1
|
Singh M, Ali H, Renuka Jyothi S, Kaur I, Kumar S, Sharma N, Siva Prasad GV, Pramanik A, Hassan Almalki W, Imran M. Tau proteins and senescent Cells: Targeting aging pathways in Alzheimer's disease. Brain Res 2024; 1844:149165. [PMID: 39155034 DOI: 10.1016/j.brainres.2024.149165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease characterized by abnormal accumulation of tau proteins and amyloid-β, leading to neuronal death and cognitive impairment. Recent studies have implicated aging pathways, including dysregulation of tau and cellular senescence in AD pathogenesis. In AD brains, tau protein, which normally stabilizes microtubules, becomes hyperphosphorylated and forms insoluble neurofibrillary tangles. These tau aggregates impair neuronal function and are propagated across the brain's neurocircuitry. Meanwhile, the number of senescent cells accumulating in the aging brain is rising, releasing a pro-inflammatory SASP responsible for neuroinflammation and neurodegeneration. This review explores potential therapeutic interventions for AD targeting tau protein and senescent cells, and tau -directed compounds, senolytics, eliminating senescent cells, and agents that modulate the SASP-senomodulators. Ultimately, a combined approach that incorporates tau-directed medications and targeted senescent cell-based therapies holds promise for reducing the harmful impact of AD's shared aging pathways.
Collapse
Affiliation(s)
- Mahaveer Singh
- School of Pharmacy and Technology Management, SVKMs NMIMS University, Shirpur campus, Maharastra India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan-303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali 140307, Punjab, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
2
|
Cummings JL, Gonzalez MI, Pritchard MC, May PC, Toledo-Sherman LM, Harris GA. The therapeutic landscape of tauopathies: challenges and prospects. Alzheimers Res Ther 2023; 15:168. [PMID: 37803386 PMCID: PMC10557207 DOI: 10.1186/s13195-023-01321-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/28/2023] [Indexed: 10/08/2023]
Abstract
Tauopathies are a group of neurodegenerative disorders characterized by the aggregation of the microtubule-associated protein tau. Aggregates of misfolded tau protein are believed to be implicated in neuronal death, which leads to a range of symptoms including cognitive decline, behavioral change, dementia, and motor deficits. Currently, there are no effective treatments for tauopathies. There are four clinical candidates in phase III trials and 16 in phase II trials. While no effective treatments are currently approved, there is increasing evidence to suggest that various therapeutic approaches may slow the progression of tauopathies or improve symptoms. This review outlines the landscape of therapeutic drugs (indexed through February 28, 2023) that target tau pathology and describes drug candidates in clinical development as well as those in the discovery and preclinical phases. The review also contains information on notable therapeutic programs that are inactive or that have been discontinued from development.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada, Las Vegas (UNLV), Henderson, NV, USA
| | | | | | - Patrick C May
- ADvantage Neuroscience Consulting LLC, Fort Wayne, IN, USA
| | | | - Glenn A Harris
- Rainwater Charitable Foundation, 777 Main Street, Suite 2250, Fort Worth, TX, 76102, USA.
| |
Collapse
|
3
|
Hu X, Meier M, Pruessner J. Challenges and opportunities of diagnostic markers of Alzheimer's disease based on structural magnetic resonance imaging. Brain Behav 2023; 13:e2925. [PMID: 36795041 PMCID: PMC10013953 DOI: 10.1002/brb3.2925] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/04/2023] [Indexed: 02/17/2023] Open
Abstract
OBJECTIVES This article aimed to carry out a narrative literature review of early diagnostic markers of Alzheimer's disease (AD) based on both micro and macro levels of pathology, indicating the shortcomings of current biomarkers and proposing a novel biomarker of structural integrity that associates the hippocampus and adjacent ventricle together. This could help to reduce the influence of individual variety and improve the accuracy and validity of structural biomarker. METHODS This review was based on presenting comprehensive background of early diagnostic markers of AD. We have compiled those markers into micro level and macro level, and discussed the advantages and disadvantages of them. Eventually the ratio of gray matter volume to ventricle volume was put forward. RESULTS The costly methodologies and related high patient burden of "micro" biomarkers (cerebrospinal fluid biomarkers) hinder the implementation in routine clinical examination. In terms of "macro" biomarkers- hippocampal volume (HV), there is a large variation of it among population, which undermines its validity Considering the gray matter atrophies while the adjacent ventricular volume enlarges, we assume the hippocampal to ventricle ratio (HVR) is a more reliable marker than HV alone the emerging evidence showed hippocampal to ventricle ratio predicts memory functions better than HV alone in elderly sample. CONCLUSIONS The ratio between gray matter structures and adjacent ventricular volumes counts as a promising superior diagnostic marker of early neurodegeneration.
