1
|
Sharma P, Giri A, Tripathi PN. Emerging Trends: Neurofilament Biomarkers in Precision Neurology. Neurochem Res 2024; 49:3208-3225. [PMID: 39347854 DOI: 10.1007/s11064-024-04244-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 10/01/2024]
Abstract
Neurofilaments are structural proteins found in the cytoplasm of neurons, particularly in axons, providing structural support and stability to the axon. They consist of multiple subunits, including NF-H, NF-M, and NF-L, which form long filaments along the axon's length. Neurofilaments are crucial for maintaining the shape and integrity of neurons, promoting axonal transport, and regulating neuronal function. They are part of the intermediate filament (IF) family, which has approximately 70 tissue-specific genes. This diversity allows for a customizable cytoplasmic meshwork, adapting to the unique structural demands of different tissues and cell types. Neurofilament proteins show increased levels in both cerebrospinal fluid (CSF) and blood after neuroaxonal damage, indicating injury regardless of the underlying etiology. Precise measurement and long-term monitoring of damage are necessary for determining prognosis, assessing disease activity, tracking therapeutic responses, and creating treatments. These investigations contribute to our understanding of the importance of proper NF composition in fundamental neuronal processes and have implications for neurological disorders associated with NF abnormalities along with its alteration in different animal and human models. Here in this review, we have highlighted various neurological disorders such as Alzheimer's, Parkinson's, Huntington's, Dementia, and paved the way to use neurofilament as a marker in managing neurological disorders.
Collapse
Affiliation(s)
- Priti Sharma
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, India
| | - Aditi Giri
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, India.
| | - Prabhash Nath Tripathi
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, India.
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| |
Collapse
|
2
|
Jang Y, Oh S, Hall AJ, Zhang Z, Tropea TF, Chen-Plotkin A, Rosenthal LS, Dawson TM, Na CH, Pantelyat AY. Biomarker discovery in progressive supranuclear palsy from human cerebrospinal fluid. Clin Proteomics 2024; 21:56. [PMID: 39342078 PMCID: PMC11437921 DOI: 10.1186/s12014-024-09507-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Progressive supranuclear palsy (PSP) is a neurodegenerative disorder often misdiagnosed as Parkinson's Disease (PD) due to shared symptoms. PSP is characterized by the accumulation of tau protein in specific brain regions, leading to loss of balance, gaze impairment, and dementia. Diagnosing PSP is challenging, and there is a significant demand for reliable biomarkers. Existing biomarkers, including tau protein and neurofilament light chain (NfL) levels in cerebrospinal fluid (CSF), show inconsistencies in distinguishing PSP from other neurodegenerative disorders. Therefore, the development of new biomarkers for PSP is imperative. METHODS We conducted an extensive proteome analysis of CSF samples from 40 PSP patients, 40 PD patients, and 40 healthy controls (HC) using tandem mass tag-based quantification. Mass spectrometry analysis of 120 CSF samples was performed across 13 batches of 11-plex TMT experiments, with data normalization to reduce batch effects. Pathway, interactome, cell-type-specific enrichment, and bootstrap receiver operating characteristic analyses were performed to identify key candidate biomarkers. RESULTS We identified a total of 3,653 unique proteins. Our analysis revealed 190, 152, and 247 differentially expressed proteins in comparisons of PSP vs. HC, PSP vs. PD, and PSP vs. both PD and HC, respectively. Gene set enrichment and interactome analysis of the differentially expressed proteins in PSP CSF showed their involvement in cell adhesion, cholesterol metabolism, and glycan biosynthesis. Cell-type enrichment analysis indicated a predominance of neuronally-derived proteins among the differentially expressed proteins. The potential biomarker classification performance demonstrated that ATP6AP2 (reduced in PSP) had the highest AUC (0.922), followed by NEFM, EFEMP2, LAMP2, CHST12, FAT2, B4GALT1, LCAT, CBLN3, FSTL5, ATP6AP1, and GGH. CONCLUSION Biomarker candidate proteins ATP6AP2, NEFM, and CHI3L1 were identified as key differentiators of PSP from the other groups. This study represents the first large-scale use of mass spectrometry-based proteome analysis to identify cerebrospinal fluid (CSF) biomarkers specific to progressive supranuclear palsy (PSP) that can differentiate it from Parkinson's disease (PD) and healthy controls. Our findings lay a crucial foundation for the development and validation of reliable biomarkers, which will enhance diagnostic accuracy and facilitate early detection of PSP.
Collapse
Affiliation(s)
- Yura Jang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sungtaek Oh
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anna J Hall
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhen Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas F Tropea
- Department of Neurology, Perelman School of Medicine, Philadelphia, PA, USA
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, Philadelphia, PA, USA
| | - Liana S Rosenthal
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Chan Hyun Na
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Alexander Y Pantelyat
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Kale MB, Bhondge HM, Wankhede NL, Shende PV, Thanekaer RP, Aglawe MM, Rahangdale SR, Taksande BG, Pandit SB, Upaganlawar AB, Umekar MJ, Kopalli SR, Koppula S. Navigating the intersection: Diabetes and Alzheimer's intertwined relationship. Ageing Res Rev 2024; 100:102415. [PMID: 39002642 DOI: 10.1016/j.arr.2024.102415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/06/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Alzheimer's disease (AD) and Diabetes mellitus (DM) exhibit comparable pathophysiological pathways. Genetic abnormalities in APP, PS-1, and PS-2 are linked to AD, with diagnostic aid from CSF and blood biomarkers. Insulin dysfunction, termed "type 3 diabetes mellitus" in AD, involves altered insulin signalling and neuronal shrinkage. Insulin influences beta-amyloid metabolism, exacerbating neurotoxicity in AD and amyloid production in DM. Both disorders display impaired glucose transporter expression, hastening cognitive decline. Mitochondrial dysfunction and Toll-like receptor 4-mediated inflammation worsen neurodegeneration in both diseases. ApoE4 raises disease risk, especially when coupled with dyslipidemia common in DM. Targeting shared pathways like insulin-degrading enzyme activation and HSP60 holds promise for therapeutic intervention. Recognizing these interconnected mechanisms underscores the imperative for developing tailored treatments addressing the overlapping pathophysiology of AD and DM, offering potential avenues for more effective management of both conditions.
Collapse
Affiliation(s)
- Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | | | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Rushikesh P Thanekaer
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Manish M Aglawe
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sandip R Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sunil B Pandit
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
4
|
Wise A, Li J, Yamakawa M, Loureiro J, Peterson B, Worringer K, Sivasankaran R, Palma JA, Mitic L, Heuer HW, Lario-Lago A, Staffaroni AM, Clark A, Taylor J, Ljubenkov PA, Vandevrede L, Grinberg LT, Spina S, Seeley WW, Miller BL, Boeve BF, Dickerson BC, Grossman M, Litvan I, Pantelyat A, Tartaglia MC, Zhang Z, Wills AMA, Rexach J, Rojas JC, Boxer AL. CSF Proteomics in Patients With Progressive Supranuclear Palsy. Neurology 2024; 103:e209585. [PMID: 38959435 PMCID: PMC11226322 DOI: 10.1212/wnl.0000000000209585] [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: 08/10/2023] [Accepted: 05/15/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Identification of fluid biomarkers for progressive supranuclear palsy (PSP) is critical to enhance therapeutic development. We implemented unbiased DNA aptamer (SOMAmer) proteomics to identify novel CSF PSP biomarkers. METHODS This is a cross-sectional study in original (18 clinically diagnosed PSP-Richardson syndrome [PSP-RS], 28 cognitively healthy controls]), validation (23 PSP-RS, 26 healthy controls), and neuropathology-confirmed (21 PSP, 52 non-PSP frontotemporal lobar degeneration) cohorts. Participants were recruited through the University of California, San Francisco, and the 4-Repeat Neuroimaging Initiative. The original and neuropathology cohorts were analyzed with the SomaScan platform version 3.0 (5026-plex) and the validation cohort with version 4.1 (7595-plex). Clinical severity was measured with the PSP Rating Scale (PSPRS). CSF proteomic data were analyzed to identify differentially expressed targets, implicated biological pathways using enrichment and weighted consensus gene coexpression analyses, diagnostic value of top targets with receiver-operating characteristic curves, and associations with disease severity with linear regressions. RESULTS A total of 136 participants were included (median age 70.6 ± 8 years, 68 [50%] women). One hundred fifty-five of 5,026 (3.1%), 959 of 7,595 (12.6%), and 321 of 5,026 (6.3%) SOMAmers were differentially expressed in PSP compared with controls in original, validation, and neuropathology-confirmed cohorts, with most of the SOMAmers showing reduced signal (83.1%, 95.1%, and 73.2%, respectively). Three coexpression modules were associated with PSP across cohorts: (1) synaptic function/JAK-STAT (β = -0.044, corrected p = 0.002), (2) vesicle cytoskeletal trafficking (β = 0.039, p = 0.007), and (3) cytokine-cytokine receptor interaction (β = -0.032, p = 0.035) pathways. Axon guidance was the top dysregulated pathway in PSP in original (strength = 1.71, p < 0.001), validation (strength = 0.84, p < 0.001), and neuropathology-confirmed (strength = 0.78, p < 0.001) cohorts. A panel of axon guidance pathway proteins discriminated between PSP and controls in original (area under the curve [AUC] = 0.924), validation (AUC = 0.815), and neuropathology-confirmed (AUC = 0.932) cohorts. Two inflammatory proteins, galectin-10 and cytotoxic T lymphocyte-associated protein-4, correlated with PSPRS scores across cohorts. DISCUSSION Axon guidance pathway proteins and several other molecular pathways are downregulated in PSP, compared with controls. Proteins in these pathways may be useful targets for biomarker or therapeutic development.
Collapse
Affiliation(s)
- Amy Wise
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Jingyao Li
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Mai Yamakawa
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Joseph Loureiro
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Brant Peterson
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Kathleen Worringer
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Rajeev Sivasankaran
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Jose-Alberto Palma
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Laura Mitic
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Hilary W Heuer
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Argentina Lario-Lago
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Adam M Staffaroni
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Annie Clark
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Jack Taylor
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Peter A Ljubenkov
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Lawren Vandevrede
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Lea T Grinberg
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Salvatore Spina
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - William W Seeley
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Bruce L Miller
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Bradley F Boeve
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Bradford C Dickerson
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Murray Grossman
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Irene Litvan
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Alexander Pantelyat
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Maria Carmela Tartaglia
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Zihan Zhang
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Anne-Marie A Wills
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Jessica Rexach
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Julio C Rojas
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Adam L Boxer
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| |
Collapse
|
5
|
Dunning EE, Decourt B, Zawia NH, Shill HA, Sabbagh MN. Pharmacotherapies for the Treatment of Progressive Supranuclear Palsy: A Narrative Review. Neurol Ther 2024; 13:975-1013. [PMID: 38743312 PMCID: PMC11263316 DOI: 10.1007/s40120-024-00614-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 03/26/2024] [Indexed: 05/16/2024] Open
Abstract
Progressive supranuclear palsy (PSP) is a neurodegenerative disorder resulting from the deposition of misfolded and neurotoxic forms of tau protein in specific areas of the midbrain, basal ganglia, and cortex. It is one of the most representative forms of tauopathy. PSP presents in several different phenotypic variations and is often accompanied by the development of concurrent neurodegenerative disorders. PSP is universally fatal, and effective disease-modifying therapies for PSP have not yet been identified. Several tau-targeting treatment modalities, including vaccines, monoclonal antibodies, and microtubule-stabilizing agents, have been investigated and have had no efficacy. The need to treat PSP and other tauopathies is critical, and many clinical trials investigating tau-targeted treatments are underway. In this review, the PubMed database was queried to collect information about preclinical and clinical research on PSP treatment. Additionally, the US National Library of Medicine's ClinicalTrials.gov website was queried to identify past and ongoing clinical trials relevant to PSP treatment. This narrative review summarizes our findings regarding these reports, which include potential disease-modifying drug trials, modifiable risk factor management, and symptom treatments.
Collapse
Affiliation(s)
- Elise E Dunning
- Creighton University School of Medicine - Phoenix, Phoenix, AZ, USA
| | - Boris Decourt
- Department of Pharmacology and Neuroscience School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Laboratory on Neurodegeneration and Translational Research, College of Medicine, Roseman University of Health Sciences, Las Vegas, NV, USA
| | - Nasser H Zawia
- Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
- Department of Biomedical and Pharmaceutical Sciences, Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
| | - Holly A Shill
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W Thomas Rd, Phoenix, AZ, 85013, USA
| | - Marwan N Sabbagh
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W Thomas Rd, Phoenix, AZ, 85013, USA.
| |
Collapse
|
6
|
Tang S, Xiao Z, Lin F, Liang X, Ma X, Wu J, Zhou X, Zhao Q, Gao J, Xiao Q, Ding D. Joint effect of testosterone and neurofilament light chain on cognitive decline in men: The Shanghai Aging Study. Alzheimers Dement 2024; 20:5290-5298. [PMID: 38837321 PMCID: PMC11350006 DOI: 10.1002/alz.13889] [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: 01/17/2024] [Revised: 04/08/2024] [Accepted: 04/22/2024] [Indexed: 06/07/2024]
Abstract
INTRODUCTION The association of testosterone and cognitive decline is inconclusive, and its joint effect with neurofilaments light chain (NfL) remains largely unknown. METHODS A total of 581 non-demented older men in the Shanghai Aging Study were included. Blood total testosterone (TT), free testosterone (FT), and NfL were measured at baseline. The relationships between TT, FT, TT/FT-NfL, and cognitive decline were explored by Cox regression models. RESULTS During a median follow-up of 6.7 years, there was an inverse association between TT/FT and cognitive decline (TT, trend p = 0.004, Q1 vs Q4, hazard ratio [HR] = 4.39, 95% confidence interval [CI] = 1.60 to 12.04; FT, trend p = 0.002, Q1 vs Q4, HR = 5.29, 95% CI = 1.50 to 16.89). Compared to participants with high TT/FT-low NfL, those with low TT/FT-high NfL had significantly higher risks of cognitive decline (TT, HR = 5.10, 95% CI = 1.11 to 23.40; FT, HR = 6.14, 95% CI = 1.34 to 28.06). DISCUSSION Our findings suggest that the combination of testosterone and neurodegenerative markers may provide reliable predictive insights into future cognitive decline. HIGHLIGHTS Testosterone is inversely associated with cognitive decline in older men. There is a joint effect of testosterone and NfL on cognitive decline. Sex hormone and neurodegeneration may synergistically contribute to cognitive deterioration.
