1
|
Galasko D, Farlow MR, Lucey BP, Honig LS, Elbert D, Bateman R, Momper J, Thomas RG, Rissman RA, Pa J, Aslanyan V, Balasubramanian A, West T, Maccecchini M, Feldman HH. A multicenter, randomized, double-blind, placebo-controlled ascending dose study to evaluate the safety, tolerability, pharmacokinetics (PK) and pharmacodynamic (PD) effects of Posiphen in subjects with early Alzheimer's Disease. Alzheimers Res Ther 2024; 16:151. [PMID: 38970127 PMCID: PMC11225352 DOI: 10.1186/s13195-024-01490-z] [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: 03/04/2024] [Accepted: 05/29/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Amyloid beta protein (Aβ) is a treatment target in Alzheimer's Disease (AD). Lowering production of its parent protein, APP, has benefits in preclinical models. Posiphen, an orally administered small molecule, binds to an iron-responsive element in APP mRNA and decreases translation of APP and Aβ. To augment human data for Posiphen, we evaluated safety, tolerability and pharmacokinetic and pharmacodynamic (PD) effects on Aβ metabolism using Stable Isotope Labeling Kinetic (SILK) analysis. METHODS Double-blind phase 1b randomized ascending dose clinical trial, at five sites, under an IRB-approved protocol. Participants with mild cognitive impairment or mild AD (Early AD) confirmed by low CSF Aβ42/40 were randomized (within each dose arm) to Posiphen or placebo. Pretreatment assessment included lumbar puncture for CSF. Participants took Posiphen or placebo for 21-23 days, then underwent CSF catheter placement, intravenous infusion of 13C6-leucine, and CSF sampling for 36 h. Safety and tolerability were assessed through participant reports, EKG and laboratory tests. CSF SILK analysis measured Aβ40, 38 and 42 with immunoprecipitation-mass spectrometry. Baseline and day 21 CSF APP, Aβ and other biomarkers were measured with immunoassays. The Mini-Mental State Exam and ADAS-cog12 were given at baseline and day 21. RESULTS From June 2017 to December 2021, 19 participants were enrolled, randomized within dose cohorts (5 active: 3 placebo) of 60 mg once/day and 60 mg twice/day; 1 participant was enrolled and completed 60 mg three times/day. 10 active drug and 5 placebo participants completed all study procedures. Posiphen was safe and well-tolerated. 8 participants had headaches related to CSF catheterization; 5 needed blood patches. Prespecified SILK analyses of Fractional Synthesis Rate (FSR) for CSF Aβ40 showed no significant overall or dose-dependent effects of Posiphen vs. placebo. Comprehensive multiparameter modeling of APP kinetics supported dose-dependent lowering of APP production by Posiphen. Cognitive measures and CSF biomarkers did not change significantly from baseline to 21 days in Posiphen vs. placebo groups. CONCLUSIONS Posiphen was safe and well-tolerated in Early AD. A multicenter SILK study was feasible. Findings are limited by small sample size but provide additional supportive safety and PK data. Comprehensive modeling of biomarker dynamics using SILK data may reveal subtle drug effects. TRIAL REGISTRATION NCT02925650 on clinicaltrials.gov (registered on 10-24-2016).
Collapse
Affiliation(s)
- Douglas Galasko
- Department of Neurosciences, UC San Diego, 9444 Medical Center Drive, Suite 1-100, La Jolla, San Diego, CA, 9209, USA.
| | | | | | | | | | | | - Jeremiah Momper
- Department of Neurosciences, UC San Diego, 9444 Medical Center Drive, Suite 1-100, La Jolla, San Diego, CA, 9209, USA
| | - Ronald G Thomas
- Department of Neurosciences, UC San Diego, 9444 Medical Center Drive, Suite 1-100, La Jolla, San Diego, CA, 9209, USA
| | - Robert A Rissman
- Department of Neurosciences, UC San Diego, 9444 Medical Center Drive, Suite 1-100, La Jolla, San Diego, CA, 9209, USA
| | - Judy Pa
- Department of Neurosciences, UC San Diego, 9444 Medical Center Drive, Suite 1-100, La Jolla, San Diego, CA, 9209, USA
| | | | - Archana Balasubramanian
- Department of Neurosciences, UC San Diego, 9444 Medical Center Drive, Suite 1-100, La Jolla, San Diego, CA, 9209, USA
| | - Tim West
- C2N Diagnostics, St Louis, MO, USA
| | | | - Howard H Feldman
- Department of Neurosciences, UC San Diego, 9444 Medical Center Drive, Suite 1-100, La Jolla, San Diego, CA, 9209, USA
| |
Collapse
|
2
|
Giroux P, Vialaret J, Kindermans J, Gabelle A, Bauchet L, Hirtz C, Lehmann S, Colinge J. Modeling the Simultaneous Dynamics of Proteins in Blood Plasma and the Cerebrospinal Fluid in Human In Vivo. J Proteome Res 2024; 23:2408-2418. [PMID: 38857467 PMCID: PMC11232576 DOI: 10.1021/acs.jproteome.4c00059] [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: 02/11/2024] [Revised: 04/12/2024] [Accepted: 05/30/2024] [Indexed: 06/12/2024]
Abstract
The analysis of protein dynamics or turnover in patients has the potential to reveal altered protein recycling, such as in Alzheimer's disease, and to provide informative data regarding drug efficacy or certain biological processes. The observed protein dynamics in a solid tissue or a fluid is the net result of not only protein synthesis and degradation but also transport across biological compartments. We report an accurate 3-biological compartment model able to simultaneously account for the protein dynamics observed in blood plasma and the cerebrospinal fluid (CSF) including a hidden central nervous system (CNS) compartment. We successfully applied this model to 69 proteins of a single individual displaying similar or very different dynamics in plasma and CSF. This study puts a strong emphasis on the methods and tools needed to develop this type of model. We believe that it will be useful to any researcher dealing with protein dynamics data modeling.
Collapse
Affiliation(s)
- Pierre Giroux
- Université
de Montpellier, 34090 Montpellier, France
- Institut
Régional du Cancer Montpellier (ICM), 34298 Montpellier, France
- Institut
de Recherche en Cancérologie de Montpellier (IRCM), Inserm
U1194, 34298 Montpellier, France
| | - Jérôme Vialaret
- LBPC-PPC
CHU Montpellier, INM, Inserm, 34295 Montpellier, France
| | - Jana Kindermans
- LBPC-PPC
CHU Montpellier, INM, Inserm, 34295 Montpellier, France
| | - Audrey Gabelle
- Université
de Montpellier, 34090 Montpellier, France
- CMRR
CHU Montpellier, INM, Inserm, 34295 Montpellier, France
| | - Luc Bauchet
- Université
de Montpellier, 34090 Montpellier, France
- Department
of Neurosurgery, CHU Montpellier, INM, Inserm, 34295 Montpellier, France
| | - Christophe Hirtz
- Université
de Montpellier, 34090 Montpellier, France
- LBPC-PPC
CHU Montpellier, INM, Inserm, 34295 Montpellier, France
- CMRR
CHU Montpellier, INM, Inserm, 34295 Montpellier, France
| | - Sylvain Lehmann
- Université
de Montpellier, 34090 Montpellier, France
- LBPC-PPC
CHU Montpellier, INM, Inserm, 34295 Montpellier, France
| | - Jacques Colinge
- Université
de Montpellier, 34090 Montpellier, France
- Institut
Régional du Cancer Montpellier (ICM), 34298 Montpellier, France
- Institut
de Recherche en Cancérologie de Montpellier (IRCM), Inserm
U1194, 34298 Montpellier, France
| |
Collapse
|
3
|
Pellegrini L, Silva-Vargas V, Patrizi A. Breakthroughs in choroid plexus and CSF biology from the first European Choroid plexus Scientific Forum (ECSF). Fluids Barriers CNS 2024; 21:43. [PMID: 38773599 PMCID: PMC11106960 DOI: 10.1186/s12987-024-00546-4] [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: 03/20/2024] [Accepted: 05/07/2024] [Indexed: 05/24/2024] Open
Abstract
The European Choroid plexus Scientific Forum (ECSF), held in Heidelberg, Germany between the 7th and 9th of November 2023, involved 21 speakers from eight countries. ECSF focused on discussing cutting-edge fundamental and medical research related to the development and functions of the choroid plexus and its implications for health, aging, and disease, including choroid plexus tumors. In addition to new findings in this expanding field, innovative approaches, animal models and 3D in vitro models were showcased to encourage further investigation into choroid plexus and cerebrospinal fluid roles.
Collapse
Affiliation(s)
- Laura Pellegrini
- Centre for Developmental Neurobiology, Guys Campus, King's College London, New Hunt's House, London, UK.
| | | | - Annarita Patrizi
- Schaller Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
4
|
Lista S, Mapstone M, Caraci F, Emanuele E, López-Ortiz S, Martín-Hernández J, Triaca V, Imbimbo C, Gabelle A, Mielke MM, Nisticò R, Santos-Lozano A, Imbimbo BP. A critical appraisal of blood-based biomarkers for Alzheimer's disease. Ageing Res Rev 2024; 96:102290. [PMID: 38580173 DOI: 10.1016/j.arr.2024.102290] [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: 11/13/2023] [Revised: 03/18/2024] [Accepted: 03/31/2024] [Indexed: 04/07/2024]
Abstract
Biomarkers that predict the clinical onset of Alzheimer's disease (AD) enable the identification of individuals in the early, preclinical stages of the disease. Detecting AD at this point may allow for more effective therapeutic interventions and optimized enrollment for clinical trials of novel drugs. The current biological diagnosis of AD is based on the AT(N) classification system with the measurement of brain deposition of amyloid-β (Aβ) ("A"), tau pathology ("T"), and neurodegeneration ("N"). Diagnostic cut-offs for Aβ1-42, the Aβ1-42/Aβ1-40 ratio, tau and hyperphosphorylated-tau concentrations in cerebrospinal fluid have been defined and may support AD clinical diagnosis. Blood-based biomarkers of the AT(N) categories have been described in the AD continuum. Cross-sectional and longitudinal studies have shown that the combination of blood biomarkers tracking neuroaxonal injury (neurofilament light chain) and neuroinflammatory pathways (glial fibrillary acidic protein) enhance sensitivity and specificity of AD clinical diagnosis and improve the prediction of AD onset. However, no international accepted cut-offs have been identified for these blood biomarkers. A kit for blood Aβ1-42/Aβ1-40 is commercially available in the U.S.; however, it does not provide a diagnosis, but simply estimates the risk of developing AD. Although blood-based AD biomarkers have a great potential in the diagnostic work-up of AD, they are not ready for the routine clinical use.
Collapse
Affiliation(s)
- Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Mark Mapstone
- Department of Neurology, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA.
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, Catania 95125, Italy; Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, Troina 94018, Italy.
| | | | - Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Juan Martín-Hernández
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Viviana Triaca
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Rome 00015, Italy.
| | - Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy.
| | - Audrey Gabelle
- Memory Resources and Research Center, Montpellier University of Excellence i-site, Montpellier 34295, France.
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA.
| | - Robert Nisticò
- School of Pharmacy, University of Rome "Tor Vergata", Rome 00133, Italy; Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome 00143, Italy.
