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Vesna Jelic
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Dag Aarsland
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden.,Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Erika Bereczki
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
14
|
Oliveira LMA, Gasser T, Edwards R, Zweckstetter M, Melki R, Stefanis L, Lashuel HA, Sulzer D, Vekrellis K, Halliday GM, Tomlinson JJ, Schlossmacher M, Jensen PH, Schulze-Hentrich J, Riess O, Hirst WD, El-Agnaf O, Mollenhauer B, Lansbury P, Outeiro TF. Alpha-synuclein research: defining strategic moves in the battle against Parkinson's disease. NPJ Parkinsons Dis 2021; 7:65. [PMID: 34312398 PMCID: PMC8313662 DOI: 10.1038/s41531-021-00203-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 05/14/2021] [Indexed: 12/13/2022] Open
Abstract
With the advent of the genetic era in Parkinson's disease (PD) research in 1997, α-synuclein was identified as an important player in a complex neurodegenerative disease that affects >10 million people worldwide. PD has been estimated to have an economic impact of $51.9 billion in the US alone. Since the initial association with PD, hundreds of researchers have contributed to elucidating the functions of α-synuclein in normal and pathological states, and these remain critical areas for continued research. With this position paper the authors strive to achieve two goals: first, to succinctly summarize the critical features that define α-synuclein's varied roles, as they are known today; and second, to identify the most pressing knowledge gaps and delineate a multipronged strategy for future research with the goal of enabling therapies to stop or slow disease progression in PD.
Collapse
Affiliation(s)
- Luis M A Oliveira
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA.
| | - Thomas Gasser
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Robert Edwards
- Departments of Neurology and Physiology, UCSF School of Medicine, San Francisco, CA, USA
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department for NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Ronald Melki
- Institut François Jacob, MIRCen, CEA and Laboratory of Neurodegenerative Diseases, CNRS, Fontenay-aux-Roses, France
| | - Leonidas Stefanis
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- First Department of Neurology, Medical School of the National and Kapodistrian University of Athens, Athens, Greece
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Faculty of Life Sciences, EPFL, Lausanne, Switzerland
| | - David Sulzer
- Department of Psychiatry, Neurology, Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Kostas Vekrellis
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Glenda M Halliday
- University of Sydney, Brain and Mind Centre and Faculty of Medicine and Health, School of Medical Sciences, Sydney, NSW, Australia
| | - Julianna J Tomlinson
- Neuroscience Program, The Ottawa Hospital, Ottawa, ON, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Michael Schlossmacher
- Neuroscience Program, The Ottawa Hospital, Ottawa, ON, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
- Division of Neurology, The Ottawa Hospital, Ottawa, ON, Canada
| | - Poul Henning Jensen
- Aarhus University, Department of Biomedicine & DANDRITE, Danish Research Institute of Translational Neuroscience, Aarhus, Denmark
| | - Julia Schulze-Hentrich
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Warren D Hirst
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, USA
| | - Omar El-Agnaf
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Paracelsus-Elena-Klinik, Kassel, Germany
| | | | - Tiago F Outeiro
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.
- Max Planck Institute for Experimental Medicine, Göttingen, Germany.
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK.
| |
Collapse
|
15
|
Fernández-Espejo E, Rodriguez de Fonseca F, Suárez J, González-Aparicio R, Santurtún A. ATP13A2 levels in serum and cerebrospinal fluid in patients with idiopathic Parkinson's disease. Parkinsonism Relat Disord 2021; 88:3-9. [PMID: 34090180 DOI: 10.1016/j.parkreldis.2021.05.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 05/11/2021] [Accepted: 05/14/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND The enzyme ATP13A2 holds promise as biomarker in Parkinson's disease (PD). No study has examined the content of ATP13A2 in serum and cerebrospinal fluid (CSF) in idiopathic PD cohorts, or how ATP13A2 relates to the clinical features of the disease. METHODS ATP13A2 concentration was evaluated with ELISA and immunoblotting. Correlations of serum and CSF ATP13A2 with clinical parameters were examined. The antiparkinsonian medication regimen was expressed as levodopa equivalent dose (LED, mg/day). RESULTS Serum ATP13A2 concentration was similar in patients and controls, and it correlated with LED and MDS-UPDRS part-IV score (p < .0001), a scale which allows evaluating motor complications. LED also correlated with MDS-UPDRS part-IV score (p < .0001). Serum ATP13A2 concentration and LED were higher in patients with motor complications than in patients without motor complications (p < .0001). The ratio of serum ATP13A2 concentration versus LED was calculated, and mean value was similar in patients with or without motor complications. ATP13A2 concentration in the CSF was undetectable in many subjects because the ELISA assay was hampered by its detection limit. Immunoblotting indicated that CSF ATP13A2 content was higher in patients relative to controls (p = .0002), and no clinical correlations were found. CONCLUSIONS Increasing LED enhanced serum ATP13A2 concentration and facilitated the development of motor complications. There is a direct relationship between serum ATP13A2 level and the dose intensity of the antiparkinsonian dopaminergic medication. The associations between serum ATP13A2 and LED suggest that serum ATP13A2 content might be a marker of dopamine replacement therapy.
Collapse
Affiliation(s)
- Emilio Fernández-Espejo
- Reial Acadèmia de Medicina de Catalunya, 08010, Barcelona, Spain; Red Andaluza de Investigación Clínica y Traslacional en Neurología (Neuro-RECA), Laboratorio de Medicina Regenerativa, Hospital Regional Universitario, 29010, Málaga, Spain.
| | - Fernando Rodriguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario, Instituto de Investigación Biomédica de Málaga (IBIMA), 29010, Málaga, Spain; Red Andaluza de Investigación Clínica y Traslacional en Neurología (Neuro-RECA), Laboratorio de Medicina Regenerativa, Hospital Regional Universitario, 29010, Málaga, Spain
| | - Juan Suárez
- Departamento de Anatomía Humana, Medicina Legal e Historia de La Ciencia, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, 29071, Málaga, Spain; Red Andaluza de Investigación Clínica y Traslacional en Neurología (Neuro-RECA), Laboratorio de Medicina Regenerativa, Hospital Regional Universitario, 29010, Málaga, Spain
| | - Ramiro González-Aparicio
- Departamento de Ciencias, San Francisco de Paula - Sevilla International College, 41003, Sevilla, Spain
| | - Ana Santurtún
- Unidad de Medicina Legal, Departamento de Fisiología y Farmacología, Universidad de Cantabria, Santander, Spain
| |
Collapse
|
16
|
Bargar C, Wang W, Gunzler SA, LeFevre A, Wang Z, Lerner AJ, Singh N, Tatsuoka C, Appleby B, Zhu X, Xu R, Haroutunian V, Zou WQ, Ma J, Chen SG. Streamlined alpha-synuclein RT-QuIC assay for various biospecimens in Parkinson's disease and dementia with Lewy bodies. Acta Neuropathol Commun 2021; 9:62. [PMID: 33827706 PMCID: PMC8028088 DOI: 10.1186/s40478-021-01175-w] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 03/31/2021] [Indexed: 11/28/2022] Open
Abstract
Definitive diagnosis of Parkinson's disease (PD) and dementia with Lewy bodies (DLB) relies on postmortem finding of disease-associated alpha-synuclein (αSynD) as misfolded protein aggregates in the central nervous system (CNS). The recent development of the real-time quaking induced conversion (RT-QuIC) assay for ultrasensitive detection of αSynD aggregates has revitalized the diagnostic values of clinically accessible biospecimens, including cerebrospinal fluid (CSF) and peripheral tissues. However, the current αSyn RT-QuIC assay platforms vary widely and are thus challenging to implement and standardize the measurements of αSynD across a wide range of biospecimens and in different laboratories. We have streamlined αSyn RT-QuIC assay based on a second generation assay platform that was assembled entirely with commercial reagents. The streamlined RT-QuIC method consisted of a simplified protocol requiring minimal hands-on time, and allowing for a uniform analysis of αSynD in different types of biospecimens from PD and DLB. Ultrasensitive and specific RT-QuIC detection of αSynD aggregates was achieved in million-fold diluted brain homogenates and in nanoliters of CSF from PD and DLB cases but not from controls. Comparative analysis revealed higher seeding activity of αSynD in DLB than PD in both brain homogenates and CSF. Our assay was further validated with CSF samples of 214 neuropathologically confirmed cases from tissue repositories (88 PD, 58 DLB, and 68 controls), yielding a sensitivity of 98% and a specificity of 100%. Finally, a single RT-QuIC assay protocol was employed uniformly to detect seeding activity of αSynD in PD samples across different types of tissues including the brain, skin, salivary gland, and colon. We anticipate that our streamlined protocol will enable interested laboratories to easily and rapidly implement the αSyn RT-QuIC assay for various clinical specimens from PD and DLB. The utilization of commercial products for all assay components will improve the robustness and standardization of the RT-QuIC assay for diagnostic applications across different sites. Due to ultralow sample consumption, the ultrasensitive RT-QuIC assay will facilitate efficient use and sharing of scarce resources of biospecimens. Our streamlined RT-QuIC assay is suitable to track the distribution of αSynD in CNS and peripheral tissues of affected patients. The ongoing evaluation of RT-QuIC assay of αSynD as a potential biomarker for PD and DLB in clinically accessible biospecimens has broad implications for understanding disease pathogenesis, improving early and differential diagnosis, and monitoring therapeutic efficacies in clinical trials.