Collapse
Affiliation(s)
- Shuning Tang
- Department of Preventive Medicine and Health EducationSchool of Public HealthThe Key Laboratory of Public Health Safety of Ministry of EducationFudan UniversityShanghaiChina
| | - Zhenxu Xiao
- Institute of NeurologyHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DisordersHuashan HospitalFudan UniversityShanghaiChina
| | - Fangting Lin
- Department of Preventive Medicine and Health EducationSchool of Public HealthThe Key Laboratory of Public Health Safety of Ministry of EducationFudan UniversityShanghaiChina
| | - Xiaoniu Liang
- Institute of NeurologyHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DisordersHuashan HospitalFudan UniversityShanghaiChina
| | - Xiaoxi Ma
- Institute of NeurologyHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DisordersHuashan HospitalFudan UniversityShanghaiChina
| | - Jie Wu
- Institute of NeurologyHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DisordersHuashan HospitalFudan UniversityShanghaiChina
| | - Xiaowen Zhou
- Institute of NeurologyHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DisordersHuashan HospitalFudan UniversityShanghaiChina
| | - Qianhua Zhao
- Institute of NeurologyHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DisordersHuashan HospitalFudan UniversityShanghaiChina
- MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Junling Gao
- Department of Preventive Medicine and Health EducationSchool of Public HealthThe Key Laboratory of Public Health Safety of Ministry of EducationFudan UniversityShanghaiChina
- Health Communication InstituteFudan UniversityShanghaiChina
| | - Qianyi Xiao
- Department of Preventive Medicine and Health EducationSchool of Public HealthThe Key Laboratory of Public Health Safety of Ministry of EducationFudan UniversityShanghaiChina
- Health Communication InstituteFudan UniversityShanghaiChina
| | - Ding Ding
- Institute of NeurologyHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DisordersHuashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
7
|
Abukuri DN. Novel Biomarkers for Alzheimer's Disease: Plasma Neurofilament Light and Cerebrospinal Fluid. Int J Alzheimers Dis 2024; 2024:6668159. [PMID: 38779175 PMCID: PMC11111307 DOI: 10.1155/2024/6668159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 03/18/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD) represent an increasingly significant public health concern. As clinical diagnosis faces challenges, biomarkers are becoming increasingly important in research, trials, and patient assessments. While biomarkers like amyloid-β peptide, tau proteins, CSF levels (Aβ, tau, and p-tau), and neuroimaging techniques are commonly used in AD diagnosis, they are often limited and invasive in monitoring and diagnosis. For this reason, blood-based biomarkers are the optimal choice for detecting neurodegeneration in brain diseases due to their noninvasiveness, affordability, reliability, and consistency. This literature review focuses on plasma neurofilament light (NfL) and CSF NfL as blood-based biomarkers used in recent AD diagnosis. The findings revealed that the core CSF biomarkers of neurodegeneration (T-tau, P-tau, and Aβ42), CSF NFL, and plasma T-tau were strongly associated with Alzheimer's disease, and the core biomarkers were strongly associated with mild cognitive impairment due to Alzheimer's disease. Elevated levels of plasma and cerebrospinal fluid NfL were linked to decreased [18F]FDG uptake in corresponding brain areas. In participants with Aβ positivity (Aβ+), NfL correlated with reduced metabolism in regions susceptible to Alzheimer's disease. In addition, CSF NfL levels correlate with brain atrophy and predict cognitive changes, while plasma total tau does not. Plasma P-tau, especially in combination with Aβ42/Aβ40, is promising for symptomatic AD stages. Though not AD-exclusive, blood NfL holds promise for neurodegeneration detection and assessing treatment efficacy. Given the consistent levels of T-tau, P-tau, Aβ42, and NFL in CSF, their incorporation into both clinical practice and research is highly recommended.
Collapse
|
8
|
Franco-O'Byrne D, Santamaría-García H, Migeot J, Ibáñez A. Emerging Theories of Allostatic-Interoceptive Overload in Neurodegeneration. Curr Top Behav Neurosci 2024. [PMID: 38637414 DOI: 10.1007/7854_2024_471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Recent integrative multilevel models offer novel insights into the etiology and course of neurodegenerative conditions. The predictive coding of allostatic-interoception theory posits that the brain adapts to environmental demands by modulating internal bodily signals through the allostatic-interoceptive system. Specifically, a domain-general allostatic-interoceptive network exerts adaptive physiological control by fine-tuning initial top-down predictions and bottom-up peripheral signaling. In this context, adequate adaptation implies the minimization of prediction errors thereby optimizing energy expenditure. Abnormalities in top-down interoceptive predictions or peripheral signaling can trigger allostatic overload states, ultimately leading to dysregulated interoceptive and bodily systems (endocrine, immunological, circulatory, etc.). In this context, environmental stress, social determinants of health, and harmful exposomes (i.e., the cumulative life-course exposition to different environmental stressors) may interact with physiological and genetic factors, dysregulating allostatic interoception and precipitating neurodegenerative processes. We review the allostatic-interoceptive overload framework across different neurodegenerative diseases, particularly in the behavioral variant frontotemporal dementia (bvFTD). We describe how concepts of allostasis and interoception could be integrated with principles of predictive coding to explain how the brain optimizes adaptive responses, while maintaining physiological stability through feedback loops with multiple organismic systems. Then, we introduce the model of allostatic-interoceptive overload of bvFTD and discuss its implications for the understanding of pathophysiological and neurocognitive abnormalities in multiple neurodegenerative conditions.
Collapse
Affiliation(s)
- Daniel Franco-O'Byrne
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibáñez, Santiago, Chile
| | - Hernando Santamaría-García
- Global Brain Health Institute, University of California-San Francisco, San Francisco, CA, USA
- Trinity College Dublin, Dublin, Ireland
- Department of Psychiatry, Pontificia Universidad Javeriana, Bogotá, Colombia
- Center of Memory and Cognition Intellectus, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Joaquín Migeot
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibáñez, Santiago, Chile
| | - Agustín Ibáñez
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile.
- Global Brain Health Institute, University of California-San Francisco, San Francisco, CA, USA.
- Trinity College Dublin, Dublin, Ireland.
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina.
- Trinity College Institute of Neuroscience (TCIN), Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
9
|
Ibanez A, Kringelbach ML, Deco G. A synergetic turn in cognitive neuroscience of brain diseases. Trends Cogn Sci 2024; 28:319-338. [PMID: 38246816 DOI: 10.1016/j.tics.2023.12.006] [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: 08/31/2023] [Revised: 12/15/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
Despite significant improvements in our understanding of brain diseases, many barriers remain. Cognitive neuroscience faces four major challenges: complex structure-function associations; disease phenotype heterogeneity; the lack of transdiagnostic models; and oversimplified cognitive approaches restricted to the laboratory. Here, we propose a synergetics framework that can help to perform the necessary dimensionality reduction of complex interactions between the brain, body, and environment. The key solutions include low-dimensional spatiotemporal hierarchies for brain-structure associations, whole-brain modeling to handle phenotype diversity, model integration of shared transdiagnostic pathophysiological pathways, and naturalistic frameworks balancing experimental control and ecological validity. Creating whole-brain models with reduced manifolds combined with ecological measures can improve our understanding of brain disease and help identify novel interventions. Synergetics provides an integrated framework for future progress in clinical and cognitive neuroscience, pushing the boundaries of brain health and disease toward more mature, naturalistic approaches.
Collapse
Affiliation(s)
- Agustin Ibanez
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile; Global Brain Health Institute (GBHI), University California San Francisco (UCSF), San Francisco, CA, USA; Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland; Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina; Department of Psychiatry, University of Oxford, Oxford, UK.
| | - Morten L Kringelbach
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Centre for Eudaimonia and Human Flourishing, University of Oxford, Oxford, UK
| | - Gustavo Deco
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Roc Boronat 138, Barcelona 08018, Spain; Institució Catalana de la Recerca i Estudis Avançats (ICREA), Passeig Lluís Companys 23, Barcelona 08010, Spain.
| |
Collapse
|
10
|
Lane-Donovan C, Boxer AL. Disentangling tau: One protein, many therapeutic approaches. Neurotherapeutics 2024; 21:e00321. [PMID: 38278659 PMCID: PMC10963923 DOI: 10.1016/j.neurot.2024.e00321] [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: 10/31/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/28/2024] Open
Abstract
The tauopathies encompass over 20 adult neurodegenerative diseases and are characterized by the dysfunction and accumulation of insoluble tau protein. Among them, Alzheimer's disease, frontotemporal dementia, and progressive supranuclear palsy collectively impact millions of patients and their families worldwide. Despite years of drug development using a variety of mechanisms of action, no therapeutic directed against tau has been approved for clinical use. This raises important questions about our current model of tau pathology and invites thoughtful consideration of our approach to nonclinical models and clinical trial design. In this article, we review what is known about the biology and genetics of tau, placing it in the context of current and failed clinical trials. We highlight potential reasons for the lack of success to date and offer suggestions for new pathways in therapeutic development. Overall, our viewpoint to the future is optimistic for this important group of neurodegenerative diseases.
Collapse
Affiliation(s)
- Courtney Lane-Donovan
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA 94158, USA.
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA 94158, USA
| |
Collapse
|
11
|
Ibanez A, Northoff G. Intrinsic timescales and predictive allostatic interoception in brain health and disease. Neurosci Biobehav Rev 2024; 157:105510. [PMID: 38104789 PMCID: PMC11184903 DOI: 10.1016/j.neubiorev.2023.105510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/29/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
The cognitive neuroscience of brain diseases faces challenges in understanding the complex relationship between brain structure and function, the heterogeneity of brain phenotypes, and the lack of dimensional and transnosological explanations. This perspective offers a framework combining the predictive coding theory of allostatic interoceptive overload (PAIO) and the intrinsic neural timescales (INT) theory to provide a more dynamic understanding of brain health in psychiatry and neurology. PAIO integrates allostasis and interoception to assess the interaction between internal patterns and environmental stressors, while INT shows that different brain regions operate on different intrinsic timescales. The allostatic overload can be understood as a failure of INT, which involves a breakdown of proper temporal integration and segregation. This can lead to dimensional disbalances between exteroceptive/interoceptive inputs across brain and whole-body levels (cardiometabolic, cardiovascular, inflammatory, immune). This approach offers new insights, presenting novel perspectives on brain spatiotemporal hierarchies and interactions. By integrating these theories, the paper opens innovative paths for studying brain health dynamics, which can inform future research in brain health and disease.
Collapse
Affiliation(s)
- Agustin Ibanez
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), CA, USA; Latin American Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile; Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina; Trinity College Dublin, Dublin, Ireland.
| | - Georg Northoff
- Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China; Center for Cognition and Brain Disorders, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, People's Republic of China; Mind, Brain Imaging and Neuroethics, Institute of Mental Health Research, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
12
|
Fang T, Dai Y, Hu X, Xu Y, Qiao J. Evaluation of serum neurofilament light chain and glial fibrillary acidic protein in the diagnosis of Alzheimer's disease. Front Neurol 2024; 15:1320653. [PMID: 38352136 PMCID: PMC10861667 DOI: 10.3389/fneur.2024.1320653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024] Open
Abstract
Purpose This study aimed to evaluate the use of serum neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) in the diagnosis of Alzheimer's disease (AD) and the differential diagnosis between AD and mild cognitive impairment (MCI). Methods From September 2021 to October 2022, we collected venous blood from patients and healthy individuals who visited our hospital's Neurology Department, and we isolated serum to detect NfL and GFAP using direct chemiluminescence. The results were analyzed using one-way analysis of variance (ANOVA) analysis and receiver operating characteristic (ROC) curves. Results Pairwise comparisons among the three groups showed that compared with the health checkup (HC) group, serum NfL and GFAP were increased in both AD and MCI (PNfL < 0.05, PGFAP < 0.01). There were significant differences in GFAP between MCI and AD groups, and the level in AD group was higher (p < 0.01), while there was no difference in NfL. Both serum NfL and serum GFAP levels can independently diagnose AD (p < 0.01). The ROC curve showed that GFAP had a higher diagnostic efficacy, with an area under the ROC curve (AUC) of 0.928. The cut-off values of the two serum markers for the diagnosis of AD were NfL > 40.09 pg./mL and GFAP >31.40 pg./mL. Sensitivity and specificity for NfL in the diagnosis of AD were 59.6 and 76.2%, respectively, and for GFAP, they were 90.4 and 82.1%, respectively. The combined diagnosis of GFAP and NfL improved the diagnostic efficiency (AUC = 0.931, sensitivity = 78.8%, specificity = 92.3%). The cut-off value of GFAP for the differential diagnosis of MCI and AD was 46.05 pg./mL. Conclusion Both serum NfL and serum GFAP can be used as biomarkers for the diagnosis of AD. Serum GFAP has better diagnostic efficacy and can distinguish AD from MCI. A combined diagnosis can improve diagnostic specificity.
Collapse
Affiliation(s)
| | | | | | - Yuanhong Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jinping Qiao
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
Palleis C, Franzmeier N, Weidinger E, Bernhardt AM, Katzdobler S, Wall S, Ferschmann C, Harris S, Schmitt J, Schuster S, Gnörich J, Finze A, Biechele G, Lindner S, Albert NL, Bartenstein P, Sabri O, Barthel H, Rupprecht R, Nuscher B, Stephens AW, Rauchmann BS, Perneczky R, Haass C, Brendel M, Levin J, Höglinger GU. Association of Neurofilament Light Chain, [ 18F]PI-2620 Tau-PET, TSPO-PET, and Clinical Progression in Patients With β-Amyloid-Negative CBS. Neurology 2024; 102:e207901. [PMID: 38165362 PMCID: PMC10834119 DOI: 10.1212/wnl.0000000000207901] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 10/03/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Corticobasal syndrome (CBS) with underlying 4-repeat tauopathy is a progressive neurodegenerative disease characterized by declining cognitive and motor functions. Biomarkers for assessing pathologic brain changes in CBS including tau-PET, 18 kDa translocator protein (TSPO)-PET, structural MRI, neurofilament light chain (NfL), or glial fibrillary acidic protein (GFAP) have recently been evaluated for differential diagnosis and disease staging, yet their association with disease trajectories remains unclear. Therefore, we performed a head-to-head comparison of neuroimaging (tau-PET, TSPO-PET, structural MRI) and plasma biomarkers (NfL, GFAP) as prognostic tools for longitudinal clinical trajectories in β-amyloid (Aβ)-negative CBS. METHODS We included patients with clinically diagnosed Aβ-negative CBS with clinical follow-up data who underwent baseline structural MRI and plasma-NfL analysis for assessing neurodegeneration, [18F]PI-2620-PET for assessing tau pathology, [18F]GE-180-PET for assessing microglia activation, and plasma-GFAP analysis for assessing astrocytosis. To quantify tau and microglia load, we assessed summary scores of whole-brain, cortical, and subcortical PET signal. For structural MRI analysis, we quantified subcortical and cortical gray matter volume. Plasma NfL and GFAP values were assessed using Simoa-based immunoassays. Symptom progression was determined using a battery of cognitive and motor tests (i.e., Progressive Supranuclear Palsy Rating Scale [PSPRS]). Using linear mixed models, we tested whether the assessed biomarkers at baseline were associated with faster symptom progression over time (i.e., time × biomarker interaction). RESULTS Overall, 21 patients with Aβ-negative CBS with ∼2-year clinical follow-up data were included. Patients with CBS with more widespread global tau-PET signal showed faster clinical progression (PSPRS: B/SE = 0.001/0.0005, p = 0.025), driven by cortical rather than subcortical tau-PET. By contrast, patients with higher global [18F]GE-180-PET readouts showed slower clinical progression (PSPRS: B/SE = -0.056/0.023, p = 0.019). No association was found between gray matter volume and clinical progression. Concerning fluid biomarkers, only higher plasma-NfL (PSPRS: B/SE = 0.176/0.046, p < 0.001) but not GFAP was associated with faster clinical deterioration. In a subsequent sensitivity analysis, we found that tau-PET, TSPO-PET, and plasma-NfL showed significant interaction effects with time on clinical trajectories when tested in the same model. DISCUSSION [18F]PI-2620 tau-PET, [18F]GE-180 TSPO-PET, and plasma-NfL show prognostic potential for clinical progression in patients with Aβ-negative CBS with probable 4-repeat tauopathy, which can be useful for clinical decision-making and stratifying patients in clinical trials.