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain; Physical Activity and Health Research Group (PaHerg), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid 28041, Spain.
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, Parma 43122, Italy.
| |
Collapse
|
5
|
Saloner R, Staffaroni A, Dammer E, Johnson ECB, Paolillo E, Wise A, Heuer H, Forsberg L, Lago AL, Webb J, Vogel J, Santillo A, Hansson O, Kramer J, Miller B, Li J, Loureiro J, Sivasankaran R, Worringer K, Seyfried N, Yokoyama J, Seeley W, Spina S, Grinberg L, VandeVrede L, Ljubenkov P, Bayram E, Bozoki A, Brushaber D, Considine C, Day G, Dickerson B, Domoto-Reilly K, Faber K, Galasko D, Geschwind D, Ghoshal N, Graff-Radford N, Hales C, Honig L, Hsiung GY, Huey E, Kornak J, Kremers W, Lapid M, Lee S, Litvan I, McMillan C, Mendez M, Miyagawa T, Pantelyat A, Pascual B, Paulson H, Petrucelli L, Pressman P, Ramos E, Rascovsky K, Roberson E, Savica R, Snyder A, Sullivan AC, Tartaglia C, Vandebergh M, Boeve B, Rosen H, Rojas J, Boxer A, Casaletto K. Large-scale network analysis of the cerebrospinal fluid proteome identifies molecular signatures of frontotemporal lobar degeneration. RESEARCH SQUARE 2024:rs.3.rs-4103685. [PMID: 38585969 PMCID: PMC10996789 DOI: 10.21203/rs.3.rs-4103685/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The pathophysiological mechanisms driving disease progression of frontotemporal lobar degeneration (FTLD) and corresponding biomarkers are not fully understood. We leveraged aptamer-based proteomics (> 4,000 proteins) to identify dysregulated communities of co-expressed cerebrospinal fluid proteins in 116 adults carrying autosomal dominant FTLD mutations (C9orf72, GRN, MAPT) compared to 39 noncarrier controls. Network analysis identified 31 protein co-expression modules. Proteomic signatures of genetic FTLD clinical severity included increased abundance of RNA splicing (particularly in C9orf72 and GRN) and extracellular matrix (particularly in MAPT) modules, as well as decreased abundance of synaptic/neuronal and autophagy modules. The generalizability of genetic FTLD proteomic signatures was tested and confirmed in independent cohorts of 1) sporadic progressive supranuclear palsy-Richardson syndrome and 2) frontotemporal dementia spectrum syndromes. Network-based proteomics hold promise for identifying replicable molecular pathways in adults living with FTLD. 'Hub' proteins driving co-expression of affected modules warrant further attention as candidate biomarkers and therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | | | - Amy Wise
- University of California, San Francisco
| | | | | | | | | | | | | | | | | | | | - Jingyao Li
- Novartis Institutes for Biomedical Research, Inc
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Suzee Lee
- University of California, San Francisco
| | | | - Corey McMillan
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Adam Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | | |
Collapse
|
6
|
Galasko D, Farlow MR, Lucey BP, Honig LS, Elbert D, Bateman R, Momper J, Thomas R, Rissman RA, Pa J, Aslanyan V, Balasubramanian A, West T, Maccecchini M, Feldman HH. A multicenter, randomized, double-blind, placebo-controlled ascending dose study to evaluate the safety, tolerability, pharmacokinetics (PK) and pharmacodynamic (PD) effects of Posiphen in subjects with Early Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.20.24304638. [PMID: 38562783 PMCID: PMC10984053 DOI: 10.1101/2024.03.20.24304638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Background Amyloid beta protein (Aβ) is a treatment target in Alzheimer's Disease (AD). Lowering production of its parent protein, APP, has benefits in preclinical models. Posiphen binds to an iron-responsive element in APP mRNA and decreases translation of APP and Aβ. To augment human data for Posiphen, we evaluated safety, tolerability and pharmacokinetic and pharmacodynamic (PD) effects on Aβ metabolism using Stable Isotope Labeling Kinetic (SILK) analysis. Methods Double-blind phase 1b randomized ascending dose clinical trial, at five sites, under an IRB-approved protocol. Participants with mild cognitive impairment or mild AD (Early AD) with positive CSF biomarkers were randomized (within each dose arm) to Posiphen or placebo. Pretreatment assessment included lumbar puncture for CSF. Participants took Posiphen or placebo for 21-23 days, then underwent CSF catheter placement, intravenous infusion of 13C6-leucine, and CSF sampling for 36 hours. Safety and tolerability were assessed through participant reports, EKG and laboratory tests. CSF SILK analysis measured Aβ40, 38 and 42 with immunoprecipitation-mass spectrometry. Baseline and day 21 CSF APP, Aβ and other biomarkers were measured with immunoassays. The Mini-Mental State Exam and ADAS-cog12 were given at baseline and day 21. Results From June 2017 to December 2021, 19 participants were enrolled, in dose cohorts (6 active: 2 placebo) of 60 mg once/day and 60 mg twice/day; 1 participant was enrolled and completed 60 mg three times/day. 10 active drug and 5 placebo participants completed all study procedures. Posiphen was safe and well-tolerated. 8 participants had headaches related to CSF catheterization; 5 needed blood patches. Prespecified SILK analyses of Fractional Synthesis Rate (FSR) for CSF Aβ40 showed no significant overall or dose-dependent effects of Posiphen vs. placebo. Comprehensive multiparameter modeling of APP kinetics supported dose-dependent lowering of APP production by Posiphen. Cognitive measures and CSF biomarkers did not change significantly from baseline to 21 days in Posiphen vs placebo groups. Conclusions Posiphen was safe and well-tolerated in Early AD. A multicenter SILK study was feasible. Findings are limited by small sample size but provide additional supportive safety and PK data. Comprehensive modeling of biomarker dynamics using SILK data may reveal subtle drug effects. Trial registration NCT02925650 on clinicaltrials.gov.
Collapse
|
7
|
van der Ende EL, In ‘t Veld SGJG, Hanskamp I, van der Lee S, Dijkstra JIR, Hok-A-Hin YS, Blujdea ER, van Swieten JC, Irwin DJ, Chen-Plotkin A, Hu WT, Lemstra AW, Pijnenburg YAL, van der Flier WM, del Campo M, Teunissen CE, Vermunt L. CSF proteomics in autosomal dominant Alzheimer's disease highlights parallels with sporadic disease. Brain 2023; 146:4495-4507. [PMID: 37348871 PMCID: PMC10629764 DOI: 10.1093/brain/awad213] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 06/24/2023] Open
Abstract
Autosomal dominant Alzheimer's disease (ADAD) offers a unique opportunity to study pathophysiological changes in a relatively young population with few comorbidities. A comprehensive investigation of proteome changes occurring in ADAD could provide valuable insights into AD-related biological mechanisms and uncover novel biomarkers and therapeutic targets. Furthermore, ADAD might serve as a model for sporadic AD, but in-depth proteome comparisons are lacking. We aimed to identify dysregulated CSF proteins in ADAD and determine the degree of overlap with sporadic AD. We measured 1472 proteins in CSF of PSEN1 or APP mutation carriers (n = 22) and age- and sex-matched controls (n = 20) from the Amsterdam Dementia Cohort using proximity extension-based immunoassays (PEA). We compared protein abundance between groups with two-sided t-tests and identified enriched biological pathways. Using the same protein panels in paired plasma samples, we investigated correlations between CSF proteins and their plasma counterparts. Finally, we compared our results with recently published PEA data from an international cohort of sporadic AD (n = 230) and non-AD dementias (n = 301). All statistical analyses were false discovery rate-corrected. We detected 66 differentially abundant CSF proteins (65 increased, 1 decreased) in ADAD compared to controls (q < 0.05). The most strongly upregulated proteins (fold change >1.8) were related to immunity (CHIT1, ITGB2, SMOC2), cytoskeletal structure (MAPT, NEFL) and tissue remodelling (TMSB10, MMP-10). Significant CSF-plasma correlations were found for the upregulated proteins SMOC2 and LILR1B. Of the 66 differentially expressed proteins, 36 had been measured previously in the sporadic dementias cohort, 34 of which (94%) were also significantly upregulated in sporadic AD, with a strong correlation between the fold changes of these proteins in both cohorts (rs = 0.730, P < 0.001). Twenty-nine of the 36 proteins (81%) were also upregulated among non-AD patients with suspected AD co-pathology. This CSF proteomics study demonstrates substantial biochemical similarities between ADAD and sporadic AD, suggesting involvement of the same biological processes. Besides known AD-related proteins, we identified several relatively novel proteins, such as TMSB10, MMP-10 and SMOC2, which have potential as novel biomarkers. With shared pathophysiological CSF changes, ADAD study findings might be translatable to sporadic AD, which could greatly expedite therapy development.
Collapse
Affiliation(s)
- Emma L van der Ende
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sjors G J G In ‘t Veld
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Iris Hanskamp
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sven van der Lee
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Janna I R Dijkstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yanaika S Hok-A-Hin
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Elena R Blujdea
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - John C van Swieten
- Alzheimer Center and Department of Neurology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William T Hu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Afina W Lemstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Epidemiology and Data Science, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Marta del Campo
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28003 Madrid, Spain
- Barcelonabeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
8
|
Spataro S, Maco B, Escrig S, Jensen L, Polerecky L, Knott G, Meibom A, Schneider BL. Stable isotope labeling and ultra-high-resolution NanoSIMS imaging reveal alpha-synuclein-induced changes in neuronal metabolism in vivo. Acta Neuropathol Commun 2023; 11:157. [PMID: 37770947 PMCID: PMC10540389 DOI: 10.1186/s40478-023-01608-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 09/30/2023] Open
Abstract
In Parkinson's disease, pathogenic factors such as the intraneuronal accumulation of the protein α-synuclein affect key metabolic processes. New approaches are required to understand how metabolic dysregulations cause degeneration of vulnerable subtypes of neurons in the brain. Here, we apply correlative electron microscopy and NanoSIMS isotopic imaging to map and quantify 13C enrichments in dopaminergic neurons at the subcellular level after pulse-chase administration of 13C-labeled glucose. To model a condition leading to neurodegeneration in Parkinson's disease, human α-synuclein was unilaterally overexpressed in the substantia nigra of one brain hemisphere in rats. When comparing neurons overexpressing α-synuclein to those located in the control hemisphere, the carbon anabolism and turnover rates revealed metabolic anomalies in specific neuronal compartments and organelles. Overexpression of α-synuclein enhanced the overall carbon turnover in nigral neurons, despite a lower relative incorporation of carbon inside the nucleus. Furthermore, mitochondria and Golgi apparatus showed metabolic defects consistent with the effects of α-synuclein on inter-organellar communication. By revealing changes in the kinetics of carbon anabolism and turnover at the subcellular level, this approach can be used to explore how neurodegeneration unfolds in specific subpopulations of neurons.