Collapse
Affiliation(s)
- Xiang Hu
- Department of Psychology, University of Konstanz, Konstanz, Germany
| | - Maria Meier
- Department of Psychology, University of Konstanz, Konstanz, Germany
| | - Jens Pruessner
- Department of Psychology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
4
|
Jann K, Boudreau J, Albrecht D, Cen SY, Cabeen RP, Ringman JM, Wang DJ, for the Alzheimer’s Disease Neuroimaging Initiative. FMRI Complexity Correlates with Tau-PET and Cognitive Decline in Late-Onset and Autosomal Dominant Alzheimer's Disease. J Alzheimers Dis 2023; 95:437-451. [PMID: 37599531 PMCID: PMC10578217 DOI: 10.3233/jad-220851] [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] [Accepted: 06/27/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND Neurofibrillary tangle pathology detected with tau-PET correlates closely with neuronal injury and cognitive symptoms in Alzheimer's disease (AD). Complexity of rs-fMRI has been demonstrated to decrease with cognitive decline in AD. OBJECTIVE We hypothesize that the rs-fMRI complexity provides an index for tau-related neuronal injury and cognitive decline in the AD process. METHODS Data was obtained from the Alzheimer's Disease Neuroimaging Initiative (ADNI3) and the Estudio de la Enfermedad de Alzheimer en Jalisciences (EEAJ) study. Associations between tau-PET and rs-fMRI complexity were calculated. Potential pathways relating complexity to cognitive function mediated through tau-PET were assessed by path analysis. RESULTS We found significant negative correlations between rs-fMRI complexity and tau-PET in medial temporal lobe of both cohorts, and associations of rs-fMRI complexity with cognitive scores were mediated through tau-PET. CONCLUSION The association of rs-fMRI complexity with tau-PET and cognition, suggests that a reduction in complexity is indicative of tau-related neuropathology and cognitive decline in AD processes.
Collapse
Affiliation(s)
- Kay Jann
- Laboratory of Functional MRI Technology (LOFT), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Julia Boudreau
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daniel Albrecht
- Laboratory of NeuroImaging (LONI), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Steven Y. Cen
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ryan P. Cabeen
- Laboratory of NeuroImaging (LONI), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John M. Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Danny J.J. Wang
- Laboratory of Functional MRI Technology (LOFT), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | |
Collapse
|
5
|
Demirbilek O, Rekik I. Predicting the evolution trajectory of population-driven connectional brain templates using recurrent multigraph neural networks. Med Image Anal 2023; 83:102649. [PMID: 36257134 DOI: 10.1016/j.media.2022.102649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022]
Abstract
The mapping of the time-dependent evolution of the human brain connectivity using longitudinal and multimodal neuroimaging datasets provides insights into the development of neurological disorders and the way they alter the brain morphology, structure and function over time. Recently, the connectional brain template (CBT) was introduced as a compact representation integrating a population of brain multigraphs, where two brain regions can have multiple connections, into a single graph. Given a population of brain multigraphs observed at a baseline timepoint t1, we aim to learn how to predict the evolution of the population CBT at follow-up timepoints t>t1. Such model will allow us to foresee the evolution of the connectivity patterns of healthy and disordered individuals at the population level. Here we present recurrent multigraph integrator network (ReMI-Net⋆) to forecast population templates at consecutive timepoints from a given single timepoint. In particular, we unprecedentedly design a graph neural network architecture to model the changes in the brain multigraph and identify the biomarkers that differentiate between the typical and atypical populations. Addressing such issues is of paramount importance in diagnosing neurodegenerative disorders at early stages and promoting new clinical studies based on the pinned-down biomarker brain regions or connectivities. In this paper, we demonstrate the design and use of the ReMI-Net⋆ model, which learns both the multigraph node level and time level dependencies concurrently. Thanks to its novel graph convolutional design and normalization layers, ReMI-Net⋆ predicts well-centered, discriminative, and topologically sound connectional templates over time. Additionally, the results show that our model outperforms all benchmarks and state-of-the-art methods by comparing and discovering the atypical connectivity alterations over time. Our ReMI-Net⋆ code is available on GitHub at https://github.com/basiralab/ReMI-Net-Star.