Collapse
Affiliation(s)
- Connor Bargar
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Wen Wang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Steven A Gunzler
- Department of Neurology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Alexandra LeFevre
- University of Maryland Brain and Tissue Bank, Baltimore, MD, 21201, USA
| | - Zerui Wang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Alan J Lerner
- Department of Neurology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Neena Singh
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Department of Neurology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Curtis Tatsuoka
- Department of Neurology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Brian Appleby
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Department of Neurology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Department of Neurology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Rong Xu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Vahram Haroutunian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Wen-Quan Zou
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
- Department of Neurology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Jiyan Ma
- Van Andel Institute, Grand Rapids, MI, 49503, USA.
| | - Shu G Chen
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
- Department of Neurology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
17
|
Zenuni H, Grillo P, Sancesario GM, Bernardini S, Mercuri NB, Schirinzi T. How Comorbidity Reflects on Cerebrospinal Fluid Biomarkers of Neurodegeneration in Aging. J Alzheimers Dis Rep 2021; 5:87-92. [PMID: 33681720 PMCID: PMC7902985 DOI: 10.3233/adr-200280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2020] [Indexed: 12/23/2022] Open
Abstract
Systemic comorbidity precipitates the risk for dementia. To comprehend the underlying mechanisms into a therapeutic perspective, we analyzed how comorbidity affects neurodegeneration-related cerebrospinal fluid (CSF) biomarkers of 55 cognitively intact subjects. The Charson Comorbidity Index (CCI) was correlated with CSF amyloid-β42 (Aβ42), amyloid-β40, total-tau, 181-phosphorylated-tau (p-tau), the Aβ42/p-tau ratio, neurogranin, and lactate. The age-related brain lesions at imaging were also considered. CCI had a raw association with Aβ42/p-tau and p-tau, and a stronger, age-independent correlation with lactate. These preliminary findings suggested that, in normal subjects, systemic comorbidity might increase CNS oxidative stress and, together with aging, contribute to develop an Alzheimer's disease-like biochemical profile.
Collapse
Affiliation(s)
- Henri Zenuni
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Piergiorgio Grillo
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | | | - Sergio Bernardini
- Department of Experimental Medicine, University of Roma Tor Vergata, Rome, Italy
- Department of Clinical Biochemistry, Tor Vergata University Hospital, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
- IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Tommaso Schirinzi
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| |
Collapse
|
18
|
CSF Biomarkers Reflecting Protein Pathology and Axonal Degeneration Are Associated with Memory, Attentional, and Executive Functioning in Early-Stage Parkinson's Disease. Int J Mol Sci 2020; 21:ijms21228519. [PMID: 33198266 PMCID: PMC7697681 DOI: 10.3390/ijms21228519] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 01/21/2023] Open
Abstract
In early-stage Parkinson′s disease (PD), cognitive impairment is common, and a variety of cognitive domains including memory, attention, and executive functioning may be affected. Cerebrospinal fluid (CSF) biomarkers are potential markers of cognitive functioning. We aimed to explore whether CSF α-synuclein species, neurofilament light chain, amyloid-β42, and tau are associated with cognitive performance in early-stage PD patients. CSF levels of total-α-synuclein and phosphorylated-α-synuclein, neurofilament light chain, amyloid-β42, and total-tau and phosphorylated-tau were measured in 26 PD patients (disease duration ≤5 years and Hoehn and Yahr stage 1–2.5). Multivariable linear regression models, adjusted for age, gender, and educational level, were used to assess the relationship between CSF biomarker levels and memory, attention, executive and visuospatial function, and language performance scores. In 26 early-stage PD patients, attention and memory were the most commonly affected domains. A higher CSF phosphorylated-α-synuclein/total-α-synuclein ratio was associated with better executive functioning (sβ = 0.40). Higher CSF neurofilament light was associated with worse memory (sβ = −0.59), attentional (sβ = −0.32), and executive functioning (sβ = −0.35). Reduced CSF amyloid-β42 levels were associated with poorer attentional functioning (sβ = 0.35). Higher CSF phosphorylated-tau was associated with worse language functioning (sβ = −0.33). Thus, CSF biomarker levels, in particular neurofilament light, were related to the most commonly affected cognitive domains in early-stage PD. This indicates that CSF biomarker levels may identify early-stage PD patients who are at an increased risk of developing cognitive impairment.
Collapse
|
19
|
Goldman JG, Forsberg LK, Boeve BF, Armstrong MJ, Irwin DJ, Ferman TJ, Galasko D, Galvin JE, Kaufer D, Leverenz J, Lippa CF, Marder K, Abler V, Biglan K, Irizarry M, Keller B, Munsie L, Nakagawa M, Taylor A, Graham T. Challenges and opportunities for improving the landscape for Lewy body dementia clinical trials. Alzheimers Res Ther 2020; 12:137. [PMID: 33121510 PMCID: PMC7597002 DOI: 10.1186/s13195-020-00703-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/08/2020] [Indexed: 01/05/2023]
Abstract
Lewy body dementia (LBD), including dementia with Lewy bodies and Parkinson's disease dementia, affects over a million people in the USA and has a substantial impact on patients, caregivers, and society. Symptomatic treatments for LBD, which can include cognitive, neuropsychiatric, autonomic, sleep, and motor features, are limited with only two drugs (cholinesterase inhibitors) currently approved by regulatory agencies for dementia in LBD. Clinical trials represent a top research priority, but there are many challenges in the development and implementation of trials in LBD. To address these issues and advance the field of clinical trials in the LBDs, the Lewy Body Dementia Association formed an Industry Advisory Council (LBDA IAC), in addition to its Research Center of Excellence program. The LBDA IAC comprises a diverse and collaborative group of experts from academic medical centers, pharmaceutical industries, and the patient advocacy foundation. The inaugural LBDA IAC meeting, held in June 2019, aimed to bring together this group, along with representatives from regulatory agencies, to address the topic of optimizing the landscape of LBD clinical trials. This review highlights the formation of the LBDA IAC, current state of LBD clinical trials, and challenges and opportunities in the field regarding trial design, study populations, diagnostic criteria, and biomarker utilization. Current gaps include a lack of standardized clinical assessment tools and evidence-based management strategies for LBD as well as difficulty and controversy in diagnosing LBD. Challenges in LBD clinical trials include the heterogeneity of LBD pathology and symptomatology, limited understanding of the trajectory of LBD cognitive and core features, absence of LBD-specific outcome measures, and lack of established standardized biologic, imaging, or genetic biomarkers that may inform study design. Demands of study participation (e.g., travel, duration, and frequency of study visits) may also pose challenges and impact trial enrollment, retention, and outcomes. There are opportunities to improve the landscape of LBD clinical trials by harmonizing clinical assessments and biomarkers across cohorts and research studies, developing and validating outcome measures in LBD, engaging the patient community to assess research needs and priorities, and incorporating biomarker and genotype profiling in study design.
Collapse
Affiliation(s)
- Jennifer G Goldman
- Parkinson's Disease and Movement Disorders Program, Shirley Ryan AbilityLab and Departments of Physical Medicine and Rehabilitation and Neurology, Northwestern University Feinberg School of Medicine, 355 E. Erie Street, Chicago, IL, 60611, USA.
| | | | | | - Melissa J Armstrong
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL, USA
| | - David J Irwin
- Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Tanis J Ferman
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, USA
| | - Doug Galasko
- Department of Neurosciences, UC San Diego, San Diego, CA, USA
| | - James E Galvin
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Daniel Kaufer
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - James Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Carol F Lippa
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Karen Marder
- Department of Neurology, Taub Institute, Sergievsky Center, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Kevin Biglan
- Neuroscience Research, Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | - Leanne Munsie
- Neuroscience Research, Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Angela Taylor
- Lewy Body Dementia Association, S.W., Lilburn, GA, USA
| | - Todd Graham
- Lewy Body Dementia Association, S.W., Lilburn, GA, USA
| |
Collapse
|
20
|
Farotti L, Paolini Paoletti F, Simoni S, Parnetti L. Unraveling Pathophysiological Mechanisms of Parkinson's Disease: Contribution of CSF Biomarkers. Biomark Insights 2020; 15:1177271920964077. [PMID: 33110345 PMCID: PMC7555566 DOI: 10.1177/1177271920964077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 01/08/2023] Open
Abstract
Diagnosis of Parkinson's disease (PD) relies on clinical history and physical examination, but misdiagnosis is common in early stages. Identification of biomarkers for PD may allow for early and more precise diagnosis and provide information about prognosis. Developments in analytical chemistry allow for the detection of a large number of molecules in cerebrospinal fluid (CSF), which are known to be associated with the pathogenesis of PD. Given the pathophysiology of PD, CSF α-synuclein species have the strongest rationale for use, also providing encouraging preliminary results in terms of early diagnosis. In the field of classical Alzheimer's disease (AD) biomarkers, low CSF Aβ42 levels have shown a robust prognostic value in terms of development of cognitive impairment. Other CSF biomarkers including lysosomal enzymes, neurofilament light chain, markers of neuroinflammation and oxidative stress, although promising, have not proved to be reliable for diagnostic and prognostic purposes yet. Overall, the implementation of CSF biomarkers may give a substantial contribution to the optimal use of disease-modifying drugs.
Collapse
Affiliation(s)
- Lucia Farotti
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | | | - Simone Simoni
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
21
|
Gmitterová K, Gawinecka J, Llorens F, Varges D, Valkovič P, Zerr I. Cerebrospinal fluid markers analysis in the differential diagnosis of dementia with Lewy bodies and Parkinson's disease dementia. Eur Arch Psychiatry Clin Neurosci 2020; 270:461-470. [PMID: 30083957 DOI: 10.1007/s00406-018-0928-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/16/2018] [Indexed: 01/18/2023]
Abstract
Dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD) share a couple of clinical similarities that is often a source of diagnostic pitfalls. We evaluated the discriminatory potential of brain-derived CSF markers [tau, p-tau (181P), Aβ1-42, NSE and S100B] across the spectrum of Lewy body disorders and assessed whether particular markers are associated with cognitive status in investigated patients. The tau CSF level, amyloid β1-42 and p-tau/tau ratio were helpful in the distinction between DLB and PDD (p = 0.04, p = 0.002 and p = 0.02, respectively) as well as from PD patients (p < 0.001, p = 0.001 and p = 0.002, respectively). Furthermore, the p-tau/tau ratio enabled the differentiation of DLB with mild dementia from PDD patients (p = 0.02). The CSF tau and p-tau levels in DLB and CSF tau and p-tau/tau ratio in PDD patients reflected the severity of dementia. Rapid disease course was associated with the decrease of Aβ1-42 in DLB but not in PDD. Elevation of S100B in DLB (p < 0.0001) as well as in PDD patients (p = 0.002) in comparison to controls was estimated. Hence, with the appropriate clinical context; the CSF marker profile could be helpful in distinguishing DLB from PDD patients even in early stages of dementia.