Collapse
Affiliation(s)
- Sofia Spataro
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Bohumil Maco
- Laboratory for Biological Geochemistry, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Stéphane Escrig
- Laboratory for Biological Geochemistry, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Louise Jensen
- Laboratory for Biological Geochemistry, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Lubos Polerecky
- Department of Earth Sciences, Faculty of Geosciences, Utrecht University, Utrecht, The Netherlands
| | - Graham Knott
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Bioelectron Microscopy Core Facility, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Anders Meibom
- Laboratory for Biological Geochemistry, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Center for Advanced Surface Analysis, Institute of Earth Sciences, University of Lausanne, Lausanne, Switzerland.
- EPFL ENAC IIE LGB, Station 2, 1015, Lausanne, Switzerland.
| | - Bernard L Schneider
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland.
- EPFL SV PTECH PTBTG, Ch. Des Mines 9, 1202, Geneva, Switzerland.
| |
Collapse
|
9
|
Ikegawa M, Kakuda N, Miyasaka T, Toyama Y, Nirasawa T, Minta K, Hanrieder J. Mass Spectrometry Imaging in Alzheimer's Disease. Brain Connect 2023; 13:319-333. [PMID: 36905365 PMCID: PMC10494909 DOI: 10.1089/brain.2022.0057] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023] Open
Abstract
Introduction: Amyloid-beta (Aβ) pathology is the precipitating histopathological characteristic of Alzheimer's disease (AD). Although the formation of amyloid plaques in human brains is suggested to be a key factor in initiating AD pathogenesis, it is still not fully understood the upstream events that lead to Aβ plaque formation and its metabolism inside the brains. Methods: Matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) has been successfully introduced to study AD pathology in brain tissue both in AD mouse models and human samples. By using MALDI-MSI, a highly selective deposition of Aβ peptides in AD brains with a variety of cerebral amyloid angiopathy (CAA) involvement was observed. Results: MALDI-MSI visualized depositions of shorter peptides in AD brains; Aβ1-36 to Aβ1-39 were quite similarly distributed with Aβ1-40 as a vascular pattern, and deposition of Aβ1-42 and Aβ1-43 was visualized with a distinct senile plaque pattern distributed in parenchyma. Moreover, how MALDI-MSI covered in situ lipidomics of plaque pathology has been reviewed, which is of interest as aberrations in neuronal lipid biochemistry have been implicated in AD pathogenesis. Discussion: In this study, we introduce the methodological concepts and challenges of MALDI-MSI for the studies of AD pathogenesis. Diverse Aβ isoforms including various C- and N-terminal truncations in AD and CAA brain tissues will be visualized. Despite the close relationship between vascular and plaque Aβ deposition, the current strategy will define cross talk between neurodegenerative and cerebrovascular processes at the level of Aβ metabolism.
Collapse
Affiliation(s)
- Masaya Ikegawa
- Department of Life and Medical Systems, Doshisha University, Kyotanabe, Kyoto, Japan
| | - Nobuto Kakuda
- Department of Life and Medical Systems, Doshisha University, Kyotanabe, Kyoto, Japan
| | - Tomohiro Miyasaka
- Department of Life and Medical Systems, Doshisha University, Kyotanabe, Kyoto, Japan
| | - Yumiko Toyama
- Department of Life and Medical Systems, Doshisha University, Kyotanabe, Kyoto, Japan
| | | | - Karolina Minta
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Amyotrophic lateral sclerosis (ALS) is an incurable, devastating neurodegenerative disease. Still, the diagnosis is mainly based on clinical symptoms, and the treatment options are strongly limited. However, the pipeline of potential treatments currently tested in clinical trials is promising. This review will discuss developments in ALS biomarker research and applications within the last 2 years and suggest future directions and needs. RECENT FINDINGS The diagnostic and prognostic utility of neurofilaments, a general marker for axoneuronal degeneration, has been confirmed by further studies in patients with ALS, and neurofilaments are finding their way into routine diagnostic and clinical trials. Additionally, there have been advancements in developing and implementing disease-specific biomarkers, especially in patients with a genetic variant, such as SOD1 or C9orf72 . Here, biomarkers have already been used as target markers and outcome parameters for novel treatment approaches. In addition, several novel biomarkers have shown encouraging results but should be discussed in the context of their early stage of assay and clinical establishment. SUMMARY The first biomarkers have found their way into clinical routine in ALS. In light of an increasing pipeline of potential treatments, further progress in discovering and implementing novel and existing biomarkers is crucial.
Collapse
Affiliation(s)
- Simon Witzel
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg
| | - Kristina Mayer
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg
| | - Patrick Oeckl
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg
- German Center for Neurodegenerative Diseases (DZNE), Site Ulm, Ulm, Germany
| |
Collapse
|
11
|
Dobrowolska Zakaria JA, Bateman RJ, Lysakowska M, Khatri A, Jean-Gilles D, Kennedy ME, Vassar R. The metabolism of human soluble amyloid precursor protein isoforms is quantifiable by a stable isotope labeling-tandem mass spectrometry method. Sci Rep 2022; 12:14985. [PMID: 36056033 PMCID: PMC9440206 DOI: 10.1038/s41598-022-18869-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 08/22/2022] [Indexed: 11/19/2022] Open
Abstract
Evidence suggests that β-secretase (BACE1), which cleaves Amyloid Precursor Protein (APP) to form sAPPβ and amyloid-β, is elevated in Alzheimer's disease (AD) brains and biofluids and, thus, BACE1 is a therapeutic target for this devastating disease. The direct product of BACE1 cleavage of APP, sAPPβ, serves as a surrogate marker of BACE1 activity in the central nervous system. This biomarker could be utilized to better understand normal APP processing, aberrant processing in the disease setting, and modulations to processing during therapeutic intervention. In this paper, we present a method for measuring the metabolism of sAPPβ and another APP proteolytic product, sAPPα, in vivo in humans using stable isotope labeling kinetics, paired with immunoprecipitation and liquid chromatography/tandem mass spectrometry. The method presented herein is robust, reproducible, and precise, and allows for the study of these analytes by taking into account their full dynamic potential as opposed to the traditional methods of absolute concentration quantitation that only provide a static view of a dynamic system. A study of in vivo cerebrospinal fluid sAPPβ and sAPPα kinetics using these methods could reveal novel insights into pathophysiological mechanisms of AD, such as increased BACE1 processing of APP.
Collapse
Affiliation(s)
- Justyna A Dobrowolska Zakaria
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- SILQ Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Monika Lysakowska
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Ammaarah Khatri
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | | | - Matthew E Kennedy
- Deparment of Neuroscience, Merck & Co., Inc., Boston, MA, 02115, USA
| | - Robert Vassar
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
12
|
Casalino E, Stine LB, Corin AJ, Thai CT, Quiroz J, Wilson SC, Labow M, Mittal S. A novel high-throughput screening strategy for targeting alpha-synuclein and other long-lived proteins. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:349-357. [PMID: 35580766 DOI: 10.1016/j.slasd.2022.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 06/15/2023]
Abstract
Small-molecule high-throughput screening (HTS) campaigns have frequently been used to identify lead molecules that can alter expression of disease-relevant proteins in cell-based assays. However, most cell-based HTS assays require short compound exposure periods to avoid toxicity and ensure that compounds are stable in media for the duration of the exposure. This limits the ability of HTS assays to detect inhibitors of the synthesis of target proteins with long half-lives, which can often exceed the exposure times utilized in most HTS campaigns. One such target is alpha-synuclein (α-syn)-a protein well-known for its pathological aggregation in Parkinson's Disease (PD) and other forms of neurodegeneration known collectively as synucleinopathies. Here, we report the development of an HTS assay using a CRISPR-engineered neuroblastoma cell line expressing a destabilized luciferase reporter inserted at the end of the coding region of the SNCA locus. The resultant destabilized fusion protein exhibited a significant reduction in half-life compared to the endogenous, unmodified α-syn protein, and accurately reported reductions in α-syn levels due to known protein translation inhibitors and specific α-syn siRNAs. The robustness and utility of this approach was shown by using the resulting cell line (dsLuc-Syn) to screen a focused library of 3,192 compounds for reduction of α-syn. These data demonstrate the general utility of converting endogenous loci into destabilized reporter genes capable of identifying inhibitors of gene expression of highly stable proteins even in short-term assays.
Collapse
Affiliation(s)
- Evan Casalino
- Neuroscience Thematic Research Center, Bristol Myers Squibb, Cambridge, MA, United States
| | - Laurel B Stine
- Inflammation, Cardiovascular and Fibrosis Thematic Research Center, Bristol Myers Squibb, Cambridge, MA, United States
| | - Aaron J Corin
- Inflammation, Cardiovascular and Fibrosis Thematic Research Center, Bristol Myers Squibb, Cambridge, MA, United States
| | - Chuong-Thu Thai
- Compound Management, Automation and Assay Technology, Bristol Myers Squibb, San Diego, CA, United States
| | - Jose Quiroz
- Compound Management, Automation and Assay Technology, Bristol Myers Squibb, San Diego, CA, United States
| | - Stephen C Wilson
- Inflammation, Cardiovascular and Fibrosis Thematic Research Center, Bristol Myers Squibb, Cambridge, MA, United States
| | - Mark Labow
- Neuroscience Thematic Research Center, Bristol Myers Squibb, Cambridge, MA, United States.
| | - Shuchi Mittal
- Neuroscience Thematic Research Center, Bristol Myers Squibb, Cambridge, MA, United States.
| |
Collapse
|
13
|
Gong X, Zhang H, Liu X, Liu Y, Liu J, Fapohunda FO, Lü P, Wang K, Tang M. Is liquid biopsy mature enough for the diagnosis of Alzheimer's disease? Front Aging Neurosci 2022; 14:977999. [PMID: 35992602 PMCID: PMC9389010 DOI: 10.3389/fnagi.2022.977999] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 07/18/2022] [Indexed: 01/10/2023] Open
Abstract
The preclinical diagnosis and clinical practice for Alzheimer's disease (AD) based on liquid biopsy have made great progress in recent years. As liquid biopsy is a fast, low-cost, and easy way to get the phase of AD, continual efforts from intense multidisciplinary studies have been made to move the research tools to routine clinical diagnostics. On one hand, technological breakthroughs have brought new detection methods to the outputs of liquid biopsy to stratify AD cases, resulting in higher accuracy and efficiency of diagnosis. On the other hand, diversiform biofluid biomarkers derived from cerebrospinal fluid (CSF), blood, urine, Saliva, and exosome were screened out and biologically verified. As a result, more detailed knowledge about the molecular pathogenesis of AD was discovered and elucidated. However, to date, how to weigh the reports derived from liquid biopsy for preclinical AD diagnosis is an ongoing question. In this review, we briefly introduce liquid biopsy and the role it plays in research and clinical practice. Then, we summarize the established fluid-based assays of the current state for AD diagnostic such as ELISA, single-molecule array (Simoa), Immunoprecipitation-Mass Spectrometry (IP-MS), liquid chromatography-MS, immunomagnetic reduction (IMR), multimer detection system (MDS). In addition, we give an updated list of fluid biomarkers in the AD research field. Lastly, the current outstanding challenges and the feasibility to use a stand-alone biomarker in the joint diagnostic strategy are discussed.