Collapse
Affiliation(s)
- Carla Palleis
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Nicolai Franzmeier
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Endy Weidinger
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Alexander M Bernhardt
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Sabrina Katzdobler
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Stephan Wall
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Christian Ferschmann
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Stefanie Harris
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Julia Schmitt
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Sebastian Schuster
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Johannes Gnörich
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Anika Finze
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Gloria Biechele
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Simon Lindner
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Nathalie L Albert
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Peter Bartenstein
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Osama Sabri
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Henryk Barthel
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Rainer Rupprecht
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Brigitte Nuscher
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Andrew W Stephens
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Boris-Stephan Rauchmann
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Robert Perneczky
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Christian Haass
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Matthias Brendel
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Johannes Levin
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Günter U Höglinger
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| |
Collapse
|
14
|
Chopra A, Outeiro TF. Aggregation and beyond: alpha-synuclein-based biomarkers in synucleinopathies. Brain 2024; 147:81-90. [PMID: 37526295 DOI: 10.1093/brain/awad260] [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: 04/13/2023] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 08/02/2023] Open
Abstract
Parkinson's disease is clinically known for the loss of dopaminergic neurons in the substantia nigra pars compacta and accumulation of intraneuronal cytoplasmic inclusions rich in alpha-synuclein called 'Lewy bodies' and 'Lewy neurites'. Together with dementia with Lewy bodies and multiple system atrophy, Parkinson's disease is part of a group of disorders called synucleinopathies. Currently, diagnosis of synucleinopathies is based on the clinical assessment which often takes place in advanced disease stages. While the causal role of alpha-synuclein aggregates in these disorders is still debatable, measuring the levels, types or seeding properties of different alpha-synuclein species hold great promise as biomarkers. Recent studies indicate significant differences in peptide, protein and RNA levels in blood samples from patients with Parkinson's disease. Seed amplification assays using CSF, blood, skin biopsy, olfactory swab samples show great promise for detecting synucleinopathies and even for discriminating between different synucleinopathies. Interestingly, small extracellular vesicles, such as exosomes, display differences in their cargoes in Parkinson's disease patients versus controls. In this update, we focus on alpha-synuclein aggregation and possible sources of disease-related species released in extracellular vesicles, which promise to revolutionize the diagnosis and the monitoring of disease progression.
Collapse
Affiliation(s)
- Avika Chopra
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37075 Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK
- Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 37075 Göttingen, Germany
| |
Collapse
|
15
|
Dey S, Yelamanchi R, Mullapudi T, Holla VV, Kamble N, Mahale RR, Sathyaprabha TN, Pal PK, Debnath M, Yadav R. Association of Insulin-like Growth Factor-1 and Neurofilament Light Chain in Patients with Progressive Supranuclear Palsy. Ann Indian Acad Neurol 2024; 27:40-45. [PMID: 38495245 PMCID: PMC10941886 DOI: 10.4103/aian.aian_507_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 11/18/2023] [Accepted: 11/24/2023] [Indexed: 03/19/2024] Open
Abstract
Background Progressive supranuclear palsy (PSP) is the most common primary tauopathy. The definite diagnosis of PSP is established by histopathologic changes in the brain. There are no reliable blood-based biomarkers to aid the diagnosis of this fatal disease at an early stage. Also, the precise etiopathology of PSP and its variants is inadequately understood. Objective Blood-based molecules such as neurofilament light chain (NfL) and insulin-like growth factor-1 (IGF-1) are shown as important markers of neurodegenerative and aging processes, respectively. These two biomarkers have not been analyzed simultaneously in PSP patients. Methods To address this knowledge gap, 40 PSP patients and equal number of healthy individuals were recruited and serum levels of NfL and IGF-1 were assayed in all the study participants by enzyme-linked immunosorbent assay (ELISA). Motor and nonmotor symptoms were evaluated in PSP patients using various scales/questionnaires. Cardiac autonomic function tests were performed in a subset of patients (n = 27). Results A significantly high serum level of NfL (P < 0.01) and a reduced level of IGF-1 (P = 0.02) were observed in PSP patients compared to healthy controls. Besides, a negative correlation (r = -0.54, P < 0.01) between NfL and IGF-1 levels was observed in PSP patients. Conclusion The finding of this study reinforces the important role of blood NfL level as a potential biomarker of PSP. Further, the current study provides novel insights into the reciprocal correlation between NfL and IGF-1 in PSP patients. Combined analysis of blood levels of these two functionally relevant markers might be useful in the prediction and diagnosis of PSP.
Collapse
Affiliation(s)
- Saikat Dey
- Department of Human Genetics, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Ramachadra Yelamanchi
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Thrinath Mullapudi
- Department of Human Genetics, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Vikram V. Holla
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Nitish Kamble
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Rohan R. Mahale
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Talakad N. Sathyaprabha
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Pramod K. Pal
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Monojit Debnath
- Department of Human Genetics, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Ravi Yadav
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| |
Collapse
|
16
|
Mohaupt P, Vialaret J, Hirtz C, Lehmann S. Increasing the sensitivity of Simoa via bead count reduction facilitates the quantification of pTau-181 in dried plasma spots. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12456. [PMID: 38476928 PMCID: PMC10927909 DOI: 10.1002/trc2.12456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/22/2024] [Indexed: 03/14/2024]
Abstract
Introduction The exclusion of affected populations from Alzheimer's disease (AD) clinical research limits our understanding of disease heterogeneity and its impact on clinical care. While micro sampling with dried plasma spots (DPS) can promote inclusivity by enabling sample collection in remote areas, current techniques lack the sensitivity required for the quantification of phosphorylated tau at Thr181 (pTau-181) in DPS extracts. Methods We developed an assay for pTau-181 with reduced bead count and improved bead read efficiency (BRE) using a prototype Simoa instrument. This novel assay's performance was evaluated against standard pTau-181 assays on two Simoa platforms, and DPS extracts were tested for pTau-181 quantification feasibility. Results The novel assay quantifies pTau-181 at concentrations up to 16x lower than traditional pTau-181 assays on HD-X and SR-X platforms. DPS extracts tested with our low-bead assay were quantified considerably above the lower limit of quantification (LLOQ), indicating the suitability of this assay for future DPS extract measurements. Discussion Implementing DPS sampling and pTau-181 quantification could increase participation from underrepresented groups in AD research. However, additional assay optimization and an in-depth study of preanalytical sample stability are essential for the transition to clinical applicability.
Collapse
Affiliation(s)
- Pablo Mohaupt
- LBPC‐PPCUniversité de MontpellierIRMB CHU de MontpellierINM INSERMMontpellierFrance
| | - Jérôme Vialaret
- LBPC‐PPCUniversité de MontpellierIRMB CHU de MontpellierINM INSERMMontpellierFrance
| | - Christophe Hirtz
- LBPC‐PPCUniversité de MontpellierIRMB CHU de MontpellierINM INSERMMontpellierFrance
| | - Sylvain Lehmann
- LBPC‐PPCUniversité de MontpellierIRMB CHU de MontpellierINM INSERMMontpellierFrance
| |
Collapse
|
17
|
Wojdała AL, Bellomo G, Gaetani L, Toja A, Chipi E, Shan D, Chiasserini D, Parnetti L. Trajectories of CSF and plasma biomarkers across Alzheimer's disease continuum: disease staging by NF-L, p-tau181, and GFAP. Neurobiol Dis 2023; 189:106356. [PMID: 37977432 DOI: 10.1016/j.nbd.2023.106356] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/02/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023] Open
Abstract
CSF-to-plasma transition will open new avenues for molecular phenotyping of Alzheimer's disease (AD). Here we evaluated a panel of AD biomarkers in matched CSF and plasma samples across the AD continuum, from preclinical AD to dementia. The aims were to: 1) compare diagnostic performance of the two biofluids, 2) evaluate trajectories of the biomarkers along AD progression. We analyzed CSF and plasma Aβ42/40, p-tau181, p-tau231, t-tau, NF-L, GFAP, UCHL-1 and CSF SNAP-25 in a cohort (n = 173) of preclinical AD, MCI-AD, AD dementia, frontotemporal dementia patients, and controls. We found a significant correlation between CSF and plasma levels of Aβ42/40, p-tau181, p-tau231, NF-L, and GFAP, while no CSF-plasma correlation was observed for t-tau and UCHL-1. Next to the core CSF biomarkers (Aβ42/40, p-tau181, t-tau), those providing the best discrimination between controls and preclinical AD were CSF p-tau231 and SNAP-25 and plasma Aβ42/40, p-tau231, and GFAP. Among plasma biomarkers, we found Aβ42/Aβ40, GFAP, and p-tau231 to show the largest rate of change at the CSF biomarker-defined cut-offs for amyloidosis and tauopathy. Finally, we identified GFAP, NF-L, and p-tau181 as the biomarkers most significantly associated with disease progression in both CSF and plasma. We suggest that a well-standardized and validated panel of selected plasma markers can facilitate early AD diagnosis, even at the asymptomatic disease stage. We propose that both CSF and plasma measurement of NF-L, p-tau181, and GFAP may play a significant role in disease staging and monitoring.
Collapse
Affiliation(s)
- Anna Lidia Wojdała
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Giovanni Bellomo
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Andrea Toja
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Elena Chipi
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Dandan Shan
- Quanterix Corporation, Billerica, MA 01821, United States of America
| | - Davide Chiasserini
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy.
| | - Lucilla Parnetti
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy.
| |
Collapse
|
18
|
Bernick C, Shan G, Ritter A, Ashton NJ, Blennow K, Lantero-Rodriguez J, Snellman A, Zetterberg H. Blood biomarkers and neurodegeneration in individuals exposed to repetitive head impacts. Alzheimers Res Ther 2023; 15:173. [PMID: 37828595 PMCID: PMC10571311 DOI: 10.1186/s13195-023-01310-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND It is unknown if fluid biomarkers reflective of brain pathologies are useful in detecting and following a neurodegenerative process in individuals exposed to repetitive head impacts. This study explores the relationship between blood biomarkers and longitudinal change in cognitive function and regional brain volumes in a cohort of professional fighters. METHODS Participants are drawn from a convenience sample of active and retired professional boxers and Mixed Martial Arts fighters and a control group with no prior exposure to head impacts. 3 T MRI brain imaging, plasma samples, and computerized cognitive testing were obtained at baseline and, for a subset, annually. MRI regional volumes were extracted, along with plasma levels of neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), p-tau231, and N-terminal tau (NTA). Statistical analyses were performed to assess the relationship between plasma levels and regional brain volumes and cognitive performance at baseline and longitudinally. RESULTS One hundred forty active boxers (mean age: 31 with standard deviation (SD) of 8), 211 active MMA (mean age of 30 with SD of 5), 69 retired boxers (mean age 49 with SD of 9), and 52 control participants (mean age 36 with SD of 12) were included in the analyses. Baseline GFAP levels were highest in the retired boxers (retired boxers v. active MMA: p = 0.0191), whereas active boxers had higher levels of NfL (active boxers v. MMA: p = 0.047). GFAP showed an increase longitudinally in retired boxers that was associated with decreasing volumes of multiple cortical and subcortical structures (e.g., hippocampus: B = - 1.25, 95% CI, - 1.65 to - 0.85) and increase in lateral ventricle size (B = 1.75, 95% CI, 1.46 to 2.04). Furthermore, performance on cognitive domains including memory, processing speed, psychomotor speed, and reaction time declined over time with increasing GFAP (e.g., processing speed: B = - 0.04, 95% CI, - 0.07 to - 0.02; reaction time: B = 0.52, 95% CI, 0.28 to 0.76). Among active fighters, increasing levels of GFAP were correlated with lower thalamic (B = - 1.42, 95% CI, - 2.34 to -0.49) and corpus callosum volumes, along with worsening scores on psychomotor speed (B = 0.14, 95% CI, 0.01 to 0.27). CONCLUSION Longitudinal plasma GFAP levels may have a role in identifying individuals exposed to repetitive head impacts who are at risk of showing progressive regional atrophy and cognitive decline.
Collapse
Affiliation(s)
- Charles Bernick
- Neurological Institute, Cleveland Clinic, Las Vegas, NV, USA.
| | - Guogen Shan
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Aaron Ritter
- Neurological Institute, Cleveland Clinic, Las Vegas, NV, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Anniina Snellman
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
19
|
Peng L, Wan L, Liu M, Long Z, Chen D, Yuan X, Tang Z, Fu Y, Zhu S, Lei L, Wang C, Peng H, Shi Y, He L, Yuan H, Wan N, Hou X, Xia K, Li J, Chen C, Qiu R, Tang B, Chen Z, Jiang H. Diagnostic and prognostic performance of plasma neurofilament light chain in multiple system atrophy: a cross-sectional and longitudinal study. J Neurol 2023; 270:4248-4261. [PMID: 37184660 DOI: 10.1007/s00415-023-11741-y] [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: 02/15/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/16/2023]
Abstract
BACKGROUND The longitudinal dynamics of neurofilament light chain (NfL) in multiple system atrophy (MSA) were incompletely illuminated. This study aimed to explore whether the plasma NfL (pNfL) could serve as a potential biomarker of clinical diagnosis and disease progression for MSA. METHODS We quantified pNfL concentrations in both a large cross-sectional cohort with 214 MSA individuals, 65 PD individuals, and 211 healthy controls (HC), and a longitudinal cohort of 84 MSA patients. Propensity score matching (PSM) was used to balance the age between the three groups. The pNfL levels between groups were compared using Kruskal-Wallis test. Linear mixed models were performed to explore the disease progression-associated factors in longitudinal MSA cohort. Random forest model as a complement to linear models was employed to quantify the importance of predictors. RESULTS Before and after matching the age by PSM, the pNfL levels could reliably differentiate MSA from HC and PD groups, but only had mild potential to distinguish PD from HC. By combining linear and nonlinear models, we demonstrated that pNfL levels at baseline, rather than the change rate of pNfL, displayed potential prognostic value for progression of MSA. The combination of baseline pNfL levels and other modifiers, such as subtypes, Hoehn-Yahr stage at baseline, was first shown to improve the diagnosis accuracy. CONCLUSIONS Our study contributed to a better understanding of longitudinal dynamics of pNfL in MSA, and validated the values of pNfL as a non-invasive sensitive biomarker for the diagnosis and progression. The combination of pNfL and other factors is recommended for better monitoring and prediction of MSA progression.
Collapse
Affiliation(s)
- Linliu Peng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Linlin Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, Hunan, China
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, 410008, Hunan, China
| | - Mingjie Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Neurology, the Affiliated Nanhua Hospital, University of South China, Hengyang, 421002, Hunan, China
| | - Zhe Long
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Daji Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xinrong Yuan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhichao Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - You Fu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Sudan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Lijing Lei
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Chunrong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Huirong Peng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yuting Shi
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Lang He
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Hongyu Yuan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Na Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xuan Hou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Kun Xia
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, 410008, Hunan, China
| | - Jinchen Li
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Chao Chen
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, 410008, Hunan, China
| | - Rong Qiu
- School of Computer Science and Engineering, Central South University, Changsha, 410083, Hunan, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Zhao Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China.
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China.
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
20
|
Alcolea D, Beeri MS, Rojas JC, Gardner RC, Lleó A. Blood Biomarkers in Neurodegenerative Diseases: Implications for the Clinical Neurologist. Neurology 2023; 101:172-180. [PMID: 36878698 PMCID: PMC10435056 DOI: 10.1212/wnl.0000000000207193] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/31/2023] [Indexed: 03/08/2023] Open
Abstract
Blood-based biomarkers offer a major advance in the clinical evaluation of neurodegenerative diseases. Currently, research studies have reported robust assays of blood markers for the detection of amyloid and tau pathologies specific to Alzheimer disease (amyloid-β peptides, and p-tau) and nonspecific blood markers of neuronal (neurofilament light, β-synuclein, and ubiquitin-C-terminal-hydrolase-L1) and glial degeneration (glial fibrillary acidic protein) that can measure key pathophysiologic processes in several neurodegenerative diseases. In the near future, these markers may be used for screening, diagnosis, or disease and treatment response monitoring. Blood-based biomarkers for neurodegenerative diseases have been rapidly implemented in research, and they have the potential to enter clinical use soon in different clinical settings. In this review, we will describe the main developments and their potential implications for the general neurologist.