Collapse
Affiliation(s)
- Karin Gmitterová
- Department of Neurology, Clinical Dementia Center and DZNE, National TSE Reference Centre, University Medical School, Georg-August University, Robert-Koch-Str. 40, 37073, Göttingen, Germany
- Second Department of Neurology, Comenius University, Bratislava, Slovakia
| | - Joanna Gawinecka
- Department of Neurology, Clinical Dementia Center and DZNE, National TSE Reference Centre, University Medical School, Georg-August University, Robert-Koch-Str. 40, 37073, Göttingen, Germany
- Institute for Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland
| | - Franc Llorens
- Department of Neurology, Clinical Dementia Center and DZNE, National TSE Reference Centre, University Medical School, Georg-August University, Robert-Koch-Str. 40, 37073, Göttingen, Germany
| | - Daniela Varges
- Department of Neurology, Clinical Dementia Center and DZNE, National TSE Reference Centre, University Medical School, Georg-August University, Robert-Koch-Str. 40, 37073, Göttingen, Germany
| | - Peter Valkovič
- Second Department of Neurology, Comenius University, Bratislava, Slovakia
| | - Inga Zerr
- Department of Neurology, Clinical Dementia Center and DZNE, National TSE Reference Centre, University Medical School, Georg-August University, Robert-Koch-Str. 40, 37073, Göttingen, Germany.
| |
Collapse
|
22
|
Schirinzi T, Di Lazzaro G, Sancesario GM, Summa S, Petrucci S, Colona VL, Bernardini S, Pierantozzi M, Stefani A, Mercuri NB, Pisani A. Young-onset and late-onset Parkinson's disease exhibit a different profile of fluid biomarkers and clinical features. Neurobiol Aging 2020; 90:119-124. [PMID: 32169356 DOI: 10.1016/j.neurobiolaging.2020.02.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 12/11/2022]
Abstract
Young-onset Parkinson's disease (YOPD) is a relevant condition whose neurobiology is questioned if different from those of typical late-onset Parkinson's disease (LOPD). Here, we explored whether the clinical-biochemical profile of Parkinson's disease (PD) could be affected by the age-of-onset (AO), as a possible result of a distinct neurodegenerative process. A panel of fluid biomarkers (CSF lactate, 42-amyloid-β peptide, total and 181-phosphorylated tau; serum uric acid) and the standard scores for motor and nonmotor signs were assessed in 76 idiopathic PD patients (genetic cases excluded; YOPD, AO ≤ 50, n = 44; LOPD, AO > 50, n = 32) and 75 sex/age-matched controls, adjusting the models for the main confounding factors. In PD, AO directly correlated to either CSF lactate and tau proteins or the nonmotor symptoms scale score. Specifically, a younger AO was associated with lower levels of biomarkers and minor burden of nonmotor symptoms. Our findings indicate that clinical-biochemical features of idiopathic PD may vary depending on the AO, accounting for different profiles in YOPD and LOPD whose recognition is fundamental for further pathophysiological implications and clinical applications.
Collapse
Affiliation(s)
- Tommaso Schirinzi
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; Department of Neurosciences, IRCCS Bambino Gesù Children's Hospital, Rome, Italy.
| | - Giulia Di Lazzaro
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Giulia Maria Sancesario
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Rome, Italy; Department of Experimental Medicine and Surgery, University of Roma Tor Vergata, Rome, Italy
| | - Susanna Summa
- Department of Neurosciences, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Simona Petrucci
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy; Department of Clinical and Molecular Medicine, S. Andrea University Hospital, Rome, Italy; IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Vito Luigi Colona
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine and Surgery, University of Roma Tor Vergata, Rome, Italy
| | | | - Alessandro Stefani
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Antonio Pisani
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
23
|
Abstract
Parkinson's disease (PD) is a chronic, debilitating neurodegenerative disorder characterized clinically by a variety of progressive motor and nonmotor symptoms. Currently, there is a dearth of diagnostic tools available to predict, diagnose or mitigate disease risk or progression, leading to a challenging dilemma within the healthcare management system. The search for a reliable biomarker for PD that reflects underlying pathology is a high priority in PD research. Currently, there is no reliable single biomarker predictive of risk for motor and cognitive decline, and there have been few longitudinal studies of temporal progression. A combination of multiple biomarkers might facilitate earlier diagnosis and more accurate prognosis in PD. In this review, we focus on the recent developments of serial biomarkers for PD from a variety of clinical, biochemical, genetic and neuroimaging perspectives.
Collapse
Affiliation(s)
- Anastasia Bougea
- Neurochemistry Laboratory, 1st Department of Neurology and Movement Disorders, Medical School, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece; Neuroscience Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
24
|
Coughlin DG, Hurtig H, Irwin DJ. Pathological Influences on Clinical Heterogeneity in Lewy Body Diseases. Mov Disord 2020; 35:5-19. [PMID: 31660655 PMCID: PMC7233798 DOI: 10.1002/mds.27867] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/06/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022] Open
Abstract
PD, PD with dementia, and dementia with Lewy bodies are clinical syndromes characterized by the neuropathological accumulation of alpha-synuclein in the CNS that represent a clinicopathological spectrum known as Lewy body disorders. These clinical entities have marked heterogeneity of motor and nonmotor symptoms with highly variable disease progression. The biological basis for this clinical heterogeneity remains poorly understood. Previous attempts to subtype patients within the spectrum of Lewy body disorders have centered on clinical features, but converging evidence from studies of neuropathology and ante mortem biomarkers, including CSF, neuroimaging, and genetic studies, suggest that Alzheimer's disease beta-amyloid and tau copathology strongly influence clinical heterogeneity and prognosis in Lewy body disorders. Here, we review previous clinical biomarker and autopsy studies of Lewy body disorders and propose that Alzheimer's disease copathology is one of several likely pathological contributors to clinical heterogeneity of Lewy body disorders, and that such pathology can be assessed in vivo. Future work integrating harmonized assessments and genetics in PD, PD with dementia, and dementia with Lewy bodies patients followed to autopsy will be critical to further refine the classification of Lewy body disorders into biologically distinct endophenotypes. This approach will help facilitate clinical trial design for both symptomatic and disease-modifying therapies to target more homogenous subsets of Lewy body disorders patients with similar prognosis and underlying biology. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- David G Coughlin
- University of Pennsylvania Health System, Department of Neurology
- Digital Neuropathology Laboratory
- Lewy Body Disease Research Center of Excellence
| | - Howard Hurtig
- University of Pennsylvania Health System, Department of Neurology
| | - David J Irwin
- University of Pennsylvania Health System, Department of Neurology
- Digital Neuropathology Laboratory
- Lewy Body Disease Research Center of Excellence
- Frontotemporal Degeneration Center, Philadelphia PA, USA 19104
| |
Collapse
|
25
|
Irwin DJ. Neuropathological Validation of Cerebrospinal Fluid Biomarkers in Neurodegenerative Diseases. J Appl Lab Med 2019; 5:jalm.2019.029876. [PMID: 31811076 DOI: 10.1373/jalm.2019.029876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022]
Affiliation(s)
- David J Irwin
- Penn Digital Neuropathology Laboratory
- Penn Frontotemporal Degeneration Center, Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
26
|
Zhou C, Guan XJ, Guo T, Zeng QL, Gao T, Huang PY, Xuan M, Gu QQ, Xu XJ, Zhang MM. Progressive brain atrophy in Parkinson's disease patients who convert to mild cognitive impairment. CNS Neurosci Ther 2019; 26:117-125. [PMID: 31278861 PMCID: PMC6930819 DOI: 10.1111/cns.13188] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/23/2019] [Accepted: 06/14/2019] [Indexed: 12/28/2022] Open
Abstract
Aims Cognitive impairment is a common symptom in the trajectory of Parkinson's disease (PD). However, the pathological underpinning is not fully known. We aimed to explore the critical structural alterations in the process of cognitive decline and its relationships with the dopaminergic deficit and the level of related cerebrospinal fluid (CSF) proteins. Methods Ninety‐four patients with PD and 32 controls were included in this study. Neuropsychological tests were performed at baseline and after 28 months to identify which patients had normal cognition and which ones developed PD‐MCI after follow‐up (“converters”). Gray matter atrophy was assessed in cross‐sectional and longitudinal analyses, respectively. The associations between altered GMV with dopamine transporter (DAT) results and the level of CSF proteins were assessed. Results Among the 94 patients with normal cognition at baseline, 24 (mean age, 63.1 years) developed PD‐MCI after 28 months of follow‐up, and 70 (mean age, 62.3 years) remained nonconverters. The converters showed significant right temporal atrophy at baseline and extensive atrophy in temporal lobe at follow‐up. Progressive bilateral frontal lobe atrophy was found in the converters. Baseline right temporal atrophy was correlated with the striatal dopaminergic degeneration in the converters. No correlation was found between the right temporal atrophy and the alterations of CSF proteins. Conclusion Early atrophy in temporal lobes and progressive atrophy in frontal lobes might be a biomarker for developing multidomain impairment of cognition and converting to PD‐MCI. Furthermore, cognition‐related temporal atrophy might be associated with dopaminergic deficit reflected by DAT scan but independent of CSF proteins in patients with PD who convert to PD‐MCI.