Collapse
Affiliation(s)
- Xun Gong
- Department of Rheumatology and Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Hantao Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Xiaoyan Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yi Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Junlin Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | | | - Peng Lü
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Kun Wang
- Children’s Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Min Tang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|
14
|
Mellinger AL, Muddiman DC, Gamcsik MP. Highlighting Functional Mass Spectrometry Imaging Methods in Bioanalysis. J Proteome Res 2022; 21:1800-1807. [PMID: 35749637 DOI: 10.1021/acs.jproteome.2c00220] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Most mass spectrometry imaging (MSI) methods provide a molecular map of tissue content but little information on tissue function. Mapping tissue function is possible using several well-known examples of "functional imaging" such as positron emission tomography and functional magnetic resonance imaging that can provide the spatial distribution of time-dependent biological processes. These functional imaging methods represent the net output of molecular networks influenced by local tissue environments that are difficult to predict from molecular/cellular content alone. However, for decades, MSI methods have also been demonstrated to provide functional imaging data on a variety of biological processes. In fact, MSI exceeds some of the classic functional imaging methods, demonstrating the ability to provide functional data from the nanoscale (subcellular) to whole tissue or organ level. This Perspective highlights several examples of how different MSI ionization and detection technologies can provide unprecedented detailed spatial maps of time-dependent biological processes, namely, nucleic acid synthesis, lipid metabolism, bioenergetics, and protein metabolism. By classifying various MSI methods under the umbrella of "functional MSI", we hope to draw attention to both the unique capabilities and accessibility with the aim of expanding this underappreciated field to include new approaches and applications.
Collapse
Affiliation(s)
- Allyson L Mellinger
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - David C Muddiman
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States.,Molecular Education, Technology and Research Innovation Center (METRIC), North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Michael P Gamcsik
- UNC/NCSU Joint Department of Biomedical Engineering, Raleigh, North Carolina 27695, United States
| |
Collapse
|
15
|
Sawa M, Overk C, Becker A, Derse D, Albay R, Weldy K, Salehi A, Beach TG, Doran E, Head E, Yu YE, Mobley WC. Impact of increased APP gene dose in Down syndrome and the Dp16 mouse model. Alzheimers Dement 2022; 18:1203-1234. [PMID: 34757693 PMCID: PMC9085977 DOI: 10.1002/alz.12463] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/21/2021] [Accepted: 07/29/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION People with Down syndrome (DS) are predisposed to Alzheimer's disease (AD). The amyloid hypothesis informs studies of AD. In AD-DS, but not sporadic AD, increased APP copy number is necessary, defining the APP gene dose hypothesis. Which amyloid precursor protein (APP) products contribute needs to be determined. METHODS Brain levels of full-length protein (fl-hAPP), C-terminal fragments (hCTFs), and amyloid beta (Aβ) peptides were measured in DS, AD-DS, non-demented controls (ND), and sporadic AD cases. The APP gene-dose hypothesis was evaluated in the Dp16 model. RESULTS DS and AD-DS differed from ND and AD for all APP products. In AD-DS, Aβ42 and Aβ40 levels exceeded AD. APP products were increased in the Dp16 model; increased APP gene dose was necessary for loss of vulnerable neurons, tau pathology, and activation of astrocytes and microglia. DISCUSSION Increases in APP products other than Aβ distinguished AD-DS from AD. Deciphering AD-DS pathogenesis necessitates deciphering which APP products contribute and how.
Collapse
Affiliation(s)
- Mariko Sawa
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Cassia Overk
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Ann Becker
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Dominique Derse
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Ricardo Albay
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Kim Weldy
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Ahmad Salehi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305
| | - Thomas G. Beach
- Brain and Body Donation Program, Banner Sun Health Research Institute, Sun City, AZ 85351
| | - Eric Doran
- Department of Pediatrics, University of California, Irvine, CA, 92697
| | - Elizabeth Head
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA, 92697
| | - Y. Eugene Yu
- The Children’s Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| | - William C Mobley
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624,Correspondence to: William Mobley M.D., Department of Neurosciences, UCSD School of Medicine, 9500 Gilman Drive, GPL 355, La Jolla, CA 92093-0624;
| |
Collapse
|
16
|
Aisen PS, Jimenez-Maggiora GA, Rafii MS, Walter S, Raman R. Early-stage Alzheimer disease: getting trial-ready. Nat Rev Neurol 2022; 18:389-399. [PMID: 35379951 PMCID: PMC8978175 DOI: 10.1038/s41582-022-00645-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 12/15/2022]
Abstract
Slowing the progression of Alzheimer disease (AD) might be the greatest unmet medical need of our time. Although one AD therapeutic has received a controversial accelerated approval from the FDA, more effective and accessible therapies are urgently needed. Consensus is growing that for meaningful disease modification in AD, therapeutic intervention must be initiated at very early (preclinical or prodromal) stages of the disease. Although the methods for such early-stage clinical trials have been developed, identification and recruitment of the required asymptomatic or minimally symptomatic study participants takes many years and requires substantial funds. As an example, in the Anti-Amyloid Treatment in Asymptomatic Alzheimer’s Disease Trial (the first phase III trial to be performed in preclinical AD), 3.5 years and more than 5,900 screens were required to recruit and randomize 1,169 participants. A new clinical trials infrastructure is required to increase the efficiency of recruitment and accelerate therapeutic progress. Collaborations in North America, Europe and Asia are now addressing this need by establishing trial-ready cohorts of individuals with preclinical and prodromal AD. These collaborations are employing innovative methods to engage the target population, assess risk of brain amyloid accumulation, select participants for biomarker studies and determine eligibility for trials. In the future, these programmes could provide effective tools for pursuing the primary prevention of AD. Here, we review the lessons learned from the AD trial-ready cohorts that have been established to date, with the aim of informing ongoing and future efforts towards efficient, cost-effective trial recruitment. Consensus is growing that intervention in the very early stages of Alzheimer disease is necessary for disease modification. Here, the authors discuss the challenges of recruiting asymptomatic or mildly symptomatic participants for clinical trials, focusing on ‘trial-ready’ cohorts as a potential solution. Trial-ready cohorts are an effective strategy for the identification of participants eligible for clinical trials in early-stage Alzheimer disease (AD). Building these cohorts requires considerable planning and technological infrastructure to facilitate recruitment, remote longitudinal assessment, data management and data storage. Trial-ready cohorts exist for genetically determined populations at risk of AD, such as those with familial AD and Down syndrome; the longitudinal data from these cohorts is improving our understanding of the disease progression in early stages, informing clinical trial design and accelerating recruitment to intervention studies. So far, the challenges experienced by trial-ready cohorts for early-stage AD have included difficulties recruiting an ethnically and racially representative cohort; and for online cohorts, difficulty retaining participants. The results of ongoing work will reveal the success of strategies to improve cohort diversity and retention, and the rates of referral to clinical trials.
Collapse
|
17
|
Sengupta U, Kayed R. Amyloid β, Tau, and α-Synuclein aggregates in the pathogenesis, prognosis, and therapeutics for neurodegenerative diseases. Prog Neurobiol 2022; 214:102270. [DOI: 10.1016/j.pneurobio.2022.102270] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/28/2022] [Accepted: 04/13/2022] [Indexed: 12/11/2022]
|
18
|
Sadygov RG. Protein turnover models for LC-MS data of heavy water metabolic labeling. Brief Bioinform 2022; 23:bbab598. [PMID: 35062023 PMCID: PMC8921656 DOI: 10.1093/bib/bbab598] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/12/2021] [Accepted: 12/26/2021] [Indexed: 01/23/2023] Open
Abstract
Protein turnover is vital for cellular functioning and is often associated with the pathophysiology of a variety of diseases. Metabolic labeling with heavy water followed by liquid chromatography coupled to mass spectrometry is a powerful tool to study in vivo protein turnover in high throughput and large scale. Heavy water is a cost-effective and easy to use labeling agent. It labels all nonessential amino acids. Due to its toxicity in high concentrations (20% or higher), small enrichments (8% or smaller) of heavy water are used with most organisms. The low concentration results in incomplete labeling of peptides/proteins. Therefore, the data processing is more challenging and requires accurate quantification of labeled and unlabeled forms of a peptide from overlapping mass isotopomer distributions. The work describes the bioinformatics aspects of the analysis of heavy water labeled mass spectral data, available software tools and current challenges and opportunities.
Collapse
Affiliation(s)
- Rovshan G Sadygov
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, 301 University of Blvd, Galveston, TX 77555, USA
| |
Collapse
|
19
|
Chhatwal JP, Schultz SA, McDade E, Schultz AP, Liu L, Hanseeuw BJ, Joseph-Mathurin N, Feldman R, Fitzpatrick CD, Sparks KP, Levin J, Berman SB, Renton AE, Esposito BT, Fernandez MV, Sung YJ, Lee JH, Klunk WE, Hofmann A, Noble JM, Graff-Radford N, Mori H, Salloway SM, Masters CL, Martins R, Karch CM, Xiong C, Cruchaga C, Perrin RJ, Gordon BA, Benzinger TLS, Fox NC, Schofield PR, Fagan AM, Goate AM, Morris JC, Bateman RJ, Johnson KA, Sperling RA. Variant-dependent heterogeneity in amyloid β burden in autosomal dominant Alzheimer's disease: cross-sectional and longitudinal analyses of an observational study. Lancet Neurol 2022; 21:140-152. [PMID: 35065037 PMCID: PMC8956209 DOI: 10.1016/s1474-4422(21)00375-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 10/13/2021] [Accepted: 10/22/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Insights gained from studying individuals with autosomal dominant Alzheimer's disease have broadly influenced mechanistic hypotheses, biomarker development, and clinical trials in both sporadic and dominantly inherited Alzheimer's disease. Although pathogenic variants causing autosomal dominant Alzheimer's disease are highly penetrant, there is substantial heterogeneity in levels of amyloid β (Aβ) between individuals. We aimed to examine whether this heterogeneity is related to disease progression and to investigate the association with mutation location within PSEN1, PSEN2, or APP. METHODS We did cross-sectional and longitudinal analyses of data from the Dominantly Inherited Alzheimer's Network (DIAN) observational study, which enrols individuals from families affected by autosomal dominant Alzheimer's disease. 340 participants in the DIAN study who were aged 18 years or older, had a history of autosomal dominant Alzheimer's disease in their family, and who were enrolled between September, 2008, and June, 2019, were included in our analysis. 206 participants were carriers of pathogenic mutations in PSEN1, PSEN2, or APP, and 134 were non-carriers. 62 unique pathogenic variants were identified in the cohort and were grouped in two ways. First, we sorted variants in PSEN1, PSEN2, or APP by the affected protein domain. Second, we divided PSEN1 variants according to position before or after codon 200. We examined variant-dependent variability in Aβ biomarkers, specifically Pittsburgh-Compound-B PET (PiB-PET) signal, levels of CSF Aβ1-42 (Aβ42), and levels of Aβ1-40 (Aβ40). FINDINGS Cortical and striatal PiB-PET signal showed striking variant-dependent variability using both grouping approaches (p<0·0001), despite similar progression on the clinical dementia rating (p>0·7), and CSF Aβ42 levels (codon-based grouping: p=0·49; domain-based grouping: p=0·095). Longitudinal PiB-PET signal also varied across codon-based groups, mirroring cross-sectional analyses. INTERPRETATION Autosomal dominant Alzheimer's disease pathogenic variants showed highly differential temporal and regional patterns of PiB-PET signal, despite similar functional progression. These findings suggest that although increased PiB-PET signal is generally seen in autosomal dominant Alzheimer's disease, higher levels of PiB-PET signal at an individual level might not reflect more severe or more advanced disease. Our results have high relevance for ongoing clinical trials in autosomal dominant Alzheimer's disease, including those using Aβ PET as a surrogate marker of disease progression. Additionally, and pertinent to both sporadic and autosomal dominant Alzheimer's disease, our results suggest that CSF and PET measures of Aβ levels are not interchangeable and might reflect different Aβ-driven pathobiological processes. FUNDING National Institute on Aging, Doris Duke Charitable Foundation, German Center for Neurodegenerative Diseases, Japanese Agency for Medical Research and Development.