Collapse
Affiliation(s)
- Daniel Alcolea
- From the Sant Pau Memory Unit (D.A., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (D.A., A.L.), CIBERNED, Madrid, Spain; Department of Psychiatry (M.S.B.), Icahn School of Medicine at Mount Sinai, New York, NY; The Joseph Sagol Neuroscience (M.S.B., R.C.G.), Center Sheba Medical Center, Tel-Hashomer, Israel; and Department of Neurology (J.C.R.), Weill Institute for Neurosciences, UCSF Memory and Aging Center, San Francisco, CA.
| | - Michal Schnaider Beeri
- From the Sant Pau Memory Unit (D.A., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (D.A., A.L.), CIBERNED, Madrid, Spain; Department of Psychiatry (M.S.B.), Icahn School of Medicine at Mount Sinai, New York, NY; The Joseph Sagol Neuroscience (M.S.B., R.C.G.), Center Sheba Medical Center, Tel-Hashomer, Israel; and Department of Neurology (J.C.R.), Weill Institute for Neurosciences, UCSF Memory and Aging Center, San Francisco, CA
| | - Julio C Rojas
- From the Sant Pau Memory Unit (D.A., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (D.A., A.L.), CIBERNED, Madrid, Spain; Department of Psychiatry (M.S.B.), Icahn School of Medicine at Mount Sinai, New York, NY; The Joseph Sagol Neuroscience (M.S.B., R.C.G.), Center Sheba Medical Center, Tel-Hashomer, Israel; and Department of Neurology (J.C.R.), Weill Institute for Neurosciences, UCSF Memory and Aging Center, San Francisco, CA
| | - Raquel C Gardner
- From the Sant Pau Memory Unit (D.A., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (D.A., A.L.), CIBERNED, Madrid, Spain; Department of Psychiatry (M.S.B.), Icahn School of Medicine at Mount Sinai, New York, NY; The Joseph Sagol Neuroscience (M.S.B., R.C.G.), Center Sheba Medical Center, Tel-Hashomer, Israel; and Department of Neurology (J.C.R.), Weill Institute for Neurosciences, UCSF Memory and Aging Center, San Francisco, CA
| | - Alberto Lleó
- From the Sant Pau Memory Unit (D.A., A.L.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB SANT PAU, Universitat Autònoma de Barcelona; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (D.A., A.L.), CIBERNED, Madrid, Spain; Department of Psychiatry (M.S.B.), Icahn School of Medicine at Mount Sinai, New York, NY; The Joseph Sagol Neuroscience (M.S.B., R.C.G.), Center Sheba Medical Center, Tel-Hashomer, Israel; and Department of Neurology (J.C.R.), Weill Institute for Neurosciences, UCSF Memory and Aging Center, San Francisco, CA.
| |
Collapse
|
21
|
Jeppsson A, Sandelius Å, Zettergren A, Kern S, Skoog I, Blennow K, Zetterberg H, Wikkelsø C, Hellström P, Tullberg M. Plasma and cerebrospinal fluid concentrations of neurofilament light protein correlate in patients with idiopathic normal pressure hydrocephalus. Fluids Barriers CNS 2023; 20:54. [PMID: 37415175 DOI: 10.1186/s12987-023-00455-y] [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: 03/14/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND Neurofilament light chain protein (NFL), a marker of neuronal axonal degeneration, is increased in cerebrospinal fluid (CSF) of patients with idiopathic normal pressure hydrocephalus (iNPH). Assays for analysis of NFL in plasma are now widely available but plasma NFL has not been reported in iNPH patients. Our aim was to examine plasma NFL in iNPH patients and to evaluate the correlation between plasma and CSF levels, and whether NFL levels are associated with clinical symptoms and outcome after shunt surgery. METHODS Fifty iNPH patients with median age 73 who had their symptoms assessed with the iNPH scale and plasma and CSF NFL sampled pre- and median 9 months post-operatively. CSF plasma was compared with 50 healthy controls (HC) matched for age and gender. Concentrations of NFL were determined in plasma using an in-house Simoa method and in CSF using a commercially available ELISA method. RESULTS Plasma NFL was elevated in patients with iNPH compared to HC (iNPH: 45 (30-64) pg/mL; HC: 33 (26-50) (median; Q1-Q3), p = 0.029). Plasma and CSF NFL concentrations correlated in iNPH patients both pre- and postoperatively (r = 0.67 and 0.72, p < 0.001). We found only weak correlations between plasma or CSF NFL and clinical symptoms and no associations with outcome. A postoperative NFL increase was seen in CSF but not in plasma. CONCLUSIONS Plasma NFL is increased in iNPH patients and concentrations correlate with CSF NFL implying that plasma NFL can be used to assess evidence of axonal degeneration in iNPH. This finding opens a window for plasma samples to be used in future studies of other biomarkers in iNPH. NFL is probably not a very useful marker of symptomatology or for prediction of outcome in iNPH.
Collapse
Affiliation(s)
- A Jeppsson
- Hydrocephalus Research Unit, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Blå Stråket 7, 41345, Gothenburg, Sweden
| | - Å Sandelius
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - A Zettergren
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - S Kern
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - I Skoog
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - K Blennow
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - H Zetterberg
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - C Wikkelsø
- Hydrocephalus Research Unit, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Blå Stråket 7, 41345, Gothenburg, Sweden
| | - P Hellström
- Hydrocephalus Research Unit, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Blå Stråket 7, 41345, Gothenburg, Sweden
| | - M Tullberg
- Hydrocephalus Research Unit, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Blå Stråket 7, 41345, Gothenburg, Sweden.
| |
Collapse
|
22
|
Asken BM, Ljubenkov PA, Staffaroni AM, Casaletto KB, Vandevrede L, Cobigo Y, Rojas-Rodriguez JC, Rankin KP, Kornak J, Heuer H, Shigenaga J, Appleby BS, Bozoki AC, Domoto-Reilly K, Ghoshal N, Huey E, Litvan I, Masdeu JC, Mendez MF, Pascual B, Pressman P, Tartaglia MC, Kremers W, Forsberg LK, Boeve BF, Boxer AL, Rosen HJ, Kramer JH. Plasma inflammation for predicting phenotypic conversion and clinical progression of autosomal dominant frontotemporal lobar degeneration. J Neurol Neurosurg Psychiatry 2023; 94:541-549. [PMID: 36977552 PMCID: PMC10313977 DOI: 10.1136/jnnp-2022-330866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/28/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND Measuring systemic inflammatory markers may improve clinical prognosis and help identify targetable pathways for treatment in patients with autosomal dominant forms of frontotemporal lobar degeneration (FTLD). METHODS We measured plasma concentrations of IL-6, TNFα and YKL-40 in pathogenic variant carriers (MAPT, C9orf72, GRN) and non-carrier family members enrolled in the ARTFL-LEFFTDS Longitudinal Frontotemporal Lobar Degeneration consortium. We evaluated associations between baseline plasma inflammation and rate of clinical and neuroimaging changes (linear mixed effects models with standardised (z) outcomes). We compared inflammation between asymptomatic carriers who remained clinically normal ('asymptomatic non-converters') and those who became symptomatic ('asymptomatic converters') using area under the curve analyses. Discrimination accuracy was compared with that of plasma neurofilament light chain (NfL). RESULTS We studied 394 participants (non-carriers=143, C9orf72=117, GRN=62, MAPT=72). In MAPT, higher TNFα was associated with faster functional decline (B=0.12 (0.02, 0.22), p=0.02) and temporal lobe atrophy. In C9orf72, higher TNFα was associated with faster functional decline (B=0.09 (0.03, 0.16), p=0.006) and cognitive decline (B=-0.16 (-0.22, -0.10), p<0.001), while higher IL-6 was associated with faster functional decline (B=0.12 (0.03, 0.21), p=0.01). TNFα was higher in asymptomatic converters than non-converters (β=0.29 (0.09, 0.48), p=0.004) and improved discriminability compared with plasma NfL alone (ΔR2=0.16, p=0.007; NfL: OR=1.4 (1.03, 1.9), p=0.03; TNFα: OR=7.7 (1.7, 31.7), p=0.007). CONCLUSIONS Systemic proinflammatory protein measurement, particularly TNFα, may improve clinical prognosis in autosomal dominant FTLD pathogenic variant carriers who are not yet exhibiting severe impairment. Integrating TNFα with markers of neuronal dysfunction like NfL could optimise detection of impending symptom conversion in asymptomatic pathogenic variant carriers and may help personalise therapeutic approaches.
Collapse
Affiliation(s)
- Breton M Asken
- Department of Clinical and Health Psychology, 1Florida Alzheimer's Disease Research Center, Fixel Institute for Neurological Diseases, University of Florida, Gainesville, Florida, USA
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, San Francisco, California, USA
| | - Peter A Ljubenkov
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, San Francisco, California, USA
| | - Adam M Staffaroni
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, San Francisco, California, USA
| | - Kaitlin B Casaletto
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, San Francisco, California, USA
| | - Lawren Vandevrede
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, San Francisco, California, USA
| | - Yann Cobigo
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, San Francisco, California, USA
| | - Julio C Rojas-Rodriguez
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, San Francisco, California, USA
| | - Katherine P Rankin
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, San Francisco, California, USA
| | - John Kornak
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
| | - Hilary Heuer
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, San Francisco, California, USA
| | - Judy Shigenaga
- Department of Medicine, Veterans Affairs Health Care System, San Francisco, California, USA
| | - Brian S Appleby
- Departments of Neurology, Psychiatry, and Pathology, Case Western Reserve, Cleveland, Ohio, USA
| | - Andrea C Bozoki
- Department of Neurology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kimiko Domoto-Reilly
- Department of Neurology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Nupur Ghoshal
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - Edward Huey
- Departments of Psychiatry and Neurology, Columbia University, New York, New York, USA
| | - Irene Litvan
- Department of Neurology, University of California, San Diego, La Jolla, California, USA
| | - Joseph C Masdeu
- Department of Neurology, Nantz National Alzheimer Center, Houston Methodist, Houston, Texas, USA
| | - Mario F Mendez
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Belen Pascual
- Department of Neurology, Nantz National Alzheimer Center, Houston Methodist, Houston, Texas, USA
| | - Peter Pressman
- Department of Neurology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario, Canada
- Canadian Sports Concussion Project, Toronto, Ontario, Canada
| | - Walter Kremers
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, Minnesota, USA
| | - Leah K Forsberg
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Brad F Boeve
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Adam L Boxer
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, San Francisco, California, USA
| | - Howie J Rosen
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, San Francisco, California, USA
| | - Joel H Kramer
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
23
|
Carlos AF, Josephs KA. The Role of Clinical Assessment in the Era of Biomarkers. Neurotherapeutics 2023; 20:1001-1018. [PMID: 37594658 PMCID: PMC10457273 DOI: 10.1007/s13311-023-01410-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2023] [Indexed: 08/19/2023] Open
Abstract
Hippocratic Medicine revolved around the three main principles of patient, disease, and physician and promoted the systematic observation of patients, rational reasoning, and interpretation of collected information. Although these remain the cardinal features of clinical assessment today, Medicine has evolved from a more physician-centered to a more patient-centered approach. Clinical assessment allows physicians to encounter, observe, evaluate, and connect with patients. This establishes the patient-physician relationship and facilitates a better understanding of the patient-disease relationship, as the ultimate goal is to diagnose, prognosticate, and treat. Biomarkers are at the core of the more disease-centered approach that is currently revolutionizing Medicine as they provide insight into the underlying disease pathomechanisms and biological changes. Genetic, biochemical, radiographic, and clinical biomarkers are currently used. Here, we define a seven-level theoretical construct for the utility of biomarkers in neurodegenerative diseases. Level 1-3 biomarkers are considered supportive of clinical assessment, capable of detecting susceptibility or risk factors, non-specific neurodegeneration or dysfunction, and/or changes at the individual level which help increase clinical diagnostic accuracy and confidence. Level 4-7 biomarkers have the potential to surpass the utility of clinical assessment through detection of early disease stages and prediction of underlying pathology. In neurodegenerative diseases, biomarkers can potentiate, but cannot substitute, clinical assessment. In this current era, aside from adding to the discovery, evaluation/validation, and implementation of more biomarkers, clinical assessment remains crucial to maintaining the personal, humanistic, and sociocultural aspects of patient care. We would argue that clinical assessment is a custom that should never go obsolete.
Collapse
Affiliation(s)
- Arenn F Carlos
- Department of Neurology, Mayo Clinic, 200 1st St. S.W., Rochester, MN, 55905, USA.
| | - Keith A Josephs
- Department of Neurology, Mayo Clinic, 200 1st St. S.W., Rochester, MN, 55905, USA
| |
Collapse
|
24
|
Huang SY, Chen SF, Cui M, Zhao M, Shen XN, Guo Y, Zhang YR, Zhang W, Wang HF, Huang YY, Cheng W, Zuo CT, Dong Q, Yu JT. Plasma Biomarkers and Positron Emission Tomography Tau Pathology in Progressive Supranuclear Palsy. Mov Disord 2023; 38:676-682. [PMID: 36781585 DOI: 10.1002/mds.29339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/12/2023] [Accepted: 01/19/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Development of disease-modifying therapeutic trials of progressive supranuclear palsy (PSP) urges the need for sensitive fluid biomarkers. OBJECTIVES The objectives of this study were to explore the utility of plasma biomarkers in the diagnosis, differential diagnosis, and assessment of disease severity, brain atrophy, and tau deposition in PSP. METHODS Plasma biomarkers were measured using a single-molecule array in a cohort composed of patients with PSP, Parkinson's disease (PD), multiple system atrophy with predominant parkinsonism (MSA-P), and healthy controls (HCs). RESULTS Plasma neurofilament light chain (NfL) outperformed other plasma makers (ie, glial fibrillary acidic protein [GFAP], phosphorylated-tau 181 [p-tau181], amyloid-β 1-40, amyloid-β 1-42) in identifying PSP from HC (area under the curve [AUC] = 0.904) and from MSA-P (AUC = 0.711). Plasma GFAP aided in distinguishing PSP from HC (AUC = 0.774) and from MSA-P (AUC = 0.832). It correlated with brainstem atrophy and higher regional tau accumulation. However, plasma p-tau181 neither helped in diagnosis nor was it associated with clinical or neuroimaging measures. CONCLUSIONS Plasma NfL and GFAP showed different values in differentiating PSP from HC or controls with other forms of neurodegenerative parkinsonism and detecting disease severity, brain atrophy, or tau deposition in PSP. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Shu-Yi Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Shu-Fen Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Mei Cui
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Meng Zhao
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xue-Ning Shen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Hui-Fu Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yu-Yuan Huang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Wei Cheng
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Chuan-Tao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, China
| |
Collapse
|
25
|
Casaletto KB, Kornack J, Paolillo EW, Rojas JC, VandeBunte A, Staffaroni AS, Lee S, Heuer H, Forsberg L, Ramos EM, Miller BL, Kramer JH, Yaffe K, Petrucelli L, Boxer A, Boeve B, Gendron TF, Rosen H. Association of Physical Activity With Neurofilament Light Chain Trajectories in Autosomal Dominant Frontotemporal Lobar Degeneration Variant Carriers. JAMA Neurol 2023; 80:82-90. [PMID: 36374516 PMCID: PMC9664369 DOI: 10.1001/jamaneurol.2022.4178] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022]
Abstract
Importance Physical activity is associated with cognitive health, even in autosomal dominant forms of dementia. Higher physical activity is associated with slowed cognitive and functional declines over time in adults carrying autosomal dominant variants for frontotemporal lobar degeneration (FTLD), but whether axonal degeneration is a potential neuroprotective target of physical activity in individuals with FTLD is unknown. Objective To examine the association between physical activity and longitudinal neurofilament light chain (NfL) trajectories in individuals with autosomal dominant forms of FTLD. Design, Setting, and Participants This cohort study included individuals from the ALLFTD Consortium, which recruited patients from sites in the US and Canada. Symptomatic and asymptomatic adults with pathogenic variants in one of 3 common genes associated with FTLD (GRN, C9orf72, or MAPT) who reported baseline physical activity levels and completed annual blood draws were assessed annually for up to 4 years. Genotype, clinical measures, and blood draws were collected between December 2014 and June 2019; data were analyzed from August 2021 to January 2022. Associations between reported baseline physical activity and longitudinal plasma NfL changes were assessed using generalized linear mixed-effects models adjusting for baseline age, sex, education, functional severity, and motor symptoms. Exposures Baseline physical activity levels reported via the Physical Activity Scale for the Elderly. To estimate effect sizes, marginal means were calculated at 3 levels of physical activity: 1 SD above the mean represented high physical activity, 0 SD represented average physical activity, and 1 SD below the mean represented low physical activity. Main Outcomes and Measures Annual plasma NfL concentrations were measured with single-molecule array technology. Results Of 160 included FTLD variant carriers, 84 (52.5%) were female, and the mean (SD) age was 50.7 (14.7) years. A total of 51 (31.8%) were symptomatic, and 77 carried the C9orf72 variant; 39, GRN variant; and 44, MAPT variant. Higher baseline physical activity was associated with slower NfL trajectories over time. On average, NfL increased 45.8% (95% CI, 22.5 to 73.7) over 4 years in variant carriers. Variant carriers with high physical activity demonstrated 14.0% (95% CI, -22.7 to -4.3) slower NfL increases compared with those with average physical activity and 30% (95% CI, -52.2 to -8.8) slower NfL increases compared with those with low physical activity. Within genotype, C9orf72 and MAPT carriers with high physical activity evidenced 18% to 21% (95% CI, -43.4 to -7.2) attenuation in NfL, while the association between physical activity and NfL trajectory was not statistically significant in GRN carriers. Activities associated with higher cardiorespiratory and cognitive demands (sports, housework, and yardwork) were most strongly correlated with slower NfL trajectories (vs walking and strength training). Conclusions and Relevance In this study, higher reported physical activity was associated with slower progression of an axonal degeneration marker in individuals with autosomal dominant FTLD. Physical activity may serve as a primary prevention target in FTLD.