Collapse
Affiliation(s)
- Cheng Zhou
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-Jun Guan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Guo
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiao-Ling Zeng
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Gao
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pei-Yu Huang
- 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
| | - Quan-Quan Gu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-Jun Xu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Min-Ming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
27
|
Parnetti L, Gaetani L, Eusebi P, Paciotti S, Hansson O, El-Agnaf O, Mollenhauer B, Blennow K, Calabresi P. CSF and blood biomarkers for Parkinson's disease. Lancet Neurol 2019; 18:573-586. [PMID: 30981640 DOI: 10.1016/s1474-4422(19)30024-9] [Citation(s) in RCA: 349] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/21/2018] [Accepted: 01/15/2019] [Indexed: 01/09/2023]
Abstract
In the management of Parkinson's disease, reliable diagnostic and prognostic biomarkers are urgently needed. The diagnosis of Parkinson's disease mostly relies on clinical symptoms, which hampers the detection of the earliest phases of the disease-the time at which treatment with forthcoming disease-modifying drugs could have the greatest therapeutic effect. Reliable prognostic markers could help in predicting the response to treatments. Evidence suggests potential diagnostic and prognostic value of CSF and blood biomarkers closely reflecting the pathophysiology of Parkinson's disease, such as α-synuclein species, lysosomal enzymes, markers of amyloid and tau pathology, and neurofilament light chain. A combination of multiple CSF biomarkers has emerged as an accurate diagnostic and prognostic model. With respect to early diagnosis, the measurement of CSF α-synuclein aggregates is providing encouraging preliminary results. Blood α-synuclein species and neurofilament light chain are also under investigation because they would provide a non-invasive tool, both for early and differential diagnosis of Parkinson's disease versus atypical parkinsonian disorders, and for disease monitoring. In view of adopting CSF and blood biomarkers for improving Parkinson's disease diagnostic and prognostic accuracy, further validation in large independent cohorts is needed.
Collapse
Affiliation(s)
- Lucilla Parnetti
- Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine, University of Perugia, Perugia, Italy.
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Paolo Eusebi
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Silvia Paciotti
- Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine, University of Perugia, Perugia, Italy; Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Omar El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Education City, Doha, Qatar
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany; University Medical Center, Department of Neurology, Göttingen, Germany
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Paolo Calabresi
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
28
|
Maass F, Schulz I, Lingor P, Mollenhauer B, Bähr M. Cerebrospinal fluid biomarker for Parkinson's disease: An overview. Mol Cell Neurosci 2018; 97:60-66. [PMID: 30543858 DOI: 10.1016/j.mcn.2018.12.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/06/2018] [Accepted: 12/08/2018] [Indexed: 01/01/2023] Open
Abstract
In Parkinson's disease (PD), there is a wide field of recent and ongoing search for useful biomarkers for early and differential diagnosis, disease monitoring or subtype characterization. Up to now, no biofluid biomarker has entered the daily clinical routine. Cerebrospinal fluid (CSF) is often used as a source for biomarker development in different neurological disorders because it reflects changes in central-nervous system homeostasis. This review article gives an overview about different biomarker approaches in PD, mainly focusing on CSF analyses. Current state and future perspectives regarding classical protein markers like alpha‑synuclein, but also different "omics" techniques are described. In conclusion, technical advancements in the field already yielded promising results, but further multicenter trials with well-defined cohorts, standardized protocols and integrated data analysis of different modalities are needed before successful translation into routine clinical application.
Collapse
Affiliation(s)
- Fabian Maass
- University Medical Center, Department of Neurology, Robert-Koch Strasse 40, 37075 Goettingen, Germany.
| | - Isabel Schulz
- University of Southampton, Faculty of Medicine, 12 University Rd, Southampton SO17 1BJ, United Kingdom
| | - Paul Lingor
- Department of Neurology, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Straße 22, 81675 Munich, Germany
| | - Brit Mollenhauer
- University Medical Center, Department of Neurology, Robert-Koch Strasse 40, 37075 Goettingen, Germany; Paracelsus-Elena-Klinik, Klinikstrasse 16, 24128 Kassel, Germany
| | - Mathias Bähr
- University Medical Center, Department of Neurology, Robert-Koch Strasse 40, 37075 Goettingen, Germany
| |
Collapse
|
29
|
Kalia LV. Diagnostic biomarkers for Parkinson's disease: focus on α-synuclein in cerebrospinal fluid. Parkinsonism Relat Disord 2018; 59:21-25. [PMID: 30466800 DOI: 10.1016/j.parkreldis.2018.11.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/09/2018] [Accepted: 11/14/2018] [Indexed: 11/24/2022]
Abstract
Diagnostic biomarkers are measures that detect or confirm the presence of a disease or identify individuals with a subtype of the disease. For Parkinson's disease, unlike other neurodegenerative diseases such as Alzheimer's disease and Creutzfeldt-Jakob disease, diagnostic biomarkers remain elusive as none are yet available or approved for clinical use. A biomarker to diagnose early or prodromal Parkinson's disease with high accuracy would significantly enhance clinical practice as well as advance clinical therapeutic trials. Multiple lines of evidence support a role of α-synuclein in the pathophysiology of Parkinson's disease and hence major ongoing efforts to identify biomarkers for Parkinson's disease are aimed at measuring α-synuclein in peripheral tissues and biofluids, including cerebrospinal fluid. This work is still in the early stages of biomarker development and has been accompanied by both losses and victories. Here, α-synuclein in cerebrospinal fluid as a diagnostic marker for Parkinson's disease is reviewed, including measures of total α-synuclein, oligomeric and phosphorylated α-synuclein, and seeding activity of α-synuclein.
Collapse
Affiliation(s)
- Lorraine V Kalia
- Division of Neurology, Department of Medicine and Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto; Morton and Gloria Shulman Movement Disorders Clinic and the Edmond J. Safra Program in Parkinson's Disease, Division of Neurology, Department of Medicine and Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
30
|
Mollenhauer B, Bowman FD, Drake D, Duong J, Blennow K, El-Agnaf O, Shaw LM, Masucci J, Taylor P, Umek RM, Dunty JM, Smith CL, Stoops E, Vanderstichele H, Schmid AW, Moniatte M, Zhang J, Kruse N, Lashuel HA, Teunissen C, Schubert T, Dave KD, Hutten SJ, Zetterberg H. Antibody-based methods for the measurement of α-synuclein concentration in human cerebrospinal fluid - method comparison and round robin study. J Neurochem 2018; 149:126-138. [PMID: 30125936 PMCID: PMC6587944 DOI: 10.1111/jnc.14569] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/06/2018] [Accepted: 08/15/2018] [Indexed: 01/12/2023]
Abstract
α‐Synuclein is the major component of Lewy bodies and a candidate biomarker for neurodegenerative diseases in which Lewy bodies are common, including Parkinson's disease and dementia with Lewy bodies. A large body of literature suggests that these disorders are characterized by reduced concentrations of α‐synuclein in cerebrospinal fluid (CSF), with overlapping concentrations compared to healthy controls and variability across studies. Several reasons can account for this variability, including technical ones, such as inter‐assay and inter‐laboratory variation (reproducibility). We compared four immunochemical methods for the quantification of α‐synuclein concentration in 50 unique CSF samples. All methods were designed to capture most of the existing α‐synuclein forms in CSF (‘total’ α‐synuclein). Each of the four methods showed high analytical precision, excellent correlation between laboratories (R2 0.83–0.99), and good correlation with each other (R2 0.64–0.93), although the slopes of the regression lines were different between the four immunoassays. The use of common reference CSF samples decreased the differences in α‐synuclein concentration between detection methods and technologies. Pilot data on an immunoprecipitation mass spectrometry (IP‐MS) method is also presented. Our results suggest that the four immunochemical methods and the IP‐MS method measure similar forms of α‐synuclein and that a common reference material would allow harmonization of results between immunoassays. ![]()
Collapse
Affiliation(s)
- Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany.,Department of Neurology, University Medical Center, Goettingen, Germany
| | - Frederick DuBois Bowman
- Department of Biostatistics, Columbia University, Mailman School of Public Health, New York City, New York, USA
| | - Daniel Drake
- Department of Biostatistics, Columbia University, Mailman School of Public Health, New York City, New York, USA
| | - Jimmy Duong
- Department of Biostatistics, Columbia University, Mailman School of Public Health, New York City, New York, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Omar El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), and College of Science and Engineering, HBKU, Education City, Qatar Foundation, Doha, Qatar
| | - Leslie M Shaw
- Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | - Adrian W Schmid
- Proteomics Core Facility, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Marc Moniatte
- Proteomics Core Facility, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Jing Zhang
- University of Washington, Seattle, WA, USA
| | - Niels Kruse
- Institute of Neuropathology, University Medical Center, Goettingen, Germany
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Institute of Physics of Biological Systems, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | | | | | - Kuldip D Dave
- Michael J. Fox Foundation for Parkinson's Research, New York City, New York, USA
| | - Samantha J Hutten
- Michael J. Fox Foundation for Parkinson's Research, New York City, New York, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Molecular Neuroscience, UCL Institute of Neurology, Queens Square, London, UK.,UK Dementia Research Institute at UCL, London, UK
| |
Collapse
|
31
|
Beach TG, Serrano GE, Kremer T, Canamero M, Dziadek S, Sade H, Derkinderen P, Corbillé AG, Letournel F, Munoz DG, White CL, Schneider J, Crary JF, Sue LI, Adler CH, Glass MJ, Intorcia AJ, Walker JE, Foroud T, Coffey CS, Ecklund D, Riss H, Goßmann J, König F, Kopil CM, Arnedo V, Riley L, Linder C, Dave KD, Jennings D, Seibyl J, Mollenhauer B, Chahine L. Immunohistochemical Method and Histopathology Judging for the Systemic Synuclein Sampling Study (S4). J Neuropathol Exp Neurol 2018; 77:793-802. [PMID: 30107604 PMCID: PMC6097838 DOI: 10.1093/jnen/nly056] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Immunohistochemical (IHC) α-synuclein (Asyn) pathology in peripheral biopsies may be a biomarker of Parkinson disease (PD). The multi-center Systemic Synuclein Sampling Study (S4) is evaluating IHC Asyn pathology within skin, colon and submandibular gland biopsies from 60 PD and 20 control subjects. Asyn pathology is being evaluated by a blinded panel of specially trained neuropathologists. Preliminary work assessed 2 candidate immunoperoxidase methods using a set of PD and control autopsy-derived sections from formalin-fixed, paraffin-embedded blocks of the 3 tissues. Both methods had 100% specificity; one, utilizing the 5C12 monoclonal antibody, was more sensitive in skin (67% vs 33%), and was chosen for further use in S4. Four trainee neuropathologists were trained to perform S4 histopathology readings; in subsequent testing, their scoring was compared to that of the trainer neuropathologist on both glass slides and digital images. Specificity and sensitivity were both close to 100% with all readers in all tissue types on both glass slides and digital images except for skin, where sensitivity averaged 75% with digital images and 83.5% with glass slides. Semiquantitative (0-3) density score agreement between trainees and trainer averaged 67% for glass slides and 62% for digital images.