Collapse
Affiliation(s)
- Oytun Demirbilek
- BASIRA lab, Faculty of Computer and Informatics, Istanbul Technical University, Istanbul, Turkey
| | - Islem Rekik
- BASIRA lab, Faculty of Computer and Informatics, Istanbul Technical University, Istanbul, Turkey; School of Science and Engineering, Computing, University of Dundee, UK.
| | | |
Collapse
|
6
|
Leach JM, Edwards LJ, Kana R, Visscher K, Yi N, Aban I, for the Alzheimer’s Disease Neuroimaging Initiative. The spike-and-slab elastic net as a classification tool in Alzheimer's disease. PLoS One 2022; 17:e0262367. [PMID: 35113902 PMCID: PMC8812870 DOI: 10.1371/journal.pone.0262367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/21/2021] [Indexed: 11/18/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia and has received considerable research attention, including using neuroimaging biomarkers to classify patients and/or predict disease progression. Generalized linear models, e.g., logistic regression, can be used as classifiers, but since the spatial measurements are correlated and often outnumber subjects, penalized and/or Bayesian models will be identifiable, while classical models often will not. Many useful models, e.g., the elastic net and spike-and-slab lasso, perform automatic variable selection, which removes extraneous predictors and reduces model variance, but neither model exploits spatial information in selecting variables. Spatial information can be incorporated into variable selection by placing intrinsic autoregressive priors on the logit probabilities of inclusion within a spike-and-slab elastic net framework. We demonstrate the ability of this framework to improve classification performance by using cortical thickness and tau-PET images from the Alzheimer's Disease Neuroimaging Initiative (ADNI) to classify subjects as cognitively normal or having dementia, and by using a simulation study to examine model performance using finer resolution images.
Collapse
Affiliation(s)
- Justin M. Leach
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Lloyd J. Edwards
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Rajesh Kana
- Department of Psychology, University of Alabama, Tuscaloosa, Alabama, United States of America
| | - Kristina Visscher
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Nengjun Yi
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Inmaculada Aban
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | | |
Collapse
|
7
|
Nuño MM, Grill JD, Gillen DL, Alzheimer’s Disease Neuroimaging Initiative. On the design of early-phase Alzheimer's disease clinical trials with cerebrospinal fluid tau outcomes. Clin Trials 2021; 18:714-723. [PMID: 34325548 PMCID: PMC8595611 DOI: 10.1177/17407745211034497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND/AIMS The focus of Alzheimer's disease studies has shifted to earlier disease stages, including mild cognitive impairment. Biomarker inclusion criteria are often incorporated into mild cognitive impairment clinical trials to identify individuals with "prodromal Alzheimer's disease" to ensure appropriate drug targets and enrich for participants likely to develop Alzheimer's disease dementia. The use of these eligibility criteria may affect study power. METHODS We investigated outcome variability and study power in the setting of proof-of-concept prodromal Alzheimer's disease trials that incorporate cerebrospinal fluid levels of total tau (t-tau) and phosphorylated (p-tau) as primary outcomes and how differing biomarker inclusion criteria affect power. We used data from the Alzheimer's Disease Neuroimaging Initiative to model trial scenarios and to estimate the variance and within-subject correlation of total and phosphorylated tau. These estimates were then used to investigate the differences in study power for trials considering these two surrogate outcomes. RESULTS Patient characteristics were similar for all eligibility criteria. The lowest outcome variance and highest within-subject correlation were obtained when phosphorylated tau was used as an eligibility criterion, compared to amyloid beta or total tau, regardless of whether total tau or phosphorylated tau were used as primary outcomes. Power increased when eligibility criteria were broadened to allow for enrollment of subjects with either low amyloid beta or high phosphorylated tau. CONCLUSION Specific biomarker inclusion criteria may impact statistical power in trials using total tau or phosphorylated tau as the primary outcome. In concert with other important considerations such as treatment target and population of clinical interest, these results may have implications to the integrity and efficiency of prodromal Alzheimer's disease trial designs.