Collapse
Affiliation(s)
- Jasmeer P Chhatwal
- Department of Neurology, Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA; Brigham and Women's Hospital, Boston, MA, USA.
| | - Stephanie A Schultz
- Department of Neurology, Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Aaron P Schultz
- Department of Neurology, Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA
| | - Lei Liu
- Department of Neurology, Harvard Medical School, Boston, MA, USA; Brigham and Women's Hospital, Boston, MA, USA
| | - Bernard J Hanseeuw
- Department of Neurology, Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA; Université Catholique de Louvain, Brussels, Belgium
| | - Nelly Joseph-Mathurin
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Rebecca Feldman
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Colleen D Fitzpatrick
- Massachusetts General Hospital, Boston, MA, USA; Brigham and Women's Hospital, Boston, MA, USA
| | | | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Neurodegenerative Diseases, Munich, Germany
| | - Sarah B Berman
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alan E Renton
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bianca T Esposito
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Yun Ju Sung
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Jae Hong Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - William E Klunk
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anna Hofmann
- German Center for Neurodegenerative Disease, Tübingen, Germany
| | - James M Noble
- Columbia University Irving Medical Center, Department of Neurology, New York, NY, USA
| | | | - Hiroshi Mori
- Osaka City University, Sumiyoshi Ward, Osaka, Japan
| | - Steven M Salloway
- Butler Hospital, Memory and Aging Program, Brown University Alpert Medical School, Providence, RI, USA
| | - Colin L Masters
- The University of Melbourne, Melbourne, VIC, Australia; Florey Institute, Melbourne, VIC, Australia
| | - Ralph Martins
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Celeste M Karch
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University in St Louis, St Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Richard J Perrin
- Department of Pathology, Washington University in St Louis, St Louis, MO, USA
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Nick C Fox
- UCL Queen Square Institute of Neurology, Dementia Research Centre, London, UK
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia; School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Anne M Fagan
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Alison M Goate
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John C Morris
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Keith A Johnson
- Department of Neurology, Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA; Brigham and Women's Hospital, Boston, MA, USA
| | - Reisa A Sperling
- Department of Neurology, Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA; Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
20
|
Budelier MM, He Y, Barthelemy NR, Jiang H, Li Y, Park E, Henson RL, Schindler SE, Holtzman DM, Bateman RJ. OUP accepted manuscript. Brain Commun 2022; 4:fcac045. [PMID: 35415607 PMCID: PMC8994116 DOI: 10.1093/braincomms/fcac045] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/19/2021] [Accepted: 02/17/2022] [Indexed: 11/12/2022] Open
Abstract
Neurofilament light is a well-established marker of both acute and chronic neuronal
damage and is increased in multiple neurodegenerative diseases. However, the protein is
not well characterized in brain tissue or body fluids, and it is unknown what
neurofilament light species are detected by commercial assays and whether additional
species exist. We developed an immunoprecipitation-mass spectrometry assay using custom
antibodies targeting various neurofilament light domains, including antibodies targeting
Coil 1A/1B of the rod domain (HJ30.13), Coil 2B of the rod domain (HJ30.4) and the tail
region (HJ30.11). We utilized our assay to characterize neurofilament light in brain
tissue and CSF of individuals with Alzheimer’s disease dementia and healthy controls. We
then validated a quantitative version of our assay and measured neurofilament light
concentrations using both our quantitative immunoprecipitation-mass spectrometry assay and
the commercially available immunoassay from Uman diagnostics in individuals with and
without Alzheimer’s disease dementia. Our validation cohort included CSF samples from 30
symptomatic amyloid-positive participants, 16 asymptomatic amyloid-positive participants,
10 symptomatic amyloid-negative participants and 25 amyloid-negative controls. We
identified at least three major neurofilament light species in CSF, including N-terminal
and C-terminal truncations, and a C-terminal fragment containing the tail domain. No
full-length neurofilament light was identified in CSF. This contrasts with brain tissue,
which contained mostly full-length neurofilament and a C-terminal tail domain fragment. We
observed an increase in neurofilament light concentrations in individuals with Alzheimer’s
disease compared with healthy controls, with larger differences for some neurofilament
light species than for others. The largest differences were observed for neurofilament
light fragments including NfL165 (in Coil 1B), NfL324 (in Coil 2B) and NfL530 (in the
C-terminal tail domain). The Uman immunoassay correlated most with NfL324. This study
provides a comprehensive evaluation of neurofilament light in brain and CSF and enables
future investigations of neurofilament light biology and utility as a biomarker.
Collapse
Affiliation(s)
- Melissa M. Budelier
- Department of Neurology, Washington University School of
Medicine, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of
Medicine, St Louis, MO, USA
| | - Yingxin He
- Department of Neurology, Washington University School of
Medicine, St Louis, MO, USA
| | - Nicolas R. Barthelemy
- Department of Neurology, Washington University School of
Medicine, St Louis, MO, USA
| | - Hong Jiang
- Department of Neurology, Washington University School of
Medicine, St Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of
Medicine, St Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington
University School of Medicine, St Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University School of
Medicine, St Louis, MO, USA
| | - Ethan Park
- Department of Biostatistics, Washington University School of
Medicine, St Louis, MO, USA
| | - Rachel L. Henson
- Department of Neurology, Washington University School of
Medicine, St Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington
University School of Medicine, St Louis, MO, USA
| | - Suzanne E. Schindler
- Department of Neurology, Washington University School of
Medicine, St Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington
University School of Medicine, St Louis, MO, USA
| | - David M. Holtzman
- Department of Neurology, Washington University School of
Medicine, St Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of
Medicine, St Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington
University School of Medicine, St Louis, MO, USA
| | - Randall J. Bateman
- Department of Neurology, Washington University School of
Medicine, St Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of
Medicine, St Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington
University School of Medicine, St Louis, MO, USA
- Correspondence to: Randall J. Bateman, MD Department of Neurology
Washington University School of Medicine 660 South Euclid Avenue, Campus Box 8111 St Louis
MO 63110, USA E-mail:
| |
Collapse
|
21
|
Khosropanah MH, Vaghasloo MA, Shakibaei M, Mueller AL, Kajbafzadeh AM, Amani L, Haririan I, Azimzadeh A, Hassannejad Z, Zolbin MM. Biomedical applications of silkworm (Bombyx Mori) proteins in regenerative medicine (a narrative review). J Tissue Eng Regen Med 2021; 16:91-109. [PMID: 34808032 DOI: 10.1002/term.3267] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/12/2021] [Accepted: 10/19/2021] [Indexed: 12/19/2022]
Abstract
Silk worm (Bombyx Mori) protein, have been considered as potential materials for a variety of advanced engineering and biomedical applications for decades. Recently, silkworm silk has gained significant importance in research attention mainly because of its remarkable and exceptional mechanical properties. Silk has already been shown to have unique interactions with cells in tissues through bio-recognition units. The natural silk contains fibroin and sericin and has been used in various tissues of the human body (skin, bone, nerve, and so on). Besides, silk also still has anti-cancer, anti-tyrosinase, anti-coagulant, anti-oxidant, anti-bacterial, and anti-diabetic properties. This article is supposed to describe the diverse biomedical capabilities of B. Mori silk as the appropriate biomaterial among the assorted natural and artificial polymers that are presently accessible, and ideal for usage in regenerative medicine fields.
Collapse
Affiliation(s)
- Mohammad Hossein Khosropanah
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Alizadeh Vaghasloo
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Anna-Lena Mueller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Abdol-Mohammad Kajbafzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Amani
- Department of Traditional Pharmacy, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy and Department of Pharmaceutics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashkan Azimzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Hassannejad
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Majidi Zolbin
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Hark TJ, Savas JN. Using stable isotope labeling to advance our understanding of Alzheimer's disease etiology and pathology. J Neurochem 2021; 159:318-329. [PMID: 33434345 PMCID: PMC8273190 DOI: 10.1111/jnc.15298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 12/18/2022]
Abstract
Stable isotope labeling with mass spectrometry (MS)-based proteomic analysis has become a powerful strategy to assess protein steady-state levels, protein turnover, and protein localization. Applying these analyses platforms to neurodegenerative disorders may uncover new aspects of the etiology of these devastating diseases. Recently, stable isotopes-MS has been used to investigate early pathological mechanisms of Alzheimer's disease (AD) with mouse models of AD-like pathology. In this review, we summarize these stable isotope-MS experimental designs and the recent application in the context of AD pathology. We also describe our current efforts aimed at using nuclear magnetic resonance (NMR) analysis of stable isotope-labeled amyloid fibrils from AD mouse model brains. Collectively, these methodologies offer new opportunities to study proteome changes in AD and other neurodegenerative diseases by elucidating mechanisms to target for treatment and prevention.
Collapse
Affiliation(s)
- Timothy J Hark
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jeffrey N Savas
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
23
|
Yuan A, Nixon RA. Neurofilament Proteins as Biomarkers to Monitor Neurological Diseases and the Efficacy of Therapies. Front Neurosci 2021; 15:689938. [PMID: 34646114 PMCID: PMC8503617 DOI: 10.3389/fnins.2021.689938] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/02/2021] [Indexed: 01/01/2023] Open
Abstract
Biomarkers of neurodegeneration and neuronal injury have the potential to improve diagnostic accuracy, disease monitoring, prognosis, and measure treatment efficacy. Neurofilament proteins (NfPs) are well suited as biomarkers in these contexts because they are major neuron-specific components that maintain structural integrity and are sensitive to neurodegeneration and neuronal injury across a wide range of neurologic diseases. Low levels of NfPs are constantly released from neurons into the extracellular space and ultimately reach the cerebrospinal fluid (CSF) and blood under physiological conditions throughout normal brain development, maturation, and aging. NfP levels in CSF and blood rise above normal in response to neuronal injury and neurodegeneration independently of cause. NfPs in CSF measured by lumbar puncture are about 40-fold more concentrated than in blood in healthy individuals. New ultra-sensitive methods now allow minimally invasive measurement of these low levels of NfPs in serum or plasma to track disease onset and progression in neurological disorders or nervous system injury and assess responses to therapeutic interventions. Any of the five Nf subunits - neurofilament light chain (NfL), neurofilament medium chain (NfM), neurofilament heavy chain (NfH), alpha-internexin (INA) and peripherin (PRPH) may be altered in a given neuropathological condition. In familial and sporadic Alzheimer's disease (AD), plasma NfL levels may rise as early as 22 years before clinical onset in familial AD and 10 years before sporadic AD. The major determinants of elevated levels of NfPs and degradation fragments in CSF and blood are the magnitude of damaged or degenerating axons of fiber tracks, the affected axon caliber sizes and the rate of release of NfP and fragments at different stages of a given neurological disease or condition directly or indirectly affecting central nervous system (CNS) and/or peripheral nervous system (PNS). NfPs are rapidly emerging as transformative blood biomarkers in neurology providing novel insights into a wide range of neurological diseases and advancing clinical trials. Here we summarize the current understanding of intracellular NfP physiology, pathophysiology and extracellular kinetics of NfPs in biofluids and review the value and limitations of NfPs and degradation fragments as biomarkers of neurodegeneration and neuronal injury.