Collapse
Affiliation(s)
- Kaitlin B. Casaletto
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - John Kornack
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Emily W. Paolillo
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Julio C. Rojas
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Anna VandeBunte
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Adam S. Staffaroni
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Shannon Lee
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Hilary Heuer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Leah Forsberg
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Eliana M. Ramos
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles
| | - Bruce L. Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Joel H. Kramer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Kristine Yaffe
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, Florida
| | - Adam Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Brad Boeve
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Tania F. Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, Florida
| | - Howard Rosen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| |
Collapse
|
26
|
Migeot JA, Duran-Aniotz CA, Signorelli CM, Piguet O, Ibáñez A. A predictive coding framework of allostatic-interoceptive overload in frontotemporal dementia. Trends Neurosci 2022; 45:838-853. [PMID: 36057473 PMCID: PMC11286203 DOI: 10.1016/j.tins.2022.08.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/27/2022] [Accepted: 08/09/2022] [Indexed: 10/31/2022]
Abstract
Recent allostatic-interoceptive explanations using predictive coding models propose that efficient regulation of the body's internal milieu is necessary to correctly anticipate environmental needs. We review this framework applied to understanding behavioral variant frontotemporal dementia (bvFTD) considering both allostatic overload and interoceptive deficits. First, we show how this framework could explain divergent deficits in bvFTD (cognitive impairments, behavioral maladjustment, brain atrophy, fronto-insular-temporal network atypicality, aberrant interoceptive electrophysiological activity, and autonomic disbalance). We develop a set of theory-driven predictions based on levels of allostatic interoception associated with bvFTD phenomenology and related physiopathological mechanisms. This approach may help further understand the disparate behavioral and physiopathological dysregulations of bvFTD, suggesting targeted interventions and strengthening clinical models of neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Joaquin A Migeot
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile; Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Claudia A Duran-Aniotz
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile; Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Camilo M Signorelli
- Department of Computer Science, University of Oxford, Oxford, UK; Physiology of Cognition, GIGA-CRC In Vivo Imaging, University of Liège, Liège, Belgium; Cognitive Neuroimaging Unit, INSERM, Saclay, France
| | - Olivier Piguet
- The University of Sydney, School of Psychology and Brain & Mind Centre, Sydney, Australia
| | - Agustín Ibáñez
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile; Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina; National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina; Global Brain Health Institute, University of California-San Francisco, San Francisco, CA, USA, and Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
27
|
Delaby C, Bousiges O, Bouvier D, Fillée C, Fourier A, Mondésert E, Nezry N, Omar S, Quadrio I, Rucheton B, Schraen-Maschke S, van Pesch V, Vicca S, Lehmann S, Bedel A. Neurofilaments contribution in clinic: state of the art. Front Aging Neurosci 2022; 14:1034684. [PMID: 36389064 PMCID: PMC9664201 DOI: 10.3389/fnagi.2022.1034684] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/10/2022] [Indexed: 07/26/2023] Open
Abstract
Neurological biomarkers are particularly valuable to clinicians as they can be used for diagnosis, prognosis, or response to treatment. This field of neurology has evolved considerably in recent years with the improvement of analytical methods, allowing the detection of biomarkers not only in cerebrospinal fluid (CSF) but also in less invasive fluids like blood. These advances greatly facilitate the repeated quantification of biomarkers, including at asymptomatic stages of the disease. Among the various informative biomarkers of neurological disorders, neurofilaments (NfL) have proven to be of particular interest in many contexts, such as neurodegenerative diseases, traumatic brain injury, multiple sclerosis, stroke, and cancer. Here we discuss these different pathologies and the potential value of NfL assay in the management of these patients, both for diagnosis and prognosis. We also describe the added value of NfL compared to other biomarkers currently used to monitor the diseases described in this review.
Collapse
Affiliation(s)
- Constance Delaby
- Université de Montpellier, IRMB, INM, INSERM, CHU de Montpellier, Laboratoire Biochimie-Protéomique clinique, Montpellier, France
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau—Biomedical Research Institute Sant Pau—Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Olivier Bousiges
- Laboratoire de biochimie et biologie moléculaire (LBBM)—Pôle de biologie Hôpital de Hautepierre—CHU de Strasbourg, CNRS, laboratoire ICube UMR 7357 et FMTS (Fédération de Médecine Translationnelle de Strasbourg), équipe IMIS, Strasbourg, France
| | - Damien Bouvier
- Service de Biochimie et Génétique Moléculaire, CHU de Clermont-Ferrand, Clermont-Ferrand, France
| | - Catherine Fillée
- Cliniques universitaires Saint-Luc UCLouvain, Service de Biochimie Médicale, Brussels, Belgium
| | - Anthony Fourier
- Biochimie et Biologie Moléculaire—LBMMS, Unité de diagnostic des pathologies dégénératives, Centre de Biologie et Pathologie Est, Groupement Hospitalier Est, Lyon, France
| | - Etienne Mondésert
- Université de Montpellier, IRMB, INM, INSERM, CHU de Montpellier, Laboratoire Biochimie-Protéomique clinique, Montpellier, France
| | - Nicolas Nezry
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, Lille, France
| | - Souheil Omar
- Laboratoire de biologie médicale de l’Institut de Neurologie de Tunis, Tunis, Tunisia
| | - Isabelle Quadrio
- Biochimie et Biologie Moléculaire—LBMMS, Unité de diagnostic des pathologies dégénératives, Centre de Biologie et Pathologie Est, Groupement Hospitalier Est, Lyon, France
| | - Benoit Rucheton
- Laboratoire de Biologie, Institut Bergonié, Bordeaux, France
| | - Susanna Schraen-Maschke
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, Lille, France
| | - Vincent van Pesch
- Cliniques universitaires Saint-Luc UCLouvain, Service de Neurologie, Brussels, Belgium
| | - Stéphanie Vicca
- Hôpital Necker-Enfants malades, Paris, Laboratoire de Biochimie générale, DMU BioPhyGen, AP-HP.Centre—Université de Paris, Paris, France
| | - Sylvain Lehmann
- Université de Montpellier, IRMB, INM, INSERM, CHU de Montpellier, Laboratoire Biochimie-Protéomique clinique, Montpellier, France
| | - Aurelie Bedel
- Service de Biochimie, CHU Pellegrin, Bordeaux, France
| |
Collapse
|
28
|
Morris HR. Blood based biomarkers for movement disorders. Acta Neurol Scand 2022; 146:353-361. [PMID: 36156206 PMCID: PMC9828103 DOI: 10.1111/ane.13700] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/12/2022] [Accepted: 08/19/2022] [Indexed: 01/12/2023]
Abstract
Movement disorders have been carefully clinically defined, based on clinico-pathological series; however there is often diagnostic and prognostic uncertainty, especially in early stage disease. Blood-based biomarkers for Alzheimer's disease (AD), particularly p-tau181 and p-tau217, may be useful in the movement disorder clinic, especially in identifying corticobasal syndrome due to AD pathology and in identifying Parkinson's disease (PD) patients at high risk for the future development of dementia. Serum or plasma neurofilament light (NfL) may be useful in separating Parkinson's plus syndromes (progressive supranuclear palsy-PSP, multiple system atrophy - MSA, and corticobasal syndrome-CBS) from PD. NfL is also a prognostic biomarker, in that the level of baseline or cross-sectional plasma/serum NfL is associated with a worse prognosis in PD and PSP. The development of protein aggregation assays in cerebrospinal fluid and multiplex assays which can measure 100 s-1000s of proteins in blood will provide new tools and insights for movement disorders for clinicians and researchers. The challenge is in efficiently integrating these tools into clinical practice and multi-modal approaches, where biomarkers are combined with clinical, genetic, and imaging data may guide the future use of these technologies.
Collapse
Affiliation(s)
- Huw R. Morris
- Department of Clinical and Movement Neurosciences, Queen Square Institute of NeurologyUniversity College LondonLondonUK
- UCL Movement Disorders CentreUniversity College LondonLondonUK
| |
Collapse
|
29
|
Jin J, Wei J, Feng Y, Cui Y, Zhou D, Yao M, He N, Mao Z, Gao F, Mao S. Plasma neurofilament light chain in Chinese children with later-onset spinal muscular atrophy. Clin Chem Lab Med 2022; 60:e237-e239. [PMID: 35953879 DOI: 10.1515/cclm-2022-0637] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/22/2022] [Indexed: 11/15/2022]
Affiliation(s)
- Jianing Jin
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, P.R. China
| | - Jia Wei
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, P.R. China
| | - Yijie Feng
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, P.R. China
| | - Yiqin Cui
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, P.R. China
| | - Dongming Zhou
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, P.R. China
| | - Mei Yao
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, P.R. China
| | - Ni He
- KingMed Center for Clinical Laboratory Co., Ltd., Hangzhou, P.R. China
| | - Zhifeng Mao
- KingMed Center for Clinical Laboratory Co., Ltd., Guangzhou, P.R. China
| | - Feng Gao
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, P.R. China
| | - Shanshan Mao
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, P.R. China
| |
Collapse
|
30
|
Parvizi T, König T, Wurm R, Silvaieh S, Altmann P, Klotz S, Rommer PS, Furtner J, Regelsberger G, Lehrner J, Traub-Weidinger T, Gelpi E, Stögmann E. Real-world applicability of glial fibrillary acidic protein and neurofilament light chain in Alzheimer's disease. Front Aging Neurosci 2022; 14:887498. [PMID: 36072480 PMCID: PMC9441692 DOI: 10.3389/fnagi.2022.887498] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Blood-based biomarkers may add a great benefit in detecting the earliest neuropathological changes in patients with Alzheimer's disease (AD). We examined the utility of neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) regarding clinical diagnosis and differentiation between amyloid positive and negative patients. To evaluate the practical application of these biomarkers in a routine clinical setting, we conducted this study in a heterogeneous memory-clinic population. Methods: We included 167 patients in this retrospective cross-sectional study, 123 patients with an objective cognitive decline [mild cognitive impairment (MCI) due to AD, n = 63, and AD-dementia, n = 60] and 44 age-matched healthy controls (HC). Cerebrospinal fluid (CSF) and plasma concentrations of NfL and GFAP were measured with single molecule array (SIMOA®) technology using the Neurology 2-Plex B kit from Quanterix. To assess the discriminatory potential of different biomarkers, age- and sex-adjusted receiver operating characteristic (ROC) curves were calculated and the area under the curve (AUC) of each model was compared. Results: We constructed a panel combining plasma NfL and GFAP with known AD risk factors (Combination panel: age+sex+APOE4+GFAP+NfL). With an AUC of 91.6% (95%CI = 0.85-0.98) for HC vs. AD and 81.7% (95%CI = 0.73-0.90) for HC vs. MCI as well as an AUC of 87.5% (95%CI = 0.73-0.96) in terms of predicting amyloid positivity, this panel showed a promising discriminatory power to differentiate these populations. Conclusion: The combination of plasma GFAP and NfL with well-established risk factors discerns amyloid positive from negative patients and could potentially be applied to identify patients who would benefit from a more invasive assessment of amyloid pathology. In the future, improved prediction of amyloid positivity with a noninvasive test may decrease the number and costs of a more invasive or expensive diagnostic approach.
Collapse
Affiliation(s)
- Tandis Parvizi
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Theresa König
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Raphael Wurm
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Sara Silvaieh
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Patrick Altmann
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Sigrid Klotz
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | | | - Julia Furtner
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Günther Regelsberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Johann Lehrner
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Tatjana Traub-Weidinger
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, University of Vienna, Vienna, Austria
| | - Ellen Gelpi
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
31
|
Ooi S, Patel SK, Eratne D, Kyndt C, Reidy N, Lewis C, Lee SC, Darby D, Brodtmann A. Plasma Neurofilament Light Chain and Clinical Diagnosis in Frontotemporal Dementia Syndromes. J Alzheimers Dis 2022; 89:1221-1231. [DOI: 10.3233/jad-220272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Frontotemporal dementia (FTD) syndromes, mimics, phenocopy (phFTD), and slowly progressive behavioral variant FTD (bvFTD) can be difficult to distinguish clinically. Biomarkers such as neurofilament light chain (NfL) may be helpful. Objective: To study plasma NfL levels in people with FTD syndromes and determine if plasma NfL can distinguish between FTD syndromes and phFTD. Methods: Plasma NfL levels were estimated using both Simoa ® Quanterix HD-X™ and SR-X™ machines grouped via final diagnosis after investigation and review. Results: Fifty participants were studied: bvFTD = 20, semantic variant FTD (svFTD) = 11, non-fluent variant FTD (nfvFTD) = 9, FTD with motor neuron disease (MND) = 4, phFTD = 2, slow progressors = 3, FTD mimic = 1, mean age 67.2 (SD 8.4) years. NfL levels were significantly higher in the FTD group compared to phenocopy group (p = 0.003). Median NfL (IQR) pg/mL was comparable in the FTD syndromes: bvFTD 41.10 (50.72), svFTD 44.38 (16.61), and nfvFTD 42.61 (22.93), highest in FTD with MND 79.67 (45.32) and lowest in both phFTD 13.99 (0.79) and slow progressors 17.97 (3.62). Conclusion: Plasma NfL appears to differentiate FTD syndromes and mimics. However, a lower NfL may predict a slower, but not necessarily lack of, neurodegeneration and therefore appears limited distinguishing slow progressors from FTD phenocopies. Larger numbers of patients from all clinical groups are required to strengthen diagnostic utility.