Collapse
Affiliation(s)
- Michelle M. Nuño
- Children’s Oncology Group, Monrovia, CA, USA
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Joshua D. Grill
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA
| | - Daniel L. Gillen
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
- Department of Statistics, University of California, Irvine, Irvine, CA, USA
| | | |
Collapse
|
8
|
Tataryn NM, Singh V, Dyke JP, Berk-Rauch HE, Clausen DM, Aronowitz E, Norris EH, Strickland S, Ahn HJ. Vascular endothelial growth factor associated dissimilar cerebrovascular phenotypes in two different mouse models of Alzheimer's Disease. Neurobiol Aging 2021; 107:96-108. [PMID: 34416494 PMCID: PMC8595520 DOI: 10.1016/j.neurobiolaging.2021.07.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 01/14/2023]
Abstract
Vascular perturbations and cerebral hypometabolism are emerging as important components of Alzheimer's disease (AD). While various in vivo imaging modalities have been designed to detect changes of cerebral perfusion and metabolism in AD patients and animal models, study results were often heterogenous with respect to imaging techniques and animal models. We therefore evaluated cerebral perfusion and glucose metabolism of two popular transgenic AD mouse strains, TgCRND8 and 5xFAD, at 7 and 12 months-of-age under identical conditions and analyzed possible molecular mechanisms underlying heterogeneous cerebrovascular phenotypes. Results revealed disparate findings in these two strains, displaying important aspects of AD progression. TgCRND8 mice showed significantly decreased cerebral blood flow and glucose metabolism with unchanged cerebral blood volume (CBV) at 12 months-of-age whereas 5xFAD mice showed unaltered glucose metabolism with significant increase in CBV at 12 months-of-age and a biphasic pattern of early hypoperfusion followed by a rebound to normal cerebral blood flow in late disease. Finally, immunoblotting assays suggested that VEGF dependent vascular tone change may restore normoperfusion and increase CBV in 5xFAD.
Collapse
Affiliation(s)
- Nicholas M Tataryn
- Tri-Institutional Training Program in Laboratory Animal Medicine and Science, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, and The Rockefeller University, New York, New York, USA and Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, Rockefeller University, New York, NY, USA; Division of Comparative Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Vishal Singh
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Jonathan P Dyke
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Hanna E Berk-Rauch
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, Rockefeller University, New York, NY, USA
| | - Dana M Clausen
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Eric Aronowitz
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Erin H Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, Rockefeller University, New York, NY, USA
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, Rockefeller University, New York, NY, USA
| | - Hyung Jin Ahn
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, USA; Brain Health Institute, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
9
|
Kroth H, Oden F, Molette J, Schieferstein H, Gabellieri E, Mueller A, Berndt M, Sreenivasachary N, Serra AM, Capotosti F, Schmitt-Willich H, Hickman D, Pfeifer A, Dinkelborg L, Stephens A. PI-2620 Lead Optimization Highlights the Importance of Off-Target Assays to Develop a PET Tracer for the Detection of Pathological Aggregated Tau in Alzheimer's Disease and Other Tauopathies. J Med Chem 2021; 64:12808-12830. [PMID: 34455780 DOI: 10.1021/acs.jmedchem.1c00861] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The first candidate PI-2014 was tested in healthy controls and subjects with Alzheimer's disease (AD). As PI-2014 displayed off-target binding to monoamine oxidase A (MAO-A), a new lead with improved binding to Tau and decreased MAO-A binding was required. For compound optimization, Tau binding assays based on both human AD brain homogenate and Tau-paired helical filaments were employed. Furthermore, two MAO-A screening assays based on (1) human-recombinant MAO-A and (2) displacement of 2-fluoro-ethyl-harmine from mouse brain homogenate were employed. Removing the N-methyl group from the tricyclic core resulted in compounds displaying improved Tau binding. For the final round of optimization, the cyclic amine substituents were replaced by pyridine derivatives. PI-2620 (2-(2-fluoropyridin-4-yl)-9H-pyrrolo[2,3-b:4,5-c']dipyridine) emerged as a best candidate displaying high Tau binding, low MAO-A binding, high brain uptake, and fast and complete brain washout. Furthermore, PI-2620 showed Tau binding on brain sections from corticobasal degeneration, progressive supranuclear palsy, and Pick's disease.