Collapse
Affiliation(s)
- Aidong Yuan
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Department of Psychiatry, NYU Neuroscience Institute, New York, NY, United States
| | - Ralph A. Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Department of Psychiatry, NYU Neuroscience Institute, New York, NY, United States
- Department of Cell Biology, New York University Grossman School of Medicine, (NYU), Neuroscience Institute, New York, NY, United States
| |
Collapse
|
24
|
Chen L, van Zijl PC, Wei Z, Lu H, Duan W, Wong PC, Li T, Xu J. Early detection of Alzheimer's disease using creatine chemical exchange saturation transfer magnetic resonance imaging. Neuroimage 2021; 236:118071. [PMID: 33878375 PMCID: PMC8321389 DOI: 10.1016/j.neuroimage.2021.118071] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/07/2021] [Accepted: 04/11/2021] [Indexed: 01/29/2023] Open
Abstract
Detecting Alzheimer's disease (AD) at an early stage brings a lot of benefits including disease management and actions to slow the progression of the disease. Here, we demonstrate that reduced creatine chemical exchange saturation transfer (CrCEST) contrast has the potential to serve as a new biomarker for early detection of AD. The results on wild type (WT) mice and two age-matched AD models, namely tauopathy (Tau) and Aβ amyloidosis (APP), indicated that CrCEST contrasts of the cortex and corpus callosum in the APP and Tau mice were significantly reduced compared to WT counterpart at an early stage (6-7 months) (p < 0.011). Two main causes of the reduced CrCEST contrast, i.e. cerebral pH and creatine concentration, were investigated. From phantom and hypercapnia experiments, CrCEST showed excellent sensitivity to pH variations. From MRS results, the creatine concentration in WT and AD mouse brain was equivalent, which suggests that the reduced CrCEST contrast was dominated by cerebral pH change involved in the progression of AD. Immunohistochemical analysis revealed that the abnormal cerebral pH in AD mice may relate to neuroinflammation, a known factor that can cause pH reduction.
Collapse
Affiliation(s)
- Lin Chen
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, School of Electronic Science and Engineering, National Model Microelectronics College, Xiamen University, Xiamen, China
| | - Peter C.M. van Zijl
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhiliang Wei
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hanzhang Lu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Philip C. Wong
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tong Li
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiadi Xu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
25
|
Michno W, Stringer KM, Enzlein T, Passarelli MK, Escrig S, Vitanova K, Wood J, Blennow K, Zetterberg H, Meibom A, Hopf C, Edwards FA, Hanrieder J. Following spatial Aβ aggregation dynamics in evolving Alzheimer's disease pathology by imaging stable isotope labeling kinetics. SCIENCE ADVANCES 2021; 7:7/25/eabg4855. [PMID: 34134980 PMCID: PMC8208724 DOI: 10.1126/sciadv.abg4855] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/04/2021] [Indexed: 05/04/2023]
Abstract
β-Amyloid (Aβ) plaque formation is the major pathological hallmark of Alzheimer's disease (AD) and constitutes a potentially critical, early inducer driving AD pathogenesis as it precedes other pathological events and cognitive symptoms by decades. It is therefore critical to understand how Aβ pathology is initiated and where and when distinct Aβ species aggregate. Here, we used metabolic isotope labeling in APPNL-G-F knock-in mice together with mass spectrometry imaging to monitor the earliest seeds of Aβ deposition through ongoing plaque development. This allowed visualizing Aβ aggregation dynamics within single plaques across different brain regions. We show that formation of structurally distinct plaques is associated with differential Aβ peptide deposition. Specifically, Aβ1-42 is forming an initial core structure followed by radial outgrowth and late secretion and deposition of Aβ1-38. These data describe a detailed picture of the earliest events of precipitating amyloid pathology at scales not previously possible.
Collapse
Affiliation(s)
- Wojciech Michno
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Katie M Stringer
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Thomas Enzlein
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, Mannheim, Germany
| | - Melissa K Passarelli
- Laboratory of Biological Geochemistry, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Department of Chemistry and Biochemistry, Concordia University, Montréal, Québec, Canada
| | - Stephane Escrig
- Laboratory of Biological Geochemistry, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Karina Vitanova
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Jack Wood
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
| | - Anders Meibom
- Laboratory of Biological Geochemistry, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Center for Advanced Surface Analysis, Institute of Earth Sciences, University of Lausanne, Lausanne, Switzerland
| | - Carsten Hopf
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, Mannheim, Germany
| | - Frances A Edwards
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
26
|
Beuckmann CT, Suzuki H, Musiek ES, Ueno T, Sato T, Bando M, Osada Y, Moline M. Evaluation of SAMP8 Mice as a Model for Sleep-Wake and Rhythm Disturbances Associated with Alzheimer's Disease: Impact of Treatment with the Dual Orexin (Hypocretin) Receptor Antagonist Lemborexant. J Alzheimers Dis 2021; 81:1151-1167. [PMID: 33843668 PMCID: PMC8293654 DOI: 10.3233/jad-201054] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background: Many patients with Alzheimer’s disease (AD) display circadian rhythm and sleep-wake disturbances. However, few mouse AD models exhibit these disturbances. Lemborexant, a dual orexin receptor antagonist, is under development for treating circadian rhythm disorders in dementia. Objective: Evaluation of senescence-accelerated mouse prone-8 (SAMP8) mice as a model for sleep-wake and rhythm disturbances in AD and the effect of lemborexant by assessing sleep-wake/diurnal rhythm behavior. Methods: SAMP8 and control senescence-accelerated mouse resistant-1 (SAMR1) mice received vehicle or lemborexant at light onset; plasma lemborexant and diurnal cerebrospinal fluid (CSF) orexin concentrations were assessed. Sleep-wake behavior and running wheel activity were evaluated. Results: Plasma lemborexant concentrations were similar between strains. The peak/nadir timing of CSF orexin concentrations were approximately opposite between strains. During lights-on, SAMP8 mice showed less non-rapid eye movement (non-REM) and REM sleep than SAMR1 mice. Lemborexant treatment normalized wakefulness/non-REM sleep in SAMP8 mice. During lights-off, lemborexant-treated SAMR1 mice showed increased non-REM sleep; lemborexant-treated SAMP8 mice displayed increased wakefulness. SAMP8 mice showed differences in electroencephalogram architecture versus SAMR1 mice. SAMP8 mice exhibited more running wheel activity during lights-on. Lemborexant treatment reduced activity during lights-on and increased activity in the latter half of lights-off, demonstrating a corrective effect on overall diurnal rhythm. Lemborexant delayed the acrophase of activity in both strains by approximately 1 hour. Conclusion: SAMP8 mice display several aspects of sleep-wake and rhythm disturbances in AD, notably mistimed activity. These findings provide some preclinical rationale for evaluating lemborexant in patients with AD who experience sleep-wake and rhythm disturbances.
Collapse
Affiliation(s)
| | | | - Erik S Musiek
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | | |
Collapse
|
27
|
Powers Carson J. A Simpler and Sensitive Mass Spectrometry Method for Quantitation of Plasma Amyloid Peptides? J Appl Lab Med 2021; 6:816-819. [PMID: 33837388 DOI: 10.1093/jalm/jfab019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/05/2021] [Indexed: 11/14/2022]
Affiliation(s)
- Jennifer Powers Carson
- Department of Medicine; Division of Endocrinology, Metabolism, and Lipid Research, Core Laboratory for Clinical Studies, Washington University, St Louis, MO, USA
| |
Collapse
|
28
|
Arber C, Alatza A, Leckey CA, Paterson RW, Zetterberg H, Wray S. Mass spectrometry analysis of tau and amyloid-beta in iPSC-derived models of Alzheimer's disease and dementia. J Neurochem 2021; 159:305-317. [PMID: 33539581 PMCID: PMC8613538 DOI: 10.1111/jnc.15315] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/18/2021] [Accepted: 01/25/2021] [Indexed: 12/11/2022]
Abstract
Induced pluripotent stem cell (iPSC) technology enables the generation of human neurons in vitro, which contain the precise genome of the cell donor, therefore permitting the generation of disease models from individuals with a disease-associated genotype of interest. This approach has been extensively used to model inherited forms of Alzheimer's disease and frontotemporal dementia. The combination of iPSC-derived neuronal models with targeted mass spectrometry analysis has provided unprecedented insights into the regulation of specific proteins in human neuronal physiology and pathology. For example enabling investigations into tau and APP/Aβ, specifically: protein isoform expression, relative levels of cleavage fragments, aggregated species and functionally critical post-translational modifications. The use of mass spectrometry has enabled a determination of how closely iPSC-derived models recapitulate disease profiles observed in the human brain. This review will highlight the progress to date in studies using iPSCs and mass spectrometry to model Alzheimer's disease and dementia. We go on to convey our optimism, as studies in the near future will make use of this precedent, together with novel techniques such as genome editing and stable isotope labelling, to provide real progress towards an in depth understanding of early neurodegenerative processes and development of novel therapeutic agents.