Collapse
Affiliation(s)
- Suyi Ooi
- Eastern Cognitive Disorders Clinic, Eastern Health, Box Hill, VIC, Australia
- Eastern Clinical Research Unit, Eastern Health Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, VIC, Australia
- Royal Melbourne Hospital, Department of Neurology, Parkville, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Sheila K Patel
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Dhamidhu Eratne
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Neuropsychiatry and Melbourne Neuropsychiatry Centre, Royal Melbourne Hospital and University of Melbourne, Parkville, VIC, Australia
| | - Christopher Kyndt
- Eastern Cognitive Disorders Clinic, Eastern Health, Box Hill, VIC, Australia
- Eastern Clinical Research Unit, Eastern Health Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, VIC, Australia
- Royal Melbourne Hospital, Department of Neurology, Parkville, VIC, Australia
| | - Natalie Reidy
- Eastern Cognitive Disorders Clinic, Eastern Health, Box Hill, VIC, Australia
| | - Courtney Lewis
- Eastern Cognitive Disorders Clinic, Eastern Health, Box Hill, VIC, Australia
- Neuropsychiatry and Melbourne Neuropsychiatry Centre, Royal Melbourne Hospital and University of Melbourne, Parkville, VIC, Australia
| | - Sarah C.M. Lee
- Eastern Clinical Research Unit, Eastern Health Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, VIC, Australia
- Calvary Health Care Bethlehem, Parkdale, VIC, Australia
| | - David Darby
- Eastern Cognitive Disorders Clinic, Eastern Health, Box Hill, VIC, Australia
- Eastern Clinical Research Unit, Eastern Health Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, VIC, Australia
- Royal Melbourne Hospital, Department of Neurology, Parkville, VIC, Australia
- Alfred Health, Department of Neurology, Prahran, Australia
| | - Amy Brodtmann
- Eastern Cognitive Disorders Clinic, Eastern Health, Box Hill, VIC, Australia
- Eastern Clinical Research Unit, Eastern Health Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, VIC, Australia
- Royal Melbourne Hospital, Department of Neurology, Parkville, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| |
Collapse
|
32
|
Garcia‐Moreno H, Prudencio M, Thomas‐Black G, Solanky N, Jansen‐West KR, Hanna AL‐Shaikh R, Heslegrave A, Zetterberg H, Santana MM, Pereira de Almeida L, Vasconcelos‐Ferreira A, Januário C, Infante J, Faber J, Klockgether T, Reetz K, Raposo M, Ferreira AF, Lima M, Schöls L, Synofzik M, Hübener‐Schmid J, Puschmann A, Gorcenco S, Wszolek ZK, Petrucelli L, Giunti P. Tau and neurofilament light-chain as fluid biomarkers in spinocerebellar ataxia type 3. Eur J Neurol 2022; 29:2439-2452. [PMID: 35478426 PMCID: PMC9543545 DOI: 10.1111/ene.15373] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/06/2022] [Accepted: 04/24/2022] [Indexed: 01/02/2023]
Abstract
BACKGROUND AND PURPOSE Clinical trials in spinocerebellar ataxia type 3 (SCA3) will require biomarkers for use as outcome measures. METHODS To evaluate total tau (t-tau), glial fibrillary acidic protein (GFAP), ubiquitin carboxy-terminal hydrolase L1 (UCHL1) and neurofilament light-chain (NfL) as fluid biomarkers in SCA3, ATXN3 mutation carriers (n = 143) and controls (n = 172) were clinically assessed, and the plasma concentrations of the four proteins were analysed on the Simoa HD-1 platform. Eleven ATXN3 mutation carrier cerebrospinal fluid samples were analysed for t-tau and phosphorylated tau (p-tau181 ). A transgenic SCA3 mouse model (MJDTg) was used to measure cerebellar t-tau levels. RESULTS Plasma t-tau levels were higher in mutation carriers below the age of 50 compared to controls, and the Inventory of Non-Ataxia Signs was associated with t-tau in ataxic patients (p = 0.004). Pre-ataxic carriers showed higher cerebrospinal fluid t-tau and p-tau181 concentrations compared to ataxic patients (p = 0.025 and p = 0.014, respectively). Cerebellar t-tau was elevated in MJDTg mice compared to wild-type (p = 0.033) only in the early stages of the disease. GFAP and UCHL1 did not show higher levels in mutation carriers compared to controls. Plasma NfL concentrations were higher in mutation carriers compared to controls, and differences were greater for younger carriers. The Scale for the Assessment and Rating of Ataxia was the strongest predictor of NfL in ataxic patients (p < 0.001). CONCLUSION Our results suggest that tau might be a marker of early disease stages in SCA3. NfL can discriminate mutation carriers from controls and is associated with different clinical variables. Longitudinal studies are required to confirm their potential role as biomarkers in clinical trials.
Collapse
Affiliation(s)
- Hector Garcia‐Moreno
- Ataxia CentreDepartment of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK,Department of NeurogeneticsNational Hospital for Neurology and NeurosurgeryUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Mercedes Prudencio
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA,Neuroscience Graduate ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Gilbert Thomas‐Black
- Ataxia CentreDepartment of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK,Department of NeurogeneticsNational Hospital for Neurology and NeurosurgeryUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Nita Solanky
- Ataxia CentreDepartment of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK,Department of NeurogeneticsNational Hospital for Neurology and NeurosurgeryUniversity College London Hospitals NHS Foundation TrustLondonUK
| | | | | | - Amanda Heslegrave
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK,UK Dementia Research Institute at UCLLondonUK
| | - Henrik Zetterberg
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK,UK Dementia Research Institute at UCLLondonUK,Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden,Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Magda M. Santana
- Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | | | | | | | - Jon Infante
- Neurology ServiceUniversity Hospital Marqués de Valdecilla‐IDIVALUniversity of CantabriaCentro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED)SantanderSpain
| | - Jennifer Faber
- Department of NeurologyUniversity Hospital BonnBonnGermany,German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Thomas Klockgether
- Department of NeurologyUniversity Hospital BonnBonnGermany,German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Kathrin Reetz
- Department of NeurologyRWTH Aachen UniversityAachenGermany,JARA‐BRAIN Institute Molecular Neuroscience and NeuroimagingForschungszentrum JülichRWTH Aachen UniversityAachenGermany
| | - Mafalda Raposo
- Faculdade de Ciências e TecnologiaUniversidade dos AçoresPonta DelgadaPortugal,Instituto de Biologia Molecular e Celular (IBMC)Instituto de Investigação e Inovação em Saúde (i3S)Universidade do PortoPortoPortugal
| | - Ana F. Ferreira
- Faculdade de Ciências e TecnologiaUniversidade dos AçoresPonta DelgadaPortugal,Instituto de Biologia Molecular e Celular (IBMC)Instituto de Investigação e Inovação em Saúde (i3S)Universidade do PortoPortoPortugal
| | - Manuela Lima
- Faculdade de Ciências e TecnologiaUniversidade dos AçoresPonta DelgadaPortugal,Instituto de Biologia Molecular e Celular (IBMC)Instituto de Investigação e Inovação em Saúde (i3S)Universidade do PortoPortoPortugal
| | - Ludger Schöls
- Department for Neurodegenerative DiseasesHertie‐Institute for Clinical Brain Research and Center for NeurologyUniversity of TübingenTübingenGermany,German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | - Matthis Synofzik
- Department for Neurodegenerative DiseasesHertie‐Institute for Clinical Brain Research and Center for NeurologyUniversity of TübingenTübingenGermany,German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | | | - Andreas Puschmann
- Lund University, Skåne University HospitalClinical Sciences, NeurologyLundSweden
| | - Sorina Gorcenco
- Lund University, Skåne University HospitalClinical Sciences, NeurologyLundSweden
| | | | - Leonard Petrucelli
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA,Neuroscience Graduate ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Paola Giunti
- Ataxia CentreDepartment of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK,Department of NeurogeneticsNational Hospital for Neurology and NeurosurgeryUniversity College London Hospitals NHS Foundation TrustLondonUK
| |
Collapse
|
33
|
Giacomucci G, Mazzeo S, Bagnoli S, Ingannato A, Leccese D, Berti V, Padiglioni S, Galdo G, Ferrari C, Sorbi S, Bessi V, Nacmias B. Plasma neurofilament light chain as a biomarker of Alzheimer's disease in Subjective Cognitive Decline and Mild Cognitive Impairment. J Neurol 2022; 269:4270-4280. [PMID: 35288777 PMCID: PMC9293849 DOI: 10.1007/s00415-022-11055-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Neurofilament light chain (NfL) is becoming increasingly notable in neurological diseases including AD, and it has been suggested as a new peripherical biomarker of neurodegeneration. We aimed to compare plasma NfL levels among Subjective Cognitive Decline (SCD), Mild Cognitive Impairment (MCI), and AD patients and to evaluate relationships between NfL and CSF biomarkers and neuropsychological scores. MATERIALS AND METHODS We enrolled 110 patients (34 SCD, 53 MCI, and 23 AD), who underwent clinical and neuropsychological evaluation, APOE genotyping, and plasma NfL analysis. Ninety-one patients underwent at least one amyloid burden biomarker (CSF and/or amyloid PET); 86 patients also underwent CSF phosphorylated-tau (p-tau) and total-tau (t-tau) measurement. Patients were classified as A + if they presented at least one positive amyloid biomarker or A- if not. RESULTS NfL levels were significantly increased in AD and MCI compared to SCD patients. These differences depend on A status, e.g., SCD A + had lower NfLs than MCI A + but comparable with MCI A-. Similarly, MCI A + had higher NfL levels than MCI A-, but comparable with AD. NfL levels correlated with p-tau in SCD, with all CSF biomarkers in MCI patients. No correlations were found in AD subgroup. In SCD, NfL levels were negatively correlated with memory test scores. CONCLUSIONS Plasma NfL levels might be a promising biomarker for neurodegeneration to discriminate cognitive decline due to AD from other conditions causing cognitive impairment in prodromal stages. Considering correlations with CSF p-tau and memory tests in SCD, NfL might be a useful peripheral biomarker also in preclinical phases of AD.
Collapse
Affiliation(s)
- Giulia Giacomucci
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Salvatore Mazzeo
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Deborah Leccese
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Valentina Berti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Sonia Padiglioni
- Regional Referral Centre for Relational Criticalities, Tuscany Region, Italy
- Unit Clinic of Organizations Careggi University Hospital, Florence, Italy
| | - Giulia Galdo
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Camilla Ferrari
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy.
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Azienda Ospedaliero-Universitaria Careggi. Largo Brambilla, 3, 50134, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| |
Collapse
|
34
|
Yao W, Zhang X, Zhao H, Xu Y, Bai F. Inflammation Disrupts Cognitive Integrity via Plasma Neurofilament Light Chain Coupling Brain Networks in Alzheimer’s Disease. J Alzheimers Dis 2022; 89:505-518. [PMID: 35871350 DOI: 10.3233/jad-220475] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Objective: Background: Plasma neurofilament light chain (NFL) is a recognized biomarker for Alzheimer’s disease (AD) and inflammation. Intrinsically organized default mode network core subsystem and frontoparietal network (FPN) and their interactions support complex cognitive function. The present study investigated the inflammatory effect on cognitive integrity via plasma NFL coupling internetwork interactions in AD. Objective: Objective: This study investigates the hypothesis that inflammation-related plasma NFL could affect the interactions of the core subsystem and FPN, which leads to the aggravation of the clinical symptoms of AD-spectrum patients. Objective: Methods: A total of 112 AD-spectrum participants underwent complete resting-state fMRI, neuropsychological tests, and plasma NFL at baseline (n = 112) and after approximately 17 months of follow-up (n = 112). The specific intersystem changes in the core subsystem and FPN were calculated and compared across groups. Then, the classifications of different AD-spectrum groups were analyzed using the association of plasma NFL and the changed intersystem interacting regions. Finally, mediation analysis was applied to investigate the significance of plasma NFL coupling networks on cognitive impairments in these subjects. Objective: Results: Discrimination of disease-related interactions of the core subsystem and FPN was found in AD-spectrum patients, which was the neural circuit fundamental to plasma NFL disrupting cognitive integrity. Furthermore, the clinical significance of plasma NFL coupling networks on AD identification and monitoring cognitive impairments were revealed in these subjects. Conclusion: The characteristic change in inflammation-related plasma NFL coupled with brain internetwork interactions could be used as a potential observation indicator in the progression of AD patients.
Collapse
Affiliation(s)
- Weina Yao
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiao Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hui Zhao
- Department of Neurology, Nanjing Drum Tower Hospital of The Affiliated Hospital of Nanjing University Medical School, and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital of The Affiliated Hospital of Nanjing University Medical School, and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | - Feng Bai
- Department of Neurology, Nanjing Drum Tower Hospital of The Affiliated Hospital of Nanjing University Medical School, and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | | |
Collapse
|
35
|
Olfati N, Shoeibi A, Litvan I. Clinical Spectrum of Tauopathies. Front Neurol 2022; 13:944806. [PMID: 35911892 PMCID: PMC9329580 DOI: 10.3389/fneur.2022.944806] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/20/2022] [Indexed: 11/20/2022] Open
Abstract
Tauopathies are both clinical and pathological heterogeneous disorders characterized by neuronal and/or glial accumulation of misfolded tau protein. It is now well understood that every pathologic tauopathy may present with various clinical phenotypes based on the primary site of involvement and the spread and distribution of the pathology in the nervous system making clinicopathological correlation more and more challenging. The clinical spectrum of tauopathies includes syndromes with a strong association with an underlying primary tauopathy, including Richardson syndrome (RS), corticobasal syndrome (CBS), non-fluent agrammatic primary progressive aphasia (nfaPPA)/apraxia of speech, pure akinesia with gait freezing (PAGF), and behavioral variant frontotemporal dementia (bvFTD), or weak association with an underlying primary tauopathy, including Parkinsonian syndrome, late-onset cerebellar ataxia, primary lateral sclerosis, semantic variant PPA (svPPA), and amnestic syndrome. Here, we discuss clinical syndromes associated with various primary tauopathies and their distinguishing clinical features and new biomarkers becoming available to improve in vivo diagnosis. Although the typical phenotypic clinical presentations lead us to suspect specific underlying pathologies, it is still challenging to differentiate pathology accurately based on clinical findings due to large phenotypic overlaps. Larger pathology-confirmed studies to validate the use of different biomarkers and prospective longitudinal cohorts evaluating detailed clinical, biofluid, and imaging protocols in subjects presenting with heterogenous phenotypes reflecting a variety of suspected underlying pathologies are fundamental for a better understanding of the clinicopathological correlations.
Collapse
Affiliation(s)
- Nahid Olfati
- Department of Neurology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- UC San Diego Department of Neurosciences, Parkinson and Other Movement Disorder Center, San Diego, CA, United States
| | - Ali Shoeibi
- Department of Neurology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Irene Litvan
- UC San Diego Department of Neurosciences, Parkinson and Other Movement Disorder Center, San Diego, CA, United States
| |
Collapse
|
36
|
Coughlin DG, Litvan I. Investigational therapeutics for the treatment of progressive supranuclear palsy. Expert Opin Investig Drugs 2022; 31:813-823. [DOI: 10.1080/13543784.2022.2087179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- David G Coughlin
- Department of Neurosciences, University of California San Diego, San Diego, 92093, CA
| | - Irene Litvan
- Department of Neurosciences, University of California San Diego, San Diego, 92093, CA
| |
Collapse
|
37
|
Ramaswamy P, Christopher R, Kumar Pal P, Debnath M, Yadav R. Plasma microRNAs as a Potential Biomarker for Identification of Progressive Supranuclear Palsy. Diagnostics (Basel) 2022; 12:diagnostics12051204. [PMID: 35626359 PMCID: PMC9139891 DOI: 10.3390/diagnostics12051204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 02/04/2023] Open
Abstract
Progressive supranuclear palsy (PSP) is the second most common Parkinsonian disorder with complex etiology. The underlying molecular mechanism of PSP pathogenesis remains unclear. The present study aims to find the feasibility of using plasma miRNAs as novel biomarkers. Plasma-focused qPCR panels were used for microRNA profiling and identified differentially expressed microRNAs in PSP compared to controls. The DIANA-miRPath v3.0 was used to perform KEGG pathway analysis. We then confirmed the expression of selected candidates by RT-qPCR and their clinical utility was assessed by ROC analysis. Profiling data revealed 28 differentially expressed microRNAs in PSP. Five overexpressed miRNAs were selected for further analysis. The KEGG pathway analysis revealed 48 high-risk pathways. The study revealed that as a single marker—miR-19b-3p, miR-33a-5p, miR-130b-3p, miR-136-3p, and miR-210-3p had a specificity of 64.71%, 82.35%, 68.75%, 82.35%, and 70.59% at sensitivity 77.78%, 77.78%, 66.67%, 73.33%, and 66.67%, respectively. The result suggests that circulating plasma miRNAs were altered in PSP compared to control. The findings of this study may provide potential biomarkers and pathways associated with PSP. Further large-scale validation studies are required to confirm the same.