Collapse
Affiliation(s)
- Thomas G Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona
| | - Geidy E Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona
| | - Thomas Kremer
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F Hoffman-La Roche, Ltd, Basel, Switzerland
- Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Penzberg, Germany
| | - Marta Canamero
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F Hoffman-La Roche, Ltd, Basel, Switzerland
- Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Penzberg, Germany
| | - Sebastian Dziadek
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F Hoffman-La Roche, Ltd, Basel, Switzerland
- Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Penzberg, Germany
| | - Hadassah Sade
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F Hoffman-La Roche, Ltd, Basel, Switzerland
- Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Penzberg, Germany
| | - Pascal Derkinderen
- Department of Neurology, CHU Nantes, Inserm, U1235, Nantes University, Nantes F-44035, France
- CHU Angers, Neurobiology and Neuropathology Laboratory, Angers F-49033, France
| | - Anne-Gaëlle Corbillé
- Department of Neurology, CHU Nantes, Inserm, U1235, Nantes University, Nantes F-44035, France
- CHU Angers, Neurobiology and Neuropathology Laboratory, Angers F-49033, France
| | - Franck Letournel
- Department of Neurology, CHU Nantes, Inserm, U1235, Nantes University, Nantes F-44035, France
- CHU Angers, Neurobiology and Neuropathology Laboratory, Angers F-49033, France
| | - David G Munoz
- Laboratory Medicine and Keenan Research Centre for Biomedical Research of the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| | - Charles L White
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | | | - John F Crary
- Department of Pathology, Fishberg Department of Neuroscience, Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lucia I Sue
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona
| | - Charles H Adler
- Department of Neurology, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Michael J Glass
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona
| | - Anthony J Intorcia
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona
| | - Jessica E Walker
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona
| | - Tatiana Foroud
- Indiana University School of Medicine, Indianapolis, Indiana
| | | | - Dixie Ecklund
- Department of Biostatistics, University of Iowa, Iowa City, Iowa
| | - Holly Riss
- Department of Biostatistics, University of Iowa, Iowa City, Iowa
| | | | - Fatima König
- Targos Molecular Pathology GmbH, Kassel, Germany
| | - Catherine M Kopil
- The Michael J. Fox Foundation for Parkinson’s Research, New York, New York
| | - Vanessa Arnedo
- The Michael J. Fox Foundation for Parkinson’s Research, New York, New York
| | - Lindsey Riley
- The Michael J. Fox Foundation for Parkinson’s Research, New York, New York
| | - Carly Linder
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kuldip D Dave
- The Michael J. Fox Foundation for Parkinson’s Research, New York, New York
| | | | - John Seibyl
- Institute for Neurodegenerative Disorders, New Haven, Connecticut
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel and University Medical Center Goettingen, Goettingen, Germany
| | - Lana Chahine
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
32
|
Irwin DJ, Hurtig HI. The Contribution of Tau, Amyloid-Beta and Alpha-Synuclein Pathology to Dementia in Lewy Body Disorders. JOURNAL OF ALZHEIMER'S DISEASE & PARKINSONISM 2018; 8:444. [PMID: 30473927 PMCID: PMC6248323 DOI: 10.4172/2161-0460.1000444] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Parkinson's Disease (PD) and the closely related Dementia with Lewy Bodies (DLB) are due to the accumulation of pathogenic alpha-synuclein protein in brain cells manifest by heterogeneous motor and non-motor symptoms, including cognitive impairment and dementia. The majority of patients with Parkinson's Disease develop Dementia (PDD) in late stages of the disease and have widespread neocortical distribution of alpha-synuclein pathology at autopsy, compared with PD without dementia, in which neocortical synuclein pathology is less prevalent. These three entities PD, DLB and PDD comprise a clinical spectrum, collectively known as Lewy Body Disorders (LBD). Recent investigations into the neuropathological basis of LBD have demonstrated that while synuclein pathology is the defining feature of these disorders, it is often accompanied by other age-related neurodegenerative pathologies. In particular, amyloid plaque and tau tangle pathology characteristic of Alzheimer's Disease (AD) (~50% of all LBD patients have sufficient pathology at autopsy for a secondary neuropathologic diagnosis of AD), appear to contribute to cognitive impairment in LBD, and the combination is associated with a shorter interval between onset of motor symptoms and development of dementia and a shorter life span. Further, the co-occurrence of neocortical alpha-synuclein, tau and amyloid pathologies found at end-stage disease suggests a potential synergistic interaction of these individual pathologies in humans during life, mirroring experimental observations in animal and cell model systems that show how pathogenic species of synuclein fibrils can promote trans-synaptic spread of both tauopathy and synucleinopathy with strain-like properties. Newer post-mortem studies using digital methods to measure pathologic burden have highlighted distinct neocortical patterns of areas with relative higher density of tau pathology in LBD compared to AD that support these model data. The emerging field of cerebrospinal fluid and molecular imaging biomarkers of synuclein, amyloid and tau pathologies in LBD is contributing to a greater understanding of how the different pathologies evolve and interact to produce clinical heterogeneity in LBD. Future work to elucidate biologically meaningful clinical subgroups of synucleinopathy and its co-pathology must focus on the full clinicopathological spectrum of LBD and use validated biomarkers, when available, to design clinical trials based on the precise selection of homogeneous patient subgroups to maximize statistical power for detecting the impact of treatment.
Collapse
Affiliation(s)
- David J. Irwin
- University of Pennsylvania Perelman School of Medicine, Department of Neurology Philadelphia PA, USA
| | - Howard I. Hurtig
- University of Pennsylvania Perelman School of Medicine, Department of Neurology Philadelphia PA, USA
| |
Collapse
|
33
|
Dolatshahi M, Pourmirbabaei S, Kamalian A, Ashraf-Ganjouei A, Yaseri M, Aarabi MH. Longitudinal Alterations of Alpha-Synuclein, Amyloid Beta, Total, and Phosphorylated Tau in Cerebrospinal Fluid and Correlations Between Their Changes in Parkinson's Disease. Front Neurol 2018; 9:560. [PMID: 30050494 PMCID: PMC6052894 DOI: 10.3389/fneur.2018.00560] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/21/2018] [Indexed: 11/13/2022] Open
Abstract
Background: Parkinson's disease (PD) is characterized by proteinopathies and these proteinopathies seem to interact synergistically and lead to protein aggregations and changes in protein cerebrospinal fluid (CSF) levels. In this study, we aimed to explore the longitudinal changes of CSF a lpha-synuclein (α-syn), total tau (t-tau), phosphorylated tau (p-tau), and beta-amyloid (Aβ1-42) and their relationships with each other and with baseline clinical entities like REM sleep behavior disorder (RBD), cognitive impairment, motor symptoms, and olfaction dysfunction. Method: One hundred and twelve non-demented PD patients and 110 controls were recruited from Parkinson's Progression Markers Initiative (PPMI).We used a linear mixed model within groups to assess longitudinal protein changes over 6 and 12 months and a random regression coefficient within the linear mixed model to investigate the correlation between proteins and their baseline clinical characteristics. Results: P-tau was lower in PDs only at baseline, but during a year, p-tau increased more rapidly in PDs than controls. Aβ1-42 was not significantly different between groups at any separate timepoint; however, when assessed longitudinally, Aβ1-42 showed significant changes in both groups. Conversely, t-tau and α-syn differed significantly between groups, but their longitudinal changes were not significant in either of the groups. Moreover, all proteins' baseline levels, except p-tau, could determine estimated longitudinal tau changes. Baseline RBDSQ scores but not UPDRS III, MoCA, or UPSIT scores were predictive of longitudinal increase in α-syn levels. Conclusion: Longitudinal changes in levels of CSF proteins are related to each other and could help researchers further understand PD pathology. In addition, RBD seems to be a potential prognostic factor for PD progression. However, in order to reach a consensus, longer follow-up times are required.
Collapse
Affiliation(s)
- Mahsa Dolatshahi
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Aida Kamalian
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mehdi Yaseri
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad H Aarabi
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Lewczuk P, Riederer P, O’Bryant SE, Verbeek MM, Dubois B, Visser PJ, Jellinger KA, Engelborghs S, Ramirez A, Parnetti L, Jack CR, Teunissen CE, Hampel H, Lleó A, Jessen F, Glodzik L, de Leon MJ, Fagan AM, Molinuevo JL, Jansen WJ, Winblad B, Shaw LM, Andreasson U, Otto M, Mollenhauer B, Wiltfang J, Turner MR, Zerr I, Handels R, Thompson AG, Johansson G, Ermann N, Trojanowski JQ, Karaca I, Wagner H, Oeckl P, van Waalwijk van Doorn L, Bjerke M, Kapogiannis D, Kuiperij HB, Farotti L, Li Y, Gordon BA, Epelbaum S, Vos SJB, Klijn CJM, Van Nostrand WE, Minguillon C, Schmitz M, Gallo C, Mato AL, Thibaut F, Lista S, Alcolea D, Zetterberg H, Blennow K, Kornhuber J, Riederer P, Gallo C, Kapogiannis D, Mato AL, Thibaut F. Cerebrospinal fluid and blood biomarkers for neurodegenerative dementias: An update of the Consensus of the Task Force on Biological Markers in Psychiatry of the World Federation of Societies of Biological Psychiatry. World J Biol Psychiatry 2018; 19:244-328. [PMID: 29076399 PMCID: PMC5916324 DOI: 10.1080/15622975.2017.1375556] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the 12 years since the publication of the first Consensus Paper of the WFSBP on biomarkers of neurodegenerative dementias, enormous advancement has taken place in the field, and the Task Force takes now the opportunity to extend and update the original paper. New concepts of Alzheimer's disease (AD) and the conceptual interactions between AD and dementia due to AD were developed, resulting in two sets for diagnostic/research criteria. Procedures for pre-analytical sample handling, biobanking, analyses and post-analytical interpretation of the results were intensively studied and optimised. A global quality control project was introduced to evaluate and monitor the inter-centre variability in measurements with the goal of harmonisation of results. Contexts of use and how to approach candidate biomarkers in biological specimens other than cerebrospinal fluid (CSF), e.g. blood, were precisely defined. Important development was achieved in neuroimaging techniques, including studies comparing amyloid-β positron emission tomography results to fluid-based modalities. Similarly, development in research laboratory technologies, such as ultra-sensitive methods, raises our hopes to further improve analytical and diagnostic accuracy of classic and novel candidate biomarkers. Synergistically, advancement in clinical trials of anti-dementia therapies energises and motivates the efforts to find and optimise the most reliable early diagnostic modalities. Finally, the first studies were published addressing the potential of cost-effectiveness of the biomarkers-based diagnosis of neurodegenerative disorders.