Collapse
Affiliation(s)
- Charles Arber
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Argyro Alatza
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Claire A Leckey
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK.,Translational Mass Spectrometry Research Group, UCL Great Ormond Street Institute of Child Health, University College London, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Ross W Paterson
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK.,UK Dementia Research Institute at UCL, London, UK.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Selina Wray
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
29
|
Chen X, Salehi A, Pearn ML, Overk C, Nguyen PD, Kleschevnikov AM, Maccecchini M, Mobley WC. Targeting increased levels of APP in Down syndrome: Posiphen-mediated reductions in APP and its products reverse endosomal phenotypes in the Ts65Dn mouse model. Alzheimers Dement 2021; 17:271-292. [PMID: 32975365 PMCID: PMC7984396 DOI: 10.1002/alz.12185] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Recent clinical trials targeting amyloid beta (Aβ) and tau in Alzheimer's disease (AD) have yet to demonstrate efficacy. Reviewing the hypotheses for AD pathogenesis and defining possible links between them may enhance insights into both upstream initiating events and downstream mechanisms, thereby promoting discovery of novel treatments. Evidence that in Down syndrome (DS), a population markedly predisposed to develop early onset AD, increased APP gene dose is necessary for both AD neuropathology and dementia points to normalization of the levels of the amyloid precursor protein (APP) and its products as a route to further define AD pathogenesis and discovering novel treatments. BACKGROUND AD and DS share several characteristic manifestations. DS is caused by trisomy of whole or part of chromosome 21; this chromosome contains about 233 protein-coding genes, including APP. Recent evidence points to a defining role for increased expression of the gene for APP and for its 99 amino acid C-terminal fragment (C99, also known as β-CTF) in dysregulating the endosomal/lysosomal system. The latter is critical for normal cellular function and in neurons for transmitting neurotrophic signals. NEW/UPDATED HYPOTHESIS We hypothesize that the increase in APP gene dose in DS initiates a process in which increased levels of full-length APP (fl-APP) and its products, including β-CTF and possibly Aβ peptides (Aβ42 and Aβ40), drive AD pathogenesis through an endosome-dependent mechanism(s), which compromises transport of neurotrophic signals. To test this hypothesis, we carried out studies in the Ts65Dn mouse model of DS and examined the effects of Posiphen, an orally available small molecule shown in prior studies to reduce fl-APP. In vitro, Posiphen lowered fl-APP and its C-terminal fragments, reversed Rab5 hyperactivation and early endosome enlargement, and restored retrograde transport of neurotrophin signaling. In vivo, Posiphen treatment (50 mg/kg/d, 26 days, intraperitoneal [i.p.]) of Ts65Dn mice was well tolerated and demonstrated no adverse effects in behavior. Treatment resulted in normalization of the levels of fl-APP, C-terminal fragments and small reductions in Aβ species, restoration to normal levels of Rab5 activity, reduced phosphorylated tau (p-tau), and reversed deficits in TrkB (tropomyosin receptor kinase B) activation and in the Akt (protein kinase B [PKB]), ERK (extracellular signal-regulated kinase), and CREB (cAMP response element-binding protein) signaling pathways. Remarkably, Posiphen treatment also restored the level of choline acetyltransferase protein to 2N levels. These findings support the APP gene dose hypothesis, point to the need for additional studies to explore the mechanisms by which increased APP gene expression acts to increase the risk for AD in DS, and to possible utility of treatments to normalize the levels of APP and its products for preventing AD in those with DS. MAJOR CHALLENGES FOR THE HYPOTHESIS Important unanswered questions are: (1) When should one intervene in those with DS; (2) would an APP-based strategy have untoward consequences on possible adaptive changes induced by chronically increased APP gene dose; (3) do other genes present on chromosome 21, or on other chromosomes whose expression is dysregulated in DS, contribute to AD pathogenesis; and (4) can one model strategies that combine the use of an APP-based treatment with those directed at other AD phenotypes including p-tau and inflammation. LINKAGE TO OTHER MAJOR THEORIES The APP gene dose hypothesis interfaces with the amyloid cascade hypothesis of AD as well as with the genetic and cell biological observations that support it. Moreover, upregulation of fl-APP protein and products may drive downstream events that dysregulate tau homeostasis and inflammatory responses that contribute to propagation of AD pathogenesis.
Collapse
Affiliation(s)
- Xu‐Qiao Chen
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Ahmad Salehi
- Department of Psychiatry & Behavioral SciencesStanford Medical SchoolPalo AltoCaliforniaUSA
| | - Matthew L. Pearn
- Department of AnesthesiologyUniversity of California San Diego, School of MedicineLa JollaCaliforniaUSA
- V.A. San Diego Healthcare SystemSan DiegoCaliforniaUSA
| | - Cassia Overk
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Phuong D. Nguyen
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | | | | | - William C. Mobley
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
30
|
Gafson AR, Barthélemy NR, Bomont P, Carare RO, Durham HD, Julien JP, Kuhle J, Leppert D, Nixon RA, Weller RO, Zetterberg H, Matthews PM. Neurofilaments: neurobiological foundations for biomarker applications. Brain 2020; 143:1975-1998. [PMID: 32408345 DOI: 10.1093/brain/awaa098] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/20/2019] [Accepted: 01/20/2020] [Indexed: 12/11/2022] Open
Abstract
Interest in neurofilaments has risen sharply in recent years with recognition of their potential as biomarkers of brain injury or neurodegeneration in CSF and blood. This is in the context of a growing appreciation for the complexity of the neurobiology of neurofilaments, new recognition of specialized roles for neurofilaments in synapses and a developing understanding of mechanisms responsible for their turnover. Here we will review the neurobiology of neurofilament proteins, describing current understanding of their structure and function, including recently discovered evidence for their roles in synapses. We will explore emerging understanding of the mechanisms of neurofilament degradation and clearance and review new methods for future elucidation of the kinetics of their turnover in humans. Primary roles of neurofilaments in the pathogenesis of human diseases will be described. With this background, we then will review critically evidence supporting use of neurofilament concentration measures as biomarkers of neuronal injury or degeneration. Finally, we will reflect on major challenges for studies of the neurobiology of intermediate filaments with specific attention to identifying what needs to be learned for more precise use and confident interpretation of neurofilament measures as biomarkers of neurodegeneration.
Collapse
Affiliation(s)
- Arie R Gafson
- Department of Brain Sciences, Imperial College, London, UK
| | - Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Pascale Bomont
- ATIP-Avenir team, INM, INSERM, Montpellier University, Montpellier, France
| | - Roxana O Carare
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Heather D Durham
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
| | - Jean-Pierre Julien
- Department of Psychiatry and Neuroscience, Laval University, Quebec, Canada.,CERVO Brain Research Center, 2601 Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - David Leppert
- Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ralph A Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA.,Departments of Psychiatry, New York University School of Medicine, New York, NY, 10016, USA.,Neuroscience Institute, New York University School of Medicine, New York, NY, 10016, USA.,Department of Cell Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Roy O Weller
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Henrik Zetterberg
- University College London Queen Square Institute of Neurology, London, UK.,UK Dementia Research Institute at University College London, London, UK.,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
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College, London, UK.,UK Dementia Research Institute at Imperial College, London
| |
Collapse
|
31
|
Cummings J. Drug Development for Psychotropic, Cognitive-Enhancing, and Disease-Modifying Treatments for Alzheimer's Disease. J Neuropsychiatry Clin Neurosci 2020; 33:3-13. [PMID: 33108950 PMCID: PMC7989572 DOI: 10.1176/appi.neuropsych.20060152] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with limited available therapies. There is progress in developing treatments for neuropsychiatric indications in AD, including agitation, psychosis, apathy, and sleep disorders. Candidate therapies progress from nonclinical and animal assessment to trials in normal volunteers (phase 1), small phase-2 trials, and larger confirmatory phase-3 trials. Biomarkers play an increasingly important role in selecting participants, stratifying populations, demonstrating target engagement, supporting disease modification, and monitoring safety. There are currently 121 agents in clinical trials, including treatments for neuropsychiatric symptoms, cognition enhancement, and disease progression. There are 27 agents in phase-1 trials, 65 in phase-2 trials, and 29 in phase-3 trials. Most of the agents in trials (80%) target disease modification. Treatments are being assessed in secondary prevention trials with cognitively normal individuals at high risk for the development of AD. There is progress in target diversification, trial designs, outcome measures, biomarkers, and trial population definitions that promise to accelerate developing new therapies for those with or at risk for AD.
Collapse
Affiliation(s)
- Jeffrey Cummings
- The Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, and the Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas
| |
Collapse
|
32
|
It's complicated: The relationship between sleep and Alzheimer's disease in humans. Neurobiol Dis 2020; 144:105031. [PMID: 32738506 DOI: 10.1016/j.nbd.2020.105031] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/21/2020] [Accepted: 07/26/2020] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by an asymptomatic period of amyloid-β (Aβ) deposition as insoluble extracellular plaque, intracellular tau aggregation, neuronal and synaptic loss, and subsequent cognitive dysfunction and dementia. A growing public health crisis, the worldwide prevalence of AD is expected to rise from 46.8 million individuals affected in 2015 to 131.5 million in 2050. Sleep disturbances have been associated with increased future risk of AD. A bi-directional relationship is hypothesized between sleep and AD with sleep disturbances as either markers for AD pathology and/or a mechanism mediating increased risk of AD. In this review, the evidence in humans supporting this complex relationship between sleep and AD will be discussed as well as the therapeutic potential and challenges of treating sleep disturbances to prevent or delay the onset of AD.
Collapse
|
33
|
Wilke C, Haas E, Reetz K, Faber J, Garcia‐Moreno H, Santana MM, van de Warrenburg B, Hengel H, Lima M, Filla A, Durr A, Melegh B, Masciullo M, Infante J, Giunti P, Neumann M, de Vries J, Pereira de Almeida L, Rakowicz M, Jacobi H, Schüle R, Kaeser SA, Kuhle J, Klockgether T, Schöls L, Barro C, Hübener‐Schmid J, Synofzik M. Neurofilaments in spinocerebellar ataxia type 3: blood biomarkers at the preataxic and ataxic stage in humans and mice. EMBO Mol Med 2020; 12:e11803. [PMID: 32510847 PMCID: PMC7338806 DOI: 10.15252/emmm.201911803] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 05/05/2020] [Accepted: 05/10/2020] [Indexed: 12/13/2022] Open
Abstract
With molecular treatments coming into reach for spinocerebellar ataxia type 3 (SCA3), easily accessible, cross-species validated biomarkers for human and preclinical trials are warranted, particularly for the preataxic disease stage. We assessed serum levels of neurofilament light (NfL) and phosphorylated neurofilament heavy (pNfH) in ataxic and preataxic subjects of two independent multicentric SCA3 cohorts and in a SCA3 knock-in mouse model. Ataxic SCA3 subjects showed increased levels of both NfL and pNfH. In preataxic subjects, NfL levels increased with proximity to the individual expected onset of ataxia, with significant NfL elevations already 7.5 years before onset. Cross-sectional NfL levels correlated with both disease severity and longitudinal disease progression. Blood NfL and pNfH increases in human SCA3 were each paralleled by similar changes in SCA3 knock-in mice, here also starting already at the presymptomatic stage, closely following ataxin-3 aggregation and preceding Purkinje cell loss in the brain. Blood neurofilaments, particularly NfL, might thus provide easily accessible, cross-species validated biomarkers in both ataxic and preataxic SCA3, associated with earliest neuropathological changes, and serve as progression, proximity-to-onset and, potentially, treatment-response markers in both human and preclinical SCA3 trials.
Collapse
|
34
|
Ehrenberg AJ, Khatun A, Coomans E, Betts MJ, Capraro F, Thijssen EH, Senkevich K, Bharucha T, Jafarpour M, Young PNE, Jagust W, Carter SF, Lashley T, Grinberg LT, Pereira JB, Mattsson-Carlgren N, Ashton NJ, Hanrieder J, Zetterberg H, Schöll M, Paterson RW. Relevance of biomarkers across different neurodegenerative diseases. ALZHEIMERS RESEARCH & THERAPY 2020; 12:56. [PMID: 32404143 PMCID: PMC7222479 DOI: 10.1186/s13195-020-00601-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/16/2020] [Indexed: 01/11/2023]
Abstract
Background The panel of fluid- and imaging-based biomarkers available for neurodegenerative disease research is growing and has the potential to close important gaps in research and the clinic. With this growth and increasing use, appropriate implementation and interpretation are paramount. Various biomarkers feature nuanced differences in strengths, limitations, and biases that must be considered when investigating disease etiology and clinical utility. For example, neuropathological investigations of Alzheimer’s disease pathogenesis can fall in disagreement with conclusions reached by biomarker-based investigations. Considering the varied strengths, limitations, and biases of different research methodologies and approaches may help harmonize disciplines within the neurodegenerative disease field. Purpose of review Along with separate review articles covering fluid and imaging biomarkers in this issue of Alzheimer’s Research and Therapy, we present the result of a discussion from the 2019 Biomarkers in Neurodegenerative Diseases course at the University College London. Here, we discuss themes of biomarker use in neurodegenerative disease research, commenting on appropriate use, interpretation, and considerations for implementation across different neurodegenerative diseases. We also draw attention to areas where biomarker use can be combined with other disciplines to understand issues of pathophysiology and etiology underlying dementia. Lastly, we highlight novel modalities that have been proposed in the landscape of neurodegenerative disease research and care.