Collapse
Affiliation(s)
- Heiko Kroth
- AC Immune SA, EPFL Innovation Park, Building B, 1015 Lausanne, Switzerland
| | - Felix Oden
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| | - Jerome Molette
- AC Immune SA, EPFL Innovation Park, Building B, 1015 Lausanne, Switzerland
| | | | | | - Andre Mueller
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| | - Mathias Berndt
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| | | | | | | | | | - David Hickman
- AC Immune SA, EPFL Innovation Park, Building B, 1015 Lausanne, Switzerland
| | - Andrea Pfeifer
- AC Immune SA, EPFL Innovation Park, Building B, 1015 Lausanne, Switzerland
| | - Ludger Dinkelborg
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| | - Andrew Stephens
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany
| |
Collapse
|
10
|
Campese N, Palermo G, Del Gamba C, Beatino MF, Galgani A, Belli E, Del Prete E, Della Vecchia A, Vergallo A, Siciliano G, Ceravolo R, Hampel H, Baldacci F. Progress regarding the context-of-use of tau as biomarker of Alzheimer's disease and other neurodegenerative diseases. Expert Rev Proteomics 2021; 18:27-48. [PMID: 33545008 DOI: 10.1080/14789450.2021.1886929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Tau protein misfolding and accumulation in toxic species is a critical pathophysiological process of Alzheimer's disease (AD) and other neurodegenerative disorders (NDDs). Tau biomarkers, namely cerebrospinal fluid (CSF) total-tau (t-tau), 181-phosphorylated tau (p-tau), and tau-PET tracers, have been recently embedded in the diagnostic criteria for AD. Nevertheless, the role of tau as a diagnostic and prognostic biomarker for other NDDs remains controversial.Areas covered: We performed a systematical PubMed-based review of the most recent advances in tau-related biomarkers for NDDs. We focused on papers published from 2015 to 2020 assessing the diagnostic or prognostic value of each biomarker.Expert opinion: The assessment of tau biomarkers in alternative easily accessible matrices, through the development of ultrasensitive techniques, represents the most significant perspective for AD-biomarker research. In NDDs, novel tau isoforms (e.g. p-tau217) or proteolytic fragments (e.g. N-terminal fragments) may represent candidate diagnostic and prognostic biomarkers and may help monitoring disease progression. Protein misfolding amplification assays, allowing the identification of different tau strains (e.g. 3 R- vs. 4 R-tau) in CSF, may constitute a breakthrough for the in vivo stratification of NDDs. Tau-PET may help tracking the spatial-temporal evolution of tau pathophysiology in AD but its application outside the AD-spectrum deserves further studies.