Collapse
Affiliation(s)
- Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, and Department of Biochemical Diagnostics, University Hospital of Białystok, Białystok, Poland
| | - Peter Riederer
- Center of Mental Health, Clinic and Policlinic of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
| | - Sid E. O’Bryant
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Marcel M. Verbeek
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer center, Nijmegen, The Netherlands
| | - Bruno Dubois
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Salpêtrièrie Hospital, INSERM UMR-S 975 (ICM), Paris 6 University, Paris, France
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
- Department of Neurology, Alzheimer Centre, Amsterdam Neuroscience VU University Medical Centre, Amsterdam, The Netherlands
| | | | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), University of Antwerp, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Alfredo Ramirez
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Lucilla Parnetti
- Section of Neurology, Center for Memory Disturbances, Lab of Clinical Neurochemistry, University of Perugia, Perugia, Italy
| | | | - Charlotte E. Teunissen
- Neurochemistry Lab and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Harald Hampel
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Alberto Lleó
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Spain
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Germany
| | - Lidia Glodzik
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - Mony J. de Leon
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - Anne M. Fagan
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - José Luis Molinuevo
- Barcelonabeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
- Alzheimer’s Disease and Other Cognitive Disorders Unit, Hospital Clínic, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Willemijn J. Jansen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Bengt Winblad
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ulf Andreasson
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel and University Medical Center Göttingen, Department of Neurology, Göttingen, Germany
| | - Jens Wiltfang
- Department of Psychiatry & Psychotherapy, University of Göttingen, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- iBiMED, Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| | - Martin R. Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Inga Zerr
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Clinical Dementia Centre, Department of Neurology, University Medical School, Göttingen, Germany
| | - Ron Handels
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | | | - Gunilla Johansson
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | - Natalia Ermann
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - John Q. Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilker Karaca
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Holger Wagner
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Patrick Oeckl
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Linda van Waalwijk van Doorn
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer center, Nijmegen, The Netherlands
| | - Maria Bjerke
- Reference Center for Biological Markers of Dementia (BIODEM), University of Antwerp, Antwerp, Belgium
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, National Institute on Aging/National Institutes of Health (NIA/NIH), Baltimore, MD, USA
| | - H. Bea Kuiperij
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer center, Nijmegen, The Netherlands
| | - Lucia Farotti
- Section of Neurology, Center for Memory Disturbances, Lab of Clinical Neurochemistry, University of Perugia, Perugia, Italy
| | - Yi Li
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - Brian A. Gordon
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Stéphane Epelbaum
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Salpêtrièrie Hospital, INSERM UMR-S 975 (ICM), Paris 6 University, Paris, France
| | - Stephanie J. B. Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Catharina J. M. Klijn
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | | | - Carolina Minguillon
- Barcelonabeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Matthias Schmitz
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Clinical Dementia Centre, Department of Neurology, University Medical School, Göttingen, Germany
| | - Carla Gallo
- Departamento de Ciencias Celulares y Moleculares/Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Andrea Lopez Mato
- Chair of Psychoneuroimmunoendocrinology, Maimonides University, Buenos Aires, Argentina
| | - Florence Thibaut
- Department of Psychiatry, University Hospital Cochin-Site Tarnier 89 rue d’Assas, INSERM 894, Faculty of Medicine Paris Descartes, Paris, France
| | - Simone Lista
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Daniel Alcolea
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Spain
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Irwin DJ, Xie SX, Coughlin D, Nevler N, Akhtar RS, McMillan CT, Lee EB, Wolk DA, Weintraub D, Chen-Plotkin A, Duda JE, Spindler M, Siderowf A, Hurtig HI, Shaw LM, Grossman M, Trojanowski JQ. CSF tau and β-amyloid predict cerebral synucleinopathy in autopsied Lewy body disorders. Neurology 2018; 90:e1038-e1046. [PMID: 29467305 PMCID: PMC5874449 DOI: 10.1212/wnl.0000000000005166] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/15/2017] [Indexed: 01/29/2023] Open
Abstract
OBJECTIVE To test the association of antemortem CSF biomarkers with postmortem pathology in Lewy body disorders (LBD). METHODS Patients with autopsy-confirmed LBD (n = 24) and autopsy-confirmed Alzheimer disease (AD) (n = 23) and cognitively normal (n = 36) controls were studied. In LBD, neuropathologic criteria defined Lewy body α-synuclein (SYN) stages with medium/high AD copathology (SYN + AD = 10) and low/no AD copathology (SYN - AD = 14). Ordinal pathology scores for tau, β-amyloid (Aβ), and SYN pathology were averaged across 7 cortical regions to obtain a global cerebral score for each pathology. CSF total tau (t-tau), phosphorylated tau at threonine181, and Aβ1-42 levels were compared between LBD and control groups and correlated with global cerebral pathology scores in LBD with linear regression. Diagnostic accuracy for postmortem categorization of LBD into SYN + AD vs SYN - AD or neocortical vs brainstem/limbic SYN stage was tested with receiver operating curves. RESULTS SYN + AD had higher CSF t-tau (mean difference 27.0 ± 8.6 pg/mL) and lower Aβ1-42 (mean difference -84.0 ± 22.9 g/mL) compared to SYN - AD (p < 0.01, both). Increasing global cerebral tau and plaque scores were associated with higher CSF t-tau (R2 = 0.15-0.16, p < 0.05, both) and lower Aβ1-42 (R2 = 0.43-0.49, p < 0.001, both), while increasing cerebral SYN scores were associated with lower CSF Aβ1-42 (R2 = 0.31, p < 0.001) and higher CSF t-tau/Aβ1-42 ratio (R2 = 0.27, p = 0.01). CSF t-tau/Aβ1-42 ratio had 100% specificity and 90% sensitivity for SYN + AD, and CSF Aβ1-42 had 77% specificity and 82% sensitivity for neocortical SYN stage. CONCLUSIONS Higher antemortem CSF t-tau/Aβ1-42 and lower Aβ1-42 levels are predictive of increasing cerebral AD and SYN pathology. These biomarkers may identify patients with LBD vulnerable to cortical SYN pathology who may benefit from both SYN and AD-targeted disease-modifying therapies.
Collapse
Affiliation(s)
- David J Irwin
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA.
| | - Sharon X Xie
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - David Coughlin
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Naomi Nevler
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Rizwan S Akhtar
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Corey T McMillan
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Edward B Lee
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - David A Wolk
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Daniel Weintraub
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Alice Chen-Plotkin
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - John E Duda
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Meredith Spindler
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Andrew Siderowf
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Howard I Hurtig
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Leslie M Shaw
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Murray Grossman
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - John Q Trojanowski
- From Penn Frontotemporal Degeneration Center (D.J.I., N.N., C.T.M., M.G.), Alzheimer's Disease Core Center (D.A.W., J.Q.T.), Department of Neurology (D.J.I., D.C., R.S.A., C.T.M., D.A.W., D.W., A.C.-P., M.S., A.S., H.I.H., M.G.), Penn Morris K. Udall Parkinson's Disease Research Center of Excellence (D.J.I., R.S.A., C.T.M., D.W., A.C.-P., J.E.D., M.G., J.Q.T.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), Translational Neuropathology Research Laboratory (E.B.L.), Department of Pathology and Laboratory Medicine (E.B.L., L.M.S., J.Q.T.), and Department of Biostatistics and Epidemiology Perelman School of Medicine (S.X.X.), University of Pennsylvania; and Parkinson's Disease Research, Education and Clinical Center (D.W., J.E.D.), Michael J. Crescenz VA Medical Center, Philadelphia, PA
| |
Collapse
|
36
|
Jellinger KA, Korczyn AD. Are dementia with Lewy bodies and Parkinson's disease dementia the same disease? BMC Med 2018; 16:34. [PMID: 29510692 PMCID: PMC5840831 DOI: 10.1186/s12916-018-1016-8] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/30/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD), which share many clinical, neurochemical, and morphological features, have been incorporated into DSM-5 as two separate entities of major neurocognitive disorders with Lewy bodies. Despite clinical overlap, their diagnosis is based on an arbitrary distinction concerning the time of onset of motor and cognitive symptoms, namely as early cognitive impairment in DLB and later onset following that of motor symptoms in PDD. Their morphological hallmarks - cortical and subcortical α-synuclein/Lewy body plus β-amyloid and tau pathologies - are similar, but clinical differences at onset suggest some dissimilar profiles. Based on recent publications, including the fourth consensus report of the DLB Consortium, a critical overview is provided herein. DISCUSSION The clinical constellations of DLB and PDD include cognitive impairment, parkinsonism, visual hallucinations, and fluctuating attention. Intravitam PET and postmortem studies have revealed a more pronounced cortical atrophy, elevated cortical and limbic Lewy body pathologies, higher Aβ and tau loads in cortex and striatum in DLB compared to PDD, and earlier cognitive defects in DLB. Conversely, multitracer PET studies have shown no differences in cortical and striatal cholinergic and dopaminergic deficits. Clinical management of both DLB and PDD includes cholinesterase inhibitors and other pharmacologic and non-drug strategies, yet with only mild symptomatic effects. Currently, no disease-modifying therapies are available. CONCLUSION DLB and PDD are important dementia syndromes that overlap in many clinical features, genetics, neuropathology, and management. They are currently considered as subtypes of an α-synuclein-associated disease spectrum (Lewy body diseases), from incidental Lewy body disease and non-demented Parkinson's disease to PDD, DLB, and DLB with Alzheimer's disease at the most severe end. Cognitive impairment in these disorders is induced not only by α-synuclein-related neurodegeneration but by multiple regional pathological scores. Both DLB and PDD show heterogeneous pathology and neurochemistry, suggesting that they share important common underlying molecular pathogenesis with Alzheimer's disease and other proteinopathies. While we prefer to view DLB and PDD as extremes on a continuum, there remains a pressing need to more clearly differentiate these syndromes and to understand the synucleinopathy processes leading to either one.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, A-1150, Vienna, Austria.