Collapse
Affiliation(s)
- Palaniswamy Ramaswamy
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560029, India; (P.R.); (P.K.P.)
| | - Rita Christopher
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560029, India;
| | - Pramod Kumar Pal
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560029, India; (P.R.); (P.K.P.)
| | - Monojit Debnath
- Department of Human Genetics, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560029, India;
| | - Ravi Yadav
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560029, India; (P.R.); (P.K.P.)
- Correspondence:
| |
Collapse
|
38
|
Parmera JB, de Oliveira MCB, Rodrigues RD, Coutinho AM. Progressive supranuclear palsy and corticobasal degeneration: novel clinical concepts and advances in biomarkers. ARQUIVOS DE NEURO-PSIQUIATRIA 2022; 80:126-136. [PMID: 35976324 PMCID: PMC9491415 DOI: 10.1590/0004-282x-anp-2022-s134] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 04/29/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD) are sporadic adult-onset primary tauopathies clinically classified among the atypical parkinsonian syndromes. They are intrinsically related with regard to their clinical features, pathology, biochemistry, and genetic risk factors. OBJECTIVES This review highlights the current knowledge on PSP and CBD, focusing on evolving clinical concepts, new diagnostic criteria, and advances in biomarkers. METHODS We performed a non-systematic literature review through the PubMed database. The search was restricted to articles written in English, published from 1964 to date. RESULTS Clinicopathologic and in vivo biomarkers studies have broadened PSP and CBD clinical phenotypes. They are now recognized as a range of motor and behavioral syndromes associated with underlying 4R-tauopathy neuropathology. The Movement Disorders Society PSP diagnostic criteria included clinical variants apart from the classical description, increasing diagnostic sensitivity. Meanwhile, imaging biomarkers have explored the complexity of symptoms and pathological processes related to corticobasal syndrome and CBD. CONCLUSIONS In recent years, several prospective or clinicopathologic studies have assessed clinical, radiological, and fluid biomarkers that have helped us gain a better understanding of the complexity of the 4R-tauopathies, mainly PSP and CBD.
Collapse
Affiliation(s)
- Jacy Bezerra Parmera
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, São Paulo, SP, Brazil
| | | | - Roberta Diehl Rodrigues
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, São Paulo, SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Radiologia, Laboratório de Medicina Nuclear (LIM 44), São Paulo, SP, Brazil
| | - Artur Martins Coutinho
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Radiologia, Centro de Medicina Nuclear, Laboratório de Medicina Nuclear (LIM 43), São Paulo, SP, Brazil
| |
Collapse
|
39
|
Alawode DOT, Fox NC, Zetterberg H, Heslegrave AJ. Alzheimer’s Disease Biomarkers Revisited From the Amyloid Cascade Hypothesis Standpoint. Front Neurosci 2022; 16:837390. [PMID: 35573283 PMCID: PMC9091905 DOI: 10.3389/fnins.2022.837390] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/04/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disease worldwide. Amyloid beta (Aβ) is one of the proteins which aggregate in AD, and its key role in the disease pathogenesis is highlighted in the amyloid cascade hypothesis, which states that the deposition of Aβ in the brain parenchyma is a crucial initiating step in the future development of AD. The sensitivity of instruments used to measure proteins in blood and cerebrospinal fluid has significantly improved, such that Aβ can now successfully be measured in plasma. However, due to the peripheral production of Aβ, there is significant overlap between diagnostic groups. The presence of pathological Aβ within the AD brain has several effects on the cells and surrounding tissue. Therefore, there is a possibility that using markers of tissue responses to Aβ may reveal more information about Aβ pathology and pathogenesis than looking at plasma Aβ alone. In this manuscript, using the amyloid cascade hypothesis as a starting point, we will delve into how the effect of Aβ on the surrounding tissue can be monitored using biomarkers. In particular, we will consider whether glial fibrillary acidic protein, triggering receptor expressed on myeloid cells 2, phosphorylated tau, and neurofilament light chain could be used to phenotype and quantify the tissue response against Aβ pathology in AD.
Collapse
Affiliation(s)
- Deborah O. T. Alawode
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- *Correspondence: Deborah O. T. Alawode,
| | - Nick C. Fox
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Amanda J. Heslegrave
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Amanda J. Heslegrave,
| |
Collapse
|
40
|
Gendron TF, Heckman MG, White LJ, Veire AM, Pedraza O, Burch AR, Bozoki AC, Dickerson BC, Domoto-Reilly K, Foroud T, Forsberg LK, Galasko DR, Ghoshal N, Graff-Radford NR, Grossman M, Heuer HW, Huey ED, Hsiung GYR, Irwin DJ, Kaufer DI, Leger GC, Litvan I, Masdeu JC, Mendez MF, Onyike CU, Pascual B, Ritter A, Roberson ED, Rojas JC, Tartaglia MC, Wszolek ZK, Rosen H, Boeve BF, Boxer AL, Petrucelli L. Comprehensive cross-sectional and longitudinal analyses of plasma neurofilament light across FTD spectrum disorders. Cell Rep Med 2022; 3:100607. [PMID: 35492244 PMCID: PMC9044101 DOI: 10.1016/j.xcrm.2022.100607] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 12/13/2021] [Accepted: 03/23/2022] [Indexed: 12/30/2022]
Abstract
Frontotemporal dementia (FTD) therapy development is hamstrung by a lack of susceptibility, diagnostic, and prognostic biomarkers. Blood neurofilament light (NfL) shows promise as a biomarker, but studies have largely focused only on core FTD syndromes, often grouping patients with different diagnoses. To expedite the clinical translation of NfL, we avail ARTFL LEFFTDS Longitudinal Frontotemporal Lobar Degeneration (ALLFTD) study resources and conduct a comprehensive investigation of plasma NfL across FTD syndromes and in presymptomatic FTD mutation carriers. We find plasma NfL is elevated in all studied syndromes, including mild cases; increases in presymptomatic mutation carriers prior to phenoconversion; and associates with indicators of disease severity. By facilitating the identification of individuals at risk of phenoconversion, and the early diagnosis of FTD, plasma NfL can aid in participant selection for prevention or early treatment trials. Moreover, its prognostic utility would improve patient care, clinical trial efficiency, and treatment outcome estimations.
Collapse
Affiliation(s)
- Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Michael G Heckman
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Launia J White
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Austin M Veire
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Otto Pedraza
- Department of Psychiatry & Psychology, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Andrea C Bozoki
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bradford C Dickerson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Leah K Forsberg
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Douglas R Galasko
- Parkinson and Other Movement Disorder Center, Department of Neuroscience, University of California, San Diego, La Jolla, CA 92037, USA
| | - Nupur Ghoshal
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Murray Grossman
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hilary W Heuer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Edward D Huey
- Department of Psychiatry, Taub Institute, Columbia University, New York, NY 10032, USA; Department of Neurology, Taub Institute, Columbia University, New York, NY 10032, USA
| | - Ging-Yuek R Hsiung
- Centre for Brain Health, Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - David J Irwin
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Daniel I Kaufer
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Gabriel C Leger
- Parkinson and Other Movement Disorder Center, Department of Neuroscience, University of California, San Diego, La Jolla, CA 92037, USA
| | - Irene Litvan
- Parkinson and Other Movement Disorder Center, Department of Neuroscience, University of California, San Diego, La Jolla, CA 92037, USA
| | - Joseph C Masdeu
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Mario F Mendez
- Department of Neurology, University of California at Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Chiadi U Onyike
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Belen Pascual
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Aaron Ritter
- Neurological Institute, Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV 89106, USA
| | - Erik D Roberson
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Julio C Rojas
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Maria Carmela Tartaglia
- Krembil Brain Institute, Tanz Centre for Research in Neurodegenerative Diseases, Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | - Howard Rosen
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
41
|
Varesi A, Carrara A, Pires VG, Floris V, Pierella E, Savioli G, Prasad S, Esposito C, Ricevuti G, Chirumbolo S, Pascale A. Blood-Based Biomarkers for Alzheimer's Disease Diagnosis and Progression: An Overview. Cells 2022; 11:1367. [PMID: 35456047 PMCID: PMC9044750 DOI: 10.3390/cells11081367] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 01/10/2023] Open
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disease characterized by amyloid-β (Aβ) plaque deposition and neurofibrillary tangle accumulation in the brain. Although several studies have been conducted to unravel the complex and interconnected pathophysiology of AD, clinical trial failure rates have been high, and no disease-modifying therapies are presently available. Fluid biomarker discovery for AD is a rapidly expanding field of research aimed at anticipating disease diagnosis and following disease progression over time. Currently, Aβ1-42, phosphorylated tau, and total tau levels in the cerebrospinal fluid are the best-studied fluid biomarkers for AD, but the need for novel, cheap, less-invasive, easily detectable, and more-accessible markers has recently led to the search for new blood-based molecules. However, despite considerable research activity, a comprehensive and up-to-date overview of the main blood-based biomarker candidates is still lacking. In this narrative review, we discuss the role of proteins, lipids, metabolites, oxidative-stress-related molecules, and cytokines as possible disease biomarkers. Furthermore, we highlight the potential of the emerging miRNAs and long non-coding RNAs (lncRNAs) as diagnostic tools, and we briefly present the role of vitamins and gut-microbiome-related molecules as novel candidates for AD detection and monitoring, thus offering new insights into the diagnosis and progression of this devastating disease.
Collapse
Affiliation(s)
- Angelica Varesi
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
- Almo Collegio Borromeo, 27100 Pavia, Italy
| | - Adelaide Carrara
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (A.C.); (V.F.)
| | - Vitor Gomes Pires
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA;
| | - Valentina Floris
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (A.C.); (V.F.)
| | - Elisa Pierella
- School of Medicine, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK;
| | - Gabriele Savioli
- Emergency Department, IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Sakshi Prasad
- Faculty of Medicine, National Pirogov Memorial Medical University, 21018 Vinnytsya, Ukraine;
| | - Ciro Esposito
- Unit of Nephrology and Dialysis, ICS Maugeri, University of Pavia, 27100 Pavia, Italy;
| | - Giovanni Ricevuti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37129 Verona, Italy;
| | - Alessia Pascale
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
42
|
Reyes-Leiva D, Dols-Icardo O, Sirisi S, Cortés-Vicente E, Turon-Sans J, de Luna N, Blesa R, Belbin O, Montal V, Alcolea D, Fortea J, Lleó A, Rojas-García R, Illán-Gala I. Pathophysiological Underpinnings of Extra-Motor Neurodegeneration in Amyotrophic Lateral Sclerosis: New Insights From Biomarker Studies. Front Neurol 2022; 12:750543. [PMID: 35115992 PMCID: PMC8804092 DOI: 10.3389/fneur.2021.750543] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) lie at opposing ends of a clinical, genetic, and neuropathological continuum. In the last decade, it has become clear that cognitive and behavioral changes in patients with ALS are more frequent than previously recognized. Significantly, these non-motor features can impact the diagnosis, prognosis, and management of ALS. Partially overlapping neuropathological staging systems have been proposed to describe the distribution of TAR DNA-binding protein 43 (TDP-43) aggregates outside the corticospinal tract. However, the relationship between TDP-43 inclusions and neurodegeneration is not absolute and other pathophysiological processes, such as neuroinflammation (with a prominent role of microglia), cortical hyperexcitability, and synaptic dysfunction also play a central role in ALS pathophysiology. In the last decade, imaging and biofluid biomarker studies have revealed important insights into the pathophysiological underpinnings of extra-motor neurodegeneration in the ALS-FTLD continuum. In this review, we first summarize the clinical and pathophysiological correlates of extra-motor neurodegeneration in ALS. Next, we discuss the diagnostic and prognostic value of biomarkers in ALS and their potential to characterize extra-motor neurodegeneration. Finally, we debate about how biomarkers could improve the diagnosis and classification of ALS. Emerging imaging biomarkers of extra-motor neurodegeneration that enable the monitoring of disease progression are particularly promising. In addition, a growing arsenal of biofluid biomarkers linked to neurodegeneration and neuroinflammation are improving the diagnostic accuracy and identification of patients with a faster progression rate. The development and validation of biomarkers that detect the pathological aggregates of TDP-43 in vivo are notably expected to further elucidate the pathophysiological underpinnings of extra-motor neurodegeneration in ALS. Novel biomarkers tracking the different aspects of ALS pathophysiology are paving the way to precision medicine approaches in the ALS-FTLD continuum. These are essential steps to improve the diagnosis and staging of ALS and the design of clinical trials testing novel disease-modifying treatments.
Collapse
Affiliation(s)
- David Reyes-Leiva
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Oriol Dols-Icardo
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Sonia Sirisi
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Elena Cortés-Vicente
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Janina Turon-Sans
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Noemi de Luna
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Olivia Belbin
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Ricard Rojas-García
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Ignacio Illán-Gala
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
- *Correspondence: Ignacio Illán-Gala
| |
Collapse
|
43
|
Sihag S, Naze S, Taghdiri F, Gumus M, Tator C, Green R, Colella B, Blennow K, Zetterberg H, Dominguez LG, Wennberg R, Mikulis DJ, Tartaglia MC, Kozloski JR. Functional brain activity constrained by structural connectivity reveals cohort-specific features for serum neurofilament light chain. COMMUNICATIONS MEDICINE 2022; 2:8. [PMID: 35603281 PMCID: PMC9053240 DOI: 10.1038/s43856-021-00065-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/07/2021] [Indexed: 11/30/2022] Open
Abstract
Background Neuro-axonal brain damage releases neurofilament light chain (NfL) proteins, which enter the blood. Serum NfL has recently emerged as a promising biomarker for grading axonal damage, monitoring treatment responses, and prognosis in neurological diseases. Importantly, serum NfL levels also increase with aging, and the interpretation of serum NfL levels in neurological diseases is incomplete due to lack of a reliable model for age-related variation in serum NfL levels in healthy subjects. Methods Graph signal processing (GSP) provides analytical tools, such as graph Fourier transform (GFT), to produce measures from functional dynamics of brain activity constrained by white matter anatomy. Here, we leveraged a set of features using GFT that quantified the coupling between blood oxygen level dependent signals and structural connectome to investigate their associations with serum NfL levels collected from healthy subjects and former athletes with history of concussions. Results Here we show that GSP feature from isthmus cingulate in the right hemisphere (r-iCg) is strongly linked with serum NfL in healthy controls. In contrast, GSP features from temporal lobe and lingual areas in the left hemisphere and posterior cingulate in the right hemisphere are the most associated with serum NfL in former athletes. Additional analysis reveals that the GSP feature from r-iCg is associated with behavioral and structural measures that predict aggressive behavior in healthy controls and former athletes. Conclusions Our results suggest that GSP-derived brain features may be included in models of baseline variance when evaluating NfL as a biomarker of neurological diseases and studying their impact on personality traits. Neurofilament light chain (NfL) is a marker released into the blood as a result of central nervous system damage or neurodegeneration. However, we know little about how NfL levels relate to brain structure and activity. Here, we use imaging data and advanced statistical methods to look at the relationship between brain activity and structure in healthy people and former athletes with a history of multiple concussions, and determine whether these can predict NfL levels in the blood. We find the relationship between brain activity and structure and NfL levels is different between the two groups. Our findings help us to understand how brain injury might impact NfL levels and their relationship with brain activity, and could guide how NfL and imaging data are used as tools in research and in the clinic. Sihag et al. analyse brain imaging data, circulating neurofilament light chain levels and personality scores in a cohort of former athletes with a history of concussions. The authors use graph signal processing to identify brain structural and connectivity features associated with neurofilament levels and with aggressive behaviour.