Collapse
Affiliation(s)
- Alexander J Ehrenberg
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA. .,Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA. .,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA.
| | - Ayesha Khatun
- Dementia Research Centre, University College London Institute of Neurology, London, UK
| | - Emma Coomans
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Matthew J Betts
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Federica Capraro
- The Francis Crick Institute, London, UK.,Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, London, UK
| | - Elisabeth H Thijssen
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.,Department of Clinical Chemistry, Amsterdam UMC, Amsterdam, The Netherlands
| | - Konstantin Senkevich
- Petersburg Nuclear Physics Institute names by B.P. Konstantinov of National Research Center, Kurchatov Institute, St. Petersburg, Russia.,First Pavlov State Medical University of St. Petersburg, St. Petersburg, Russia
| | - Tehmina Bharucha
- Oxford Glycobiology Institute, Department of Biochemistry , University of Oxford, Oxford, UK
| | - Mehrsa Jafarpour
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK
| | - Peter N E Young
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Center for Molecular and Translational Medicine, Lund University, Lund, Sweden
| | - William Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA.,Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Stephen F Carter
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK.,Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Tammaryn Lashley
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK.,Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - Lea T Grinberg
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.,University of São Paulo Medical School, São Paulo, Brazil.,Global Brain Health Institute, San Francisco, CA, USA
| | - Joana B Pereira
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Clinical Memory Research Unit, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Center for Molecular and Translational Medicine, Lund University, Lund, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Center for Molecular and Translational Medicine, Lund University, Lund, 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
| | - Jörg Hanrieder
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK.,Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK.,Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,UK Dementia Research Institute at University College London, London, UK
| | - Michael Schöll
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK.,Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Memory Research Unit, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Ross W Paterson
- Dementia Research Centre, University College London Institute of Neurology, London, UK
| |
Collapse
|
35
|
Castor KJ, Shenoi S, Edminster SP, Tran T, King KS, Chui H, Pogoda JM, Fonteh AN, Harrington MG. Urine dicarboxylic acids change in pre-symptomatic Alzheimer's disease and reflect loss of energy capacity and hippocampal volume. PLoS One 2020; 15:e0231765. [PMID: 32298384 PMCID: PMC7162508 DOI: 10.1371/journal.pone.0231765] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Non-invasive biomarkers will enable widespread screening and early diagnosis of Alzheimer’s disease (AD). We hypothesized that the considerable loss of brain tissue in AD will result in detection of brain lipid components in urine, and that these will change in concert with CSF and brain biomarkers of AD. We examined urine dicarboxylic acids (DCA) of carbon length 3–10 to reflect products of oxidative damage and energy generation or balance that may account for changes in brain function in AD. Mean C4-C5 DCAs were lower and mean C7-C10 DCAs were higher in the urine from AD compared to cognitively healthy (CH) individuals. Moreover, mean C4-C5 DCAs were lower and mean C7-C9 were higher in urine from CH individuals with abnormal compared to normal CSF amyloid and Tau levels; i.e., the apparent urine changes in AD also appeared to be present in CH individuals that have CSF risk factors of early AD pathology. In examining the relationship between urine DCAs and AD biomarkers, we found short chain DCAs positively correlated with CSF Aβ42, while C7-C10 DCAs negatively correlated with CSF Aβ42 and positively correlated with CSF Tau levels. Furthermore, we found a negative correlation of C7-C10 DCAs with hippocampal volume (p < 0.01), which was not found in the occipital volume. Urine measures of DCAs have an 82% ability to predict cognitively healthy participants with normal CSF amyloid/Tau. These data suggest that urine measures of increased lipoxidation and dysfunctional energy balance reflect early AD pathology from brain and CSF biomarkers. Measures of urine DCAs may contribute to personalized healthcare by indicating AD pathology and may be utilized to explore population wellness or monitor the efficacy of therapies in clinical trials.
Collapse
Affiliation(s)
- K. J. Castor
- Neurosciences, Huntington Medical Research Institutes, Pasadena, CA, United States of America
| | - S. Shenoi
- Neurosciences, Huntington Medical Research Institutes, Pasadena, CA, United States of America
| | - S. P. Edminster
- Neurosciences, Huntington Medical Research Institutes, Pasadena, CA, United States of America
| | - T. Tran
- Clinical MR Unit, Huntington Medical Research Institutes, Pasadena, CA, United States of America
| | - K. S. King
- Clinical MR Unit, Huntington Medical Research Institutes, Pasadena, CA, United States of America
| | - H. Chui
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - J. M. Pogoda
- Cipher Biostatistics & Reporting, Reno, NV, United States of America
| | - A. N. Fonteh
- Neurosciences, Huntington Medical Research Institutes, Pasadena, CA, United States of America
- * E-mail: (ANF); (MGH)
| | - M. G. Harrington
- Neurosciences, Huntington Medical Research Institutes, Pasadena, CA, United States of America
- * E-mail: (ANF); (MGH)
| |
Collapse
|
36
|
Chatterjee K, Carman-Esparza CM, Munson JM. Methods to measure, model and manipulate fluid flow in brain. J Neurosci Methods 2020; 333:108541. [PMID: 31838183 PMCID: PMC7607555 DOI: 10.1016/j.jneumeth.2019.108541] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/01/2019] [Accepted: 12/04/2019] [Indexed: 01/15/2023]
Abstract
The brain consists of a complex network of cells and matrix that is cushioned and nourished by multiple types of fluids: cerebrospinal fluid, blood, and interstitial fluid. The movement of these fluids through the tissues has recently gained more attention due to implications in Alzheimer's Disease and glioblastoma. Therefore, methods to study these fluid flows are necessary and timely for the current study of neuroscience. Imaging modalities such as magnetic resonance imaging have been used clinically and pre-clinically to image flows in healthy and diseased brains. These measurements have been used to both parameterize and validate models of fluid flow both computational and in vitro. Both of these models can elucidate the changes to fluid flow that occur during disease and can assist in linking the compartments of fluid flow with one another, a difficult challenge experimentally. In vitro models, though in limited use with fluid flow, allow the examination of cellular responses to physiological flow. To determine causation, in vivo methods have been developed to manipulate flow, including both physical and pharmacological manipulations, at each point of fluid movement of origination resulting in exciting findings in the preclinical setting. With new targets, such as the brain-draining lymphatics and glymphatic system, fluid flow and tissue drainage within the brain is an exciting and growing research area. In this review, we discuss the methods that currently exist to examine and test hypotheses related to fluid flow in the brain as we attempt to determine its impact on neural function.
Collapse
Affiliation(s)
- Krishnashis Chatterjee
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Cora M Carman-Esparza
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jennifer M Munson
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States.
| |
Collapse
|
37
|
Abrahamson EE, Ikonomovic MD. Brain injury-induced dysfunction of the blood brain barrier as a risk for dementia. Exp Neurol 2020; 328:113257. [PMID: 32092298 DOI: 10.1016/j.expneurol.2020.113257] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/31/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) is a complex and dynamic physiological interface between brain parenchyma and cerebral vasculature. It is composed of closely interacting cells and signaling molecules that regulate movement of solutes, ions, nutrients, macromolecules, and immune cells into the brain and removal of products of normal and abnormal brain cell metabolism. Dysfunction of multiple components of the BBB occurs in aging, inflammatory diseases, traumatic brain injury (TBI, severe or mild repetitive), and in chronic degenerative dementing disorders for which aging, inflammation, and TBI are considered risk factors. BBB permeability changes after TBI result in leakage of serum proteins, influx of immune cells, perivascular inflammation, as well as impairment of efflux transporter systems and accumulation of aggregation-prone molecules involved in hallmark pathologies of neurodegenerative diseases with dementia. In addition, cerebral vascular dysfunction with persistent alterations in cerebral blood flow and neurovascular coupling contribute to brain ischemia, neuronal degeneration, and synaptic dysfunction. While the idea of TBI as a risk factor for dementia is supported by many shared pathological features, it remains a hypothesis that needs further testing in experimental models and in human studies. The current review focusses on pathological mechanisms shared between TBI and neurodegenerative disorders characterized by accumulation of pathological protein aggregates, such as Alzheimer's disease and chronic traumatic encephalopathy. We discuss critical knowledge gaps in the field that need to be explored to clarify the relationship between TBI and risk for dementia and emphasize the need for longitudinal in vivo studies using imaging and biomarkers of BBB dysfunction in people with single or multiple TBI.
Collapse
Affiliation(s)
- Eric E Abrahamson
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Milos D Ikonomovic
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
38
|
Bassendine MF, Taylor-Robinson SD, Fertleman M, Khan M, Neely D. Is Alzheimer's Disease a Liver Disease of the Brain? J Alzheimers Dis 2020; 75:1-14. [PMID: 32250293 PMCID: PMC7306895 DOI: 10.3233/jad-190848] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
Clinical specialization is not only a force for progress, but it has also led to the fragmentation of medical knowledge. The focus of research in the field of Alzheimer's disease (AD) is neurobiology, while hepatologists focus on liver diseases and lipid specialists on atherosclerosis. This article on AD focuses on the role of the liver and lipid homeostasis in the development of AD. Amyloid-β (Aβ) deposits accumulate as plaques in the brain of an AD patient long before cognitive decline is evident. Aβ generation is a normal physiological process; the steady-state level of Aβ in the brain is determined by balance between Aβ production and its clearance. We present evidence suggesting that the liver is the origin of brain Aβ deposits and that it is involved in peripheral clearance of circulating Aβ in the blood. Hence the liver could be targeted to decrease Aβ production or increase peripheral clearance.
Collapse
Affiliation(s)
- Margaret F. Bassendine
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
- Department of Hepatology & Gastroenterology, Division of Surgery and Cancer, Imperial College London, St Mary’s Campus, UK
| | - Simon D. Taylor-Robinson
- Department of Hepatology & Gastroenterology, Division of Surgery and Cancer, Imperial College London, St Mary’s Campus, UK
| | - Michael Fertleman
- Department of Hepatology & Gastroenterology, Division of Surgery and Cancer, Imperial College London, St Mary’s Campus, UK
- Department of Bioengineering, Imperial College London, UK
| | - Michael Khan
- University of Warwick & University Hospitals of Coventry and Warwickshire NHS Trust, UK
| | - Dermot Neely
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
- Department of Blood Sciences, Newcastle upon Tyne Hospitals NHS Foundation Trust, UK
| |
Collapse
|