| | - Amos D Korczyn
- Tel-Aviv University, Sackler Faculty of Medicine, Ramat Aviv, Israel
| |
Collapse
|
37
|
Jellinger KA. Dementia with Lewy bodies and Parkinson's disease-dementia: current concepts and controversies. J Neural Transm (Vienna) 2017; 125:615-650. [PMID: 29222591 DOI: 10.1007/s00702-017-1821-9] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 11/28/2017] [Indexed: 12/15/2022]
Abstract
Dementia with Lewy bodies (DLB) and Parkinson's disease-dementia (PDD), although sharing many clinical, neurochemical and morphological features, according to DSM-5, are two entities of major neurocognitive disorders with Lewy bodies of unknown etiology. Despite considerable clinical overlap, their diagnosis is based on an arbitrary distinction between the time of onset of motor and cognitive symptoms: dementia often preceding parkinsonism in DLB and onset of cognitive impairment after onset of motor symptoms in PDD. Both are characterized morphologically by widespread cortical and subcortical α-synuclein/Lewy body plus β-amyloid and tau pathologies. Based on recent publications, including the fourth consensus report of the DLB Consortium, a critical overview is given. The clinical features of DLB and PDD include cognitive impairment, parkinsonism, visual hallucinations, and fluctuating attention. Intravitam PET and post-mortem studies revealed more pronounced cortical atrophy, elevated cortical and limbic Lewy pathologies (with APOE ε4), apart from higher prevalence of Alzheimer pathology in DLB than PDD. These changes may account for earlier onset and greater severity of cognitive defects in DLB, while multitracer PET studies showed no differences in cholinergic and dopaminergic deficits. DLB and PDD sharing genetic, neurochemical, and morphologic factors are likely to represent two subtypes of an α-synuclein-associated disease spectrum (Lewy body diseases), beginning with incidental Lewy body disease-PD-nondemented-PDD-DLB (no parkinsonism)-DLB with Alzheimer's disease (DLB-AD) at the most severe end, although DLB does not begin with PD/PDD and does not always progress to DLB-AD, while others consider them as the same disease. Both DLB and PDD show heterogeneous pathology and neurochemistry, suggesting that they share important common underlying molecular pathogenesis with AD and other proteinopathies. Cognitive impairment is not only induced by α-synuclein-caused neurodegeneration but by multiple regional pathological scores. Recent animal models and human post-mortem studies have provided important insights into the pathophysiology of DLB/PDD showing some differences, e.g., different spreading patterns of α-synuclein pathology, but the basic pathogenic mechanisms leading to the heterogeneity between both disorders deserve further elucidation. In view of the controversies about the nosology and pathogenesis of both syndromes, there remains a pressing need to differentiate them more clearly and to understand the processes leading these synucleinopathies to cause one disorder or the other. Clinical management of both disorders includes cholinesterase inhibitors, other pharmacologic and nonpharmacologic strategies, but these have only a mild symptomatic effect. Currently, no disease-modifying therapies are available.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
38
|
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
|
39
|
Jellinger KA. Potential clinical utility of multiple system atrophy biomarkers. Expert Rev Neurother 2017; 17:1189-1208. [DOI: 10.1080/14737175.2017.1392239] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
40
|
Mollenhauer B, Batrla R, El-Agnaf O, Galasko DR, Lashuel HA, Merchant KM, Shaw LM, Selkoe DJ, Umek R, Vanderstichele H, Zetterberg H, Zhang J, Caspell-Garcia C, Coffey C, Hutten SJ, Frasier M, Taylor P. A user's guide for α-synuclein biomarker studies in biological fluids: Perianalytical considerations. Mov Disord 2017; 32:1117-1130. [PMID: 28734051 PMCID: PMC5638072 DOI: 10.1002/mds.27090] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/30/2017] [Accepted: 06/02/2017] [Indexed: 01/13/2023] Open
Abstract
Parkinson's disease biomarkers are needed to increase diagnostic accuracy, to objectively monitor disease progression and to assess therapeutic efficacy as well as target engagement when evaluating novel drug and therapeutic strategies. This article summarizes perianalytical considerations for biomarker studies (based on immunoassays) in Parkinson's disease, with emphasis on quantifying total α‐synuclein protein in biological fluids. Current knowledge and pitfalls are discussed, and selected perianalytical variables are presented systematically, including different temperature of sample collection and types of collection tubes, gradient sampling, the addition of detergent, aliquot volume, the freezing time, and the different thawing methods. We also discuss analytical confounders. We identify gaps in the knowledge and delineate specific areas that require further investigation, such as the need to identify posttranslational modifications of α‐synuclein and antibody‐independent reference methods for quantification, as well as the analysis of potential confounders, such as comorbidities, medication, and phenotypes of Parkinson's disease in larger cohorts. This review could be used as a guideline for future Parkinson's disease biomarker studies and will require regular updating as more information arises in this growing field, including new technical developments as they become available. In addition to reviewing best practices, we also identify the current technical limitations and gaps in the knowledge that should be addressed to enable accurate and quantitative assessment of α‐synuclein levels in the clinical setting. © 2017 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany.,Department of Neurology, University Medical Center, Göttingen, Germany
| | - Richard Batrla
- Roche Diagnostics International Ltd, Rotkreuz, Switzerland
| | - Omar El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), and College of Science and Engineering, HBKU, Education City, Qatar Foundation, Doha, Qatar
| | | | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Faculty of Life Science, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | | | - Lesley M Shaw
- Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dennis J Selkoe
- Center for Neurodegenerative Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert Umek
- MesoScale Discovery, Gaithersburg, Maryland, USA
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK; UK Dementia Research Institute, London, UK
| | - Jing Zhang
- University of Washington, Seattle, Washington, USA
| | - Chelsea Caspell-Garcia
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | - Chris Coffey
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | - Samantha J Hutten
- Michael J. Fox Foundation for Parkinson's Research, New York, New York, USA
| | - Mark Frasier
- Michael J. Fox Foundation for Parkinson's Research, New York, New York, USA
| | | | | |
Collapse
|
41
|
Abstract
Measures of the severity of cognitive impairment or parkinsonism are the usual endpoints in clinical trials for Alzheimer’s disease (AD) and Parkinson’s disease (PD), but are critically hampered by their lack of disease sensitivity and specificity. Due to the high failure rate of clinical trials, the rate of regulatory approval for efficacious new drugs has stagnated in the past few decades, with the gap between basic science discovery and clinical application metaphorically termed the “Valley of Death”. While the causes for this are probably multiple and complex, the usage of biomarkers as surrogate endpoints, particularly when they are molecularly-specific for the disease, has achieved some success in cancer trials, and it is likely that neurodegenerative disease trials would benefit from the same approach. As dementia and parkinsonism are not disease-specific clinical syndromes, both AD and PD trials have been flawed by reliance on clinical diagnosis and clinical endpoints. Clinical improvement has been a requirement for regulatory approval, but molecularly-specific biomarkers should improve both diagnostic accuracy and tracking of disease progression, allowing quicker screening of drug candidates. However, even when a molecularly-specific biomarker is found, such as amyloid imaging for AD, it may not reflect the entire extant molecular disease repertoire and may not serve equally well in the different roles of preclinical detection, diagnostic confirmation and surrogate endpoint, necessitating the usage of two, three or more biomarkers, deployed in series or in parallel.
Collapse
|
42
|
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
|
43
|
Tomlinson JJ, Shutinoski B, Dong L, Meng F, Elleithy D, Lengacher NA, Nguyen AP, Cron GO, Jiang Q, Roberson ED, Nussbaum RL, Majbour NK, El-Agnaf OM, Bennett SA, Lagace DC, Woulfe JM, Sad S, Brown EG, Schlossmacher MG. Holocranohistochemistry enables the visualization of α-synuclein expression in the murine olfactory system and discovery of its systemic anti-microbial effects. J Neural Transm (Vienna) 2017; 124:721-738. [PMID: 28477284 PMCID: PMC5446848 DOI: 10.1007/s00702-017-1726-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 04/18/2017] [Indexed: 01/19/2023]
Abstract
Braak and Del Tredici have proposed that typical Parkinson disease (PD) has its origins in the olfactory bulb and gastrointestinal tract. However, the role of the olfactory system has insufficiently been explored in the pathogeneses of PD and Alzheimer disease (AD) in laboratory models. Here, we demonstrate applications of a new method to process mouse heads for microscopy by sectioning, mounting, and staining whole skulls (‘holocranohistochemistry’). This technique permits the visualization of the olfactory system from the nasal cavity to mitral cells and dopamine-producing interneurons of glomeruli in the olfactory bulb. We applied this method to two specific goals: first, to visualize PD- and AD-linked gene expression in the olfactory system, where we detected abundant, endogenous α-synuclein and tau expression in the olfactory epithelium. Furthermore, we observed amyloid-β plaques and proteinase-K-resistant α-synuclein species, respectively, in cranial nerve-I of APP- and human SNCA-over-expressing mice. The second application of the technique was to the modeling of gene–environment interactions in the nasal cavity of mice. We tracked the infection of a neurotropic respiratory-enteric-orphan virus from the nose pad into cranial nerves-I (and -V) and monitored the ensuing brain infection. Given its abundance in the olfactory epithelia, we questioned whether α-synuclein played a role in innate host defenses to modify the outcome of infections. Indeed, Snca-null mice were more likely to succumb to viral encephalitis versus their wild-type littermates. Moreover, using a bacterial sepsis model, Snca-null mice were less able to control infection after intravenous inoculation with Salmonella typhimurium. Together, holocranohistochemistry enabled new discoveries related to α-synuclein expression and its function in mice. Future studies will address: the role of Mapt and mutant SNCA alleles in infection paradigms; the contribution of xenobiotics in the initiation of idiopathic PD; and the safety to the host when systemically targeting α-synuclein by immunotherapy.