Collapse
|
44
|
Acute peri‐operative neurocognitive disorders: a narrative review. Anaesthesia 2022; 77 Suppl 1:34-42. [DOI: 10.1111/anae.15613] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2021] [Indexed: 12/18/2022]
|
45
|
Benussi A, Ashton NJ, Karikari TK, Alberici A, Saraceno C, Ghidoni R, Benussi L, Zetterberg H, Blennow K, Borroni B. Prodromal frontotemporal dementia: clinical features and predictors of progression. Alzheimers Res Ther 2021; 13:188. [PMID: 34782010 PMCID: PMC8594126 DOI: 10.1186/s13195-021-00932-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 11/04/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND The prodromal phase of frontotemporal dementia (FTD) is still not well characterized, and conversion rates to dementia and predictors of progression at 1-year follow-up are currently unknown. METHODS In this retrospective study, disease severity was assessed using the global CDR plus NACC FTLD. Prodromal FTD was defined to reflect mild cognitive or behavioural impairment with relatively preserved functional independence (global CDR plus NACC = 0.5) as well as mild, moderate and severe dementia (classified as global CDR plus NACC = 1, 2, 3, respectively). Disease progression at 1-year follow-up and serum NfL measurements were acquired in a subgroup of patients. RESULTS Of 563 participants, 138 were classified as prodromal FTD, 130 as mild, 175 as moderate and 120 as severe FTD. In the prodromal and mild phases, we observed an early increase in serum NfL levels followed by behavioural disturbances and deficits in executive functions. Negative symptoms, such as apathy, inflexibility and loss of insight, predominated in the prodromal phase. Serum NfL levels were significantly increased in the prodromal phase compared with healthy controls (average difference 14.5, 95% CI 2.9 to 26.1 pg/mL), but lower than in patients with mild FTD (average difference -15.5, 95% CI -28.4 to -2.7 pg/mL). At 1-year follow-up, 51.2% of patients in the prodromal phase had converted to dementia. Serum NfL measurements at baseline were the strongest predictors of disease progression at 1-year follow-up (OR 1.07, 95% CI 1.03 to 1.11, p < 0.001). CONCLUSIONS Prodromal FTD is a mutable stage with high rate of progression to fully symptomatic disease at 1-year follow-up. High serum NfL levels may support prodromal FTD diagnosis and represent a helpful marker to assess disease progression.
Collapse
Affiliation(s)
- Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, P.le Spedali Civili 1, 25123, Brescia, Italy
| | - Nicholas J Ashton
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Mölndal, Sweden
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Thomas K Karikari
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
| | - Antonella Alberici
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, P.le Spedali Civili 1, 25123, Brescia, Italy
| | - Claudia Saraceno
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, P.le Spedali Civili 1, 25123, Brescia, Italy.
| |
Collapse
|
46
|
Brodtmann A, Werden E, Khlif MS, Bird LJ, Egorova N, Veldsman M, Pardoe H, Jackson G, Bradshaw J, Darby D, Cumming T, Churilov L, Donnan G. Neurodegeneration Over 3 Years Following Ischaemic Stroke: Findings From the Cognition and Neocortical Volume After Stroke Study. Front Neurol 2021; 12:754204. [PMID: 34744989 PMCID: PMC8570373 DOI: 10.3389/fneur.2021.754204] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/27/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Stroke survivors are at high risk of dementia, associated with increasing age and vascular burden and with pre-existing cognitive impairment, older age. Brain atrophy patterns are recognised as signatures of neurodegenerative conditions, but the natural history of brain atrophy after stroke remains poorly described. We sought to determine whether stroke survivors who were cognitively normal at time of stroke had greater total brain (TBV) and hippocampal volume (HV) loss over 3 years than controls. We examined whether stroke survivors who were cognitively impaired (CI) at 3 months following their stroke had greater brain volume loss than cognitively normal (CN) stroke participants over the next 3 years. Methods: Cognition And Neocortical Volume After Stroke (CANVAS) study is a multi-centre cohort study of first-ever or recurrent adult ischaemic stroke participants compared to age- and sex-matched community controls. Participants were followed with MRI and cognitive assessments over 3 years and were free of a history of cognitive impairment or decline at inclusion. Our primary outcome measure was TBV change between 3 months and 3 years; secondary outcomes were TBV and HV change comparing CI and CN participants. We investigated associations between group status and brain volume change using a baseline-volume adjusted linear regression model with robust standard error. Results: Ninety-three stroke (26 women, 66.7 ± 12 years) and 39 control participants (15 women, 68.7 ± 7 years) were available at 3 years. TBV loss in stroke patients was greater than controls: stroke mean (M) = 20.3 cm3 ± SD 14.8 cm3; controls M = 14.2 cm3 ± SD 13.2 cm3; [adjusted mean difference 7.88 95%CI (2.84, 12.91) p-value = 0.002]. TBV decline was greater in those stroke participants who were cognitively impaired (M = 30.7 cm3; SD = 14.2 cm3) at 3 months (M = 19.6 cm3; SD = 13.8 cm3); [adjusted mean difference 10.42; 95%CI (3.04, 17.80), p-value = 0.006]. No statistically significant differences in HV change were observed. Conclusions: Ischaemic stroke survivors exhibit greater neurodegeneration compared to stroke-free controls. Brain atrophy is greater in stroke participants who were cognitively impaired early after their stroke. Early cognitive impairment was associated greater subsequent atrophy, reflecting the combined impacts of stroke and vascular brain burden. Atrophy rates could serve as a useful biomarker for trials testing interventions to reduce post-stroke secondary neurodegeneration. Clinical Trail Registration:http://www.clinicaltrials.gov, identifier: NCT02205424.
Collapse
Affiliation(s)
- Amy Brodtmann
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia.,Melbourne Dementia Research Centre, Florey Institute and University of Melbourne, Parkville, VIC, Australia.,Melbourne Medical School, University of Melbourne, Melbourne, VIC, Australia
| | - Emilio Werden
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Mohamed Salah Khlif
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Laura J Bird
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Natalia Egorova
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia.,Melbourne Dementia Research Centre, Florey Institute and University of Melbourne, Parkville, VIC, Australia.,Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Michele Veldsman
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia.,Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Heath Pardoe
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Graeme Jackson
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia.,Melbourne Medical School, University of Melbourne, Melbourne, VIC, Australia
| | - Jennifer Bradshaw
- Department of Clinical Neuropsychology, Austin Health, Heidelberg, VIC, Australia
| | - David Darby
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia.,Melbourne Dementia Research Centre, Florey Institute and University of Melbourne, Parkville, VIC, Australia
| | - Toby Cumming
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Leonid Churilov
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia.,Melbourne Medical School, University of Melbourne, Melbourne, VIC, Australia
| | - Geoffrey Donnan
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia.,Melbourne Medical School, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
47
|
[Neuroprotective treatment of tauopathies]. DER NERVENARZT 2021; 92:1227-1238. [PMID: 34652482 DOI: 10.1007/s00115-021-01210-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
Tau pathology is now considered to be the main cause of a wide spectrum of neurodegenerative diseases, which are collectively referred to as tauopathies. These include primary tauopathies, in which tau plays the main role in the pathogenesis as well as secondary tauopathies, such as Alzheimer's disease, in which amyloid beta also plays a substantial role in the disease process in addition to the tau pathology. Primary tauopathies include progressive supranuclear palsy, corticobasal degeneration, Pick's disease and rare hereditary tauopathies, which are referred to as frontotemporal lobar degeneration with microtubule-associated protein tau (MAPT) mutation. Tauopathies differ from each other pathologically by the affected brain regions and cell types as well as by the biochemical characteristics of the aggregated tau protein. Various tau-centered neuroprotective treatment approaches are currently in preclinical and clinical development. They target different mechanisms, including the reduction of tau expression, inhibition of tau aggregation, dissolution of tau aggregates, improvement of cellular mechanisms to eliminate toxic tau species, stabilization of microtubules and prevention of intercellular tau spreading. This review article gives an overview of tauopathies and the current concepts for the development of disease-modifying treatment.
Collapse
|
48
|
Stamelou M, Respondek G, Giagkou N, Whitwell JL, Kovacs GG, Höglinger GU. Evolving concepts in progressive supranuclear palsy and other 4-repeat tauopathies. Nat Rev Neurol 2021; 17:601-620. [PMID: 34426686 DOI: 10.1038/s41582-021-00541-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Tauopathies are classified according to whether tau deposits predominantly contain tau isoforms with three or four repeats of the microtubule-binding domain. Those in which four-repeat (4R) tau predominates are known as 4R-tauopathies, and include progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease, globular glial tauopathies and conditions associated with specific MAPT mutations. In these diseases, 4R-tau deposits are found in various cell types and anatomical regions of the brain and the conditions share pathological, pathophysiological and clinical characteristics. Despite being considered 'prototype' tauopathies and, therefore, ideal for studying neuroprotective agents, 4R-tauopathies are still severe and untreatable diseases for which no validated biomarkers exist. However, advances in research have addressed the issues of phenotypic overlap, early clinical diagnosis, pathophysiology and identification of biomarkers, setting a road map towards development of treatments. New clinical criteria have been developed and large cohorts with early disease are being followed up in prospective studies. New clinical trial readouts are emerging and biomarker research is focused on molecular pathways that have been identified. Lessons learned from failed trials of neuroprotective drugs are being used to design new trials. In this Review, we present an overview of the latest research in 4R-tauopathies, with a focus on progressive supranuclear palsy, and discuss how current evidence dictates ongoing and future research goals.
Collapse
Affiliation(s)
- Maria Stamelou
- Parkinson's Disease and Movement Disorders Dept, HYGEIA Hospital, Athens, Greece. .,European University of Cyprus, Nicosia, Cyprus. .,Philipps University, Marburg, Germany.
| | - Gesine Respondek
- Department of Neurology, Hanover Medical School, Hanover, Germany
| | - Nikolaos Giagkou
- Parkinson's Disease and Movement Disorders Dept, HYGEIA Hospital, Athens, Greece
| | | | - Gabor G Kovacs
- Department of Laboratory Medicine and Pathobiology and Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Toronto, Ontario, Canada.,Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
| | - Günter U Höglinger
- Department of Neurology, Hanover Medical School, Hanover, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| |
Collapse
|
49
|
A novel orally active HDAC6 inhibitor T-518 shows a therapeutic potential for Alzheimer's disease and tauopathy in mice. Sci Rep 2021; 11:15423. [PMID: 34326423 PMCID: PMC8322070 DOI: 10.1038/s41598-021-94923-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/14/2021] [Indexed: 11/14/2022] Open
Abstract
Accumulation of tau protein is a key pathology of age-related neurodegenerative diseases such as Alzheimer's disease and progressive supranuclear palsy. Those diseases are collectively termed tauopathies. Tau pathology is associated with axonal degeneration because tau binds to microtubules (MTs), a component of axon and regulates their stability. The acetylation state of MTs contributes to stability and histone deacetylase 6 (HDAC6) is a major regulator of MT acetylation status, suggesting that pharmacological HDAC6 inhibition could improve axonal function and may slow the progression of tauopathy. Here we characterize N-[(1R,2R)-2-{3-[5-(difluoromethyl)-1,3,4-oxadiazol-2-yl]-5-oxo-5H,6H,7H-pyrrolo[3,4-b]pyridin-6-yl}cyclohexyl]-2,2,3,3,3-pentafluoropropanamide (T-518), a novel, potent, highly selective HDAC6 inhibitor with clinically favorable pharmacodynamics. T-518 shows potent inhibitory activity against HDAC6 and superior selectivity over other HDACs compared with the known HDAC6 inhibitors in the enzyme and cellular assays. T-518 showed brain penetration in an oral dose and blocked HDAC6-dependent tubulin deacetylation at Lys40 in mouse hippocampus. A 2-week treatment restored impaired axonal transport and novel object recognition in the P301S tau Tg mouse, tauopathy model, while a 3-month treatment also decreased RIPA-insoluble tau accumulation. Pharmaceutical inhibition of HDAC6 is a potential therapeutic strategy for tauopathy, and T-518 is a particularly promising drug candidate.
Collapse
|
50
|
Simrén J, Leuzy A, Karikari TK, Hye A, Benedet AL, Lantero‐Rodriguez J, Mattsson‐Carlgren N, Schöll M, Mecocci P, Vellas B, Tsolaki M, Kloszewska I, Soininen H, Lovestone S, Aarsland D, Hansson O, Rosa‐Neto P, Westman E, Blennow K, Zetterberg H, Ashton NJ. The diagnostic and prognostic capabilities of plasma biomarkers in Alzheimer's disease. Alzheimers Dement 2021; 17:1145-1156. [PMID: 33491853 PMCID: PMC8359457 DOI: 10.1002/alz.12283] [Citation(s) in RCA: 181] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/28/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION This study investigated the diagnostic and disease-monitoring potential of plasma biomarkers in mild cognitive impairment (MCI) and Alzheimer's disease (AD) dementia and cognitively unimpaired (CU) individuals. METHODS Plasma was analyzed using Simoa assays from 99 CU, 107 MCI, and 103 AD dementia participants. RESULTS Phosphorylated-tau181 (P-tau181), neurofilament light, amyloid-β (Aβ42/40), Total-tau and Glial fibrillary acidic protein were altered in AD dementia but P-tau181 significantly outperformed all biomarkers in differentiating AD dementia from CU (area under the curve [AUC] = 0.91). P-tau181 was increased in MCI converters compared to non-converters. Higher P-tau181 was associated with steeper cognitive decline and gray matter loss in temporal regions. Longitudinal change of P-tau181 was strongly associated with gray matter loss in the full sample and with Aβ measures in CU individuals. DISCUSSION P-tau181 detected AD at MCI and dementia stages and was strongly associated with cognitive decline and gray matter loss. These findings highlight the potential value of plasma P-tau181 as a non-invasive and cost-effective diagnostic and prognostic biomarker in AD.
Collapse
Affiliation(s)
- Joel Simrén
- Department of Psychiatry and NeurochemistryThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGothenburgSweden
| | - Antoine Leuzy
- Clinical Memory Research UnitLund UniversityMalmöSweden
| | - Thomas K. Karikari
- Department of Psychiatry and NeurochemistryThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Abdul Hye
- Department of Old Age PsychiatryInstitute of Psychiatry, Psychology & Neuroscience, King's College LondonLondonUK
| | | | - Juan Lantero‐Rodriguez
- Department of Psychiatry and NeurochemistryThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Niklas Mattsson‐Carlgren
- Clinical Memory Research UnitLund UniversityMalmöSweden
- Department of NeurologySkåne University HospitalLundSweden
- Wallenberg Centre for Molecular MedicineLund UniversityLundSweden
| | - Michael Schöll
- Department of Psychiatry and NeurochemistryThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - Patrizia Mecocci
- Department of MedicineInstitute of Gerontology and GeriatricsUniversity of PerugiaPerugiaItaly
| | | | - Magda Tsolaki
- Aristotle University of ThessalonikiThessalonikiGreece
| | | | - Hilkka Soininen
- Institute of Clinical MedicineUniversity of Eastern FinlandKuopioFinland
| | | | - Dag Aarsland
- Department of Old Age PsychiatryInstitute of Psychiatry, Psychology & Neuroscience, King's College LondonLondonUK
- Centre for Age‐Related MedicineStavanger University HospitalStavangerNorway
| | | | - Oskar Hansson
- Clinical Memory Research UnitLund UniversityMalmöSweden
- Memory ClinicSkåne University HospitalMalmöSweden
| | - Pedro Rosa‐Neto
- Translational Neuroimaging LaboratoryMcGill UniversityMontréalCanada
| | - Eric Westman
- Division of Clinical GeriatricsDepartment of NeurobiologyCare Sciences and SocietyKarolinska InstituteStockholmSweden
- Department of NeuroimagingCentre for Neuroimaging SciencesPsychology and NeuroscienceKing's College LondonInstitute of PsychiatryLondonUK
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGothenburgSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGothenburgSweden
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Department of Old Age PsychiatryInstitute of Psychiatry, Psychology & Neuroscience, King's College LondonLondonUK
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
| |
Collapse
|