Collapse
Affiliation(s)
- Julianna J Tomlinson
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada. .,University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada. .,University of Ottawa, 451 Smyth Road, RGH #1464, Ottawa, ON, K1H 8M5, Canada.
| | - Bojan Shutinoski
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Li Dong
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Fanyi Meng
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Dina Elleithy
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | | | - Angela P Nguyen
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Greg O Cron
- Department of Medical Imaging, The Ottawa Hospital, Ottawa, ON, Canada.,Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, Department of Radiology, University of Ottawa, Ottawa, ON, Canada
| | - Qiubo Jiang
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Erik D Roberson
- Department of Neurology, University of Alabama, Birmingham, AL, USA
| | - Robert L Nussbaum
- Division of Medical Genetics, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Nour K Majbour
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Omar M El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Steffany A Bennett
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Diane C Lagace
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - John M Woulfe
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.,Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Subash Sad
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Earl G Brown
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michael G Schlossmacher
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada. .,University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada. .,Division of Neurology, Department of Medicine, Faculty of Medicine, The Ottawa Hospital, Ottawa, ON, Canada. .,University of Ottawa, 451 Smyth Road, RGH #1464, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
44
|
Abstract
Dementia is a frequent problem encountered in advanced stages of Parkinson disease (PD). In recent years, research has focused on the pre-dementia stages of cognitive impairment in PD, including mild cognitive impairment (MCI). Several longitudinal studies have shown that MCI is a harbinger of dementia in PD, although the course is variable, and stabilization of cognition - or even reversal to normal cognition - is not uncommon. In addition to limbic and cortical spread of Lewy pathology, several other mechanisms are likely to contribute to cognitive decline in PD, and a variety of biomarker studies, some using novel structural and functional imaging techniques, have documented in vivo brain changes associated with cognitive impairment. The evidence consistently suggests that low cerebrospinal fluid levels of amyloid-β42, a marker of comorbid Alzheimer disease (AD), predict future cognitive decline and dementia in PD. Emerging genetic evidence indicates that in addition to the APOE*ε4 allele (an established risk factor for AD), GBA mutations and SCNA mutations and triplications are associated with cognitive decline in PD, whereas the findings are mixed for MAPT polymorphisms. Cognitive enhancing medications have some effect in PD dementia, but no convincing evidence that progression from MCI to dementia can be delayed or prevented is available, although cognitive training has shown promising results.
Collapse
|
45
|
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
|
46
|
Chen J, Schwarz E. Opportunities and Challenges of Multiplex Assays: A Machine Learning Perspective. Methods Mol Biol 2017; 1546:115-122. [PMID: 27896760 DOI: 10.1007/978-1-4939-6730-8_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Multiplex assays that allow the simultaneous measurement of multiple analytes in small sample quantities have developed into a widely used technology. Their implementation spans across multiple assay systems and can provide readouts of similar quality as the respective single-plex measures, albeit at far higher throughput. Multiplex assay systems are therefore an important element for biomarker discovery and development strategies but analysis of the derived data can face substantial challenges that may limit the possibility of identifying meaningful biological markers. This chapter gives an overview of opportunities and challenges of multiplexed biomarker analysis, in particular from the perspective of machine learning aimed at identification of predictive biological signatures.
Collapse
Affiliation(s)
- Junfang Chen
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J 5, Mannheim, 68159, Germany
| | - Emanuel Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J 5, Mannheim, 68159, Germany.
| |
Collapse
|
47
|
Schulz JB, Hausmann L, Hardy J. 199 years of Parkinson disease - what have we learned and what is the path to the future? J Neurochem 2016; 139 Suppl 1:3-7. [PMID: 27581372 DOI: 10.1111/jnc.13733] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 06/29/2016] [Indexed: 12/21/2022]
Abstract
In 1817, 199 years ago, James Parkinson described for the first time in 'An Essay on the Shaking Palsy' the symptoms of the disease that was later named Parkinson Disease. The current special issue of the Journal of Neurochemistry is dedicated to the discoveries and advances that have been made since, leading to a better understanding of this neurodegenerative disease and of potential treatment options. Reputed researchers cover various aspects from neuroanatomical basics; genetic and molecular risk factors such as LRRK2; the available cell and animal models that mimic crucial features of the pathophysiology; to clinical aspects and treatments, including deep brain stimulation. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Jörg B Schulz
- Department of Neurology, University Hospital, RWTH Aachen University, Aachen, Germany. .,JARA-BRAIN Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany.
| | - Laura Hausmann
- Department of Neurology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - John Hardy
- Reta Lila Weston Research Laboratories, UCL Institute of Neurology, Queen Square, London, WC1N3BG, UK
| |
Collapse
|
48
|
van Steenoven I, Aarsland D, Weintraub D, Londos E, Blanc F, van der Flier WM, Teunissen CE, Mollenhauer B, Fladby T, Kramberger MG, Bonanni L, Lemstra AW. Cerebrospinal Fluid Alzheimer's Disease Biomarkers Across the Spectrum of Lewy Body Diseases: Results from a Large Multicenter Cohort. J Alzheimers Dis 2016; 54:287-95. [PMID: 27567832 PMCID: PMC5535729 DOI: 10.3233/jad-160322] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Concomitant Alzheimer's disease (AD) pathology is observed in Lewy body diseases (LBD), but the clinical impact is unknown. Only a few biomarker studies in LBD exist and have included small cohorts from single centers. OBJECTIVE We aimed to evaluate the prevalence of abnormal cerebrospinal fluid (CSF) AD biomarkers across the spectrum of LBD in a large multicenter cohort and to assess whether an AD biomarker profile was associated with demographic and clinical differences in dementia with Lewy bodies (DLB). METHODS We included 375 DLB patients, 164 Parkinson's disease (PD) patients without dementia, and 55 PD patients with dementia (PDD) from 10 centers. CSF amyloid-beta42 (Aβ42), total tau (t-tau), and phosphorylated tau (p-tau) values were dichotomized as abnormal or normal according to locally available cut-off values. A CSF AD profile was defined as abnormal Aβ42 combined with abnormal t-tau and/or p-tau. RESULTS A substantial proportion of DLB patients had abnormal values for CSF Aβ42, t-tau, and p-tau, while abnormal values were uncommon in PD without dementia. Patients with PDD had values in between. A CSF AD profile was observed in 25% of DLB patients, compared with only 9% of PDD and 3% of PD without dementia. Within DLB, patients with a CSF AD profile were older, more often female, performed worse on the Mini-Mental State Examination, and had shorter disease duration compared with patients with normal CSF. CONCLUSION A CSF AD profile is more common in DLB compared with PDD and PD, and is associated with more severe cognitive impairment in DLB.
Collapse
Affiliation(s)
- Inger van Steenoven
- Department of Neurology & Alzheimer Centre, VU University Medical Center, Amsterdam, The Netherlands
| | - Dag Aarsland
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institute, Stockholm, Sweden
- Center for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Daniel Weintraub
- Departments of Psychiatry and Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Elisabet Londos
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Frédéric Blanc
- Neuropsychology Unit and Geriatric Day Hospital (Strasbourg Resource and Research Memory Centre, CMRR), University Hospital of Strasbourg and ICube Laboratory, FMTS, University of Strasbourg and CNRS, Strasbourg, France
| | - Wiesje M. van der Flier
- Department of Neurology & Alzheimer Centre and Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry Lab and Biobank, Department of Clinical Chemistry, VU University Medical Center Amsterdam, The Netherlands
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel and University Medical Center, Department of Neurosurgery and Institute of Neuropathology, Göttingen, Germany
| | - Tormod Fladby
- Department of Neurology, Akershus University Hospital and Faculty of Medicine, University of Oslo, Norway
| | | | - Laura Bonanni
- Department of Neuroscience and Imaging and Clinical Science, and Aging Research Centre, G. d’Annunzio University, Chieti, Italy
| | - Afina W. Lemstra
- Department of Neurology & Alzheimer Centre, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
49
|
Abdelnour C, van Steenoven I, Londos E, Blanc F, Auestad B, Kramberger MG, Zetterberg H, Mollenhauer B, Boada M, Aarsland D. Alzheimer's disease cerebrospinal fluid biomarkers predict cognitive decline in lewy body dementia. Mov Disord 2016; 31:1203-8. [DOI: 10.1002/mds.26668] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/11/2016] [Accepted: 04/13/2016] [Indexed: 01/13/2023] Open
Affiliation(s)
- Carla Abdelnour
- Fundació ACE, Alzheimer Research Center and Memory ClinicInstitut Català de Neurociències AplicadesBarcelona Spain
| | - Inger van Steenoven
- Alzheimer Center, Department of NeurologyVU Medical CenterAmsterdam The Netherlands
| | - Elisabet Londos
- Clinical Memory Research Unit, Department of Clinical SciencesLund UniversityMalmö Sweden
| | - Frédéric Blanc
- Neuropsychology Unit and Geriatric Day Hospital (Strasbourg Resource and Research Memory Center, CMRR), University Hospital of Strasbourg and ICube laboratory, FMTSUniversity of Strasbourg and CNRSStrasbourg France
| | - Bjørn Auestad
- Research DepartmentStavanger University HospitalStavanger Norway
- Department of Mathematics and Natural SciencesUniversity of StavangerStavanger Norway
| | | | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistrythe Sahlgrenska Academy at the University of GothenburgMölndal Sweden
- Department of Molecular NeuroscienceUCL Institute of NeurologyLondon United Kingdom
| | - Brit Mollenhauer
- Paracelsus‐Elena‐Klinik, Kassel and University Medical Center, Department of Neurosurgery and Institute of NeuropathologyGöttingen Germany
| | - Mercè Boada
- Fundació ACE, Alzheimer Research Center and Memory ClinicInstitut Català de Neurociències AplicadesBarcelona Spain
| | - Dag Aarsland
- Research DepartmentStavanger University HospitalStavanger Norway
- Karolinska Institutet, Department of Neurobiology, Care sciences and Society (NVS)Center for Alzheimer Research Division for NeurogeriatricsStockholm Sweden
| | | |
Collapse
|
50
|
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
|