Collapse
Affiliation(s)
- Nicole Campese
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giovanni Palermo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Claudia Del Gamba
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Alessandro Galgani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elisabetta Belli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Eleonora Del Prete
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Andrea Vergallo
- GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Harald Hampel
- GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| |
Collapse
|
11
|
Tamil Selvan S, Ravichandar R, Kanta Ghosh K, Mohan A, Mahalakshmi P, Gulyás B, Padmanabhan P. Coordination chemistry of ligands: Insights into the design of amyloid beta/tau-PET imaging probes and nanoparticles-based therapies for Alzheimer’s disease. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2020.213659] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
12
|
Clinical Evaluation of 18F-PI-2620 as a Potent PET Radiotracer Imaging Tau Protein in Alzheimer Disease and Other Neurodegenerative Diseases Compared With 18F-THK-5351. Clin Nucl Med 2020; 45:841-847. [PMID: 32910050 DOI: 10.1097/rlu.0000000000003261] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE PET is a useful tool for detecting the presence and extent of brain tau accumulation. However, most first-generation tau PET tracers are limited for high off-target binding and detection of tau in non-Alzheimer disease (AD). This study evaluated potential clinical applications of F-PI-2620 as a novel PET tracer with a high binding affinity for tau deposition in AD and non-AD tauopathies. METHODS Twenty-six participants diagnosed with either mild cognitive impairment, probable AD, frontotemporal dementia, or parkinsonism, as well as healthy controls underwent a 60- to 90-minute brain PET scan after 7 mci (259 MBq) injection of F-PI-2620. Some participants had previous PET scans using F-THK-5351 or F-FP-CIT for dopamine transporter imaging. RESULTS All participants showed no increase in off-target binding in basal ganglia on F-PI-2620 PET images, as noted for first-generation tau tracers. Aβ+ mild cognitive impairment or AD patients showed diverse cortical F-PI-2620 uptake in frontotemporoparietal cortex that correlated with Mini-Mental Status Examination (ρ = -0.692, P = 0.013). Aβ+ Parkinson disease with dementia and (Aβ unknown) primary progressive aphasia patients also showed increased F-PI-2620 uptakes in the frontotemporoparietal cortex. Patients with parkinsonism showed increased uptakes in the pallidum compared with Aβ- healthy controls (left: 1.41 ± 0.14 vs 1.04 ± 0.13, P = 0.014; right: 1.18 ± 0.16 vs 0.95 ± 0.07, P = 0.014). CONCLUSIONS F-PI-2620 PET might be a sensitive tool to detect cortical tau deposits in patients with Aβ+ AD and Aβ+ non-AD tauopathies. Furthermore, this study showed that "off-target" binding in the basal ganglia does not affect F-PI-2620.
Collapse
|
13
|
Tau PTM Profiles Identify Patient Heterogeneity and Stages of Alzheimer's Disease. Cell 2020; 183:1699-1713.e13. [PMID: 33188775 DOI: 10.1016/j.cell.2020.10.029] [Citation(s) in RCA: 434] [Impact Index Per Article: 86.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 08/27/2020] [Accepted: 10/16/2020] [Indexed: 01/06/2023]
Abstract
To elucidate the role of Tau isoforms and post-translational modification (PTM) stoichiometry in Alzheimer's disease (AD), we generated a high-resolution quantitative proteomics map of 95 PTMs on multiple isoforms of Tau isolated from postmortem human tissue from 49 AD and 42 control subjects. Although Tau PTM maps reveal heterogeneity across subjects, a subset of PTMs display high occupancy and frequency for AD, suggesting importance in disease. Unsupervised analyses indicate that PTMs occur in an ordered manner, leading to Tau aggregation. The processive addition and minimal set of PTMs associated with seeding activity was further defined by analysis of size-fractionated Tau. To summarize, features in the Tau protein critical for disease intervention at different stages of disease are identified, including enrichment of 0N and 4R isoforms, underrepresentation of the C terminus, an increase in negative charge in the proline-rich region (PRR), and a decrease in positive charge in the microtubule binding domain (MBD).
Collapse
|
14
|
Gabellieri E, Capotosti F, Molette J, Sreenivasachary N, Mueller A, Berndt M, Schieferstein H, Juergens T, Varisco Y, Oden F, Schmitt-Willich H, Hickman D, Dinkelborg L, Stephens A, Pfeifer A, Kroth H. Discovery of 2-(4-(2-fluoroethoxy)piperidin-1-yl)-9-methyl-9H-pyrrolo[2,3-b:4,5-c']dipyridine ([18F]PI-2014) as PET tracer for the detection of pathological aggregated tau in Alzheimer's disease and other tauopathies. Eur J Med Chem 2020; 204:112615. [PMID: 32771872 DOI: 10.1016/j.ejmech.2020.112615] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/04/2020] [Accepted: 06/21/2020] [Indexed: 12/29/2022]
Abstract
The compound screening was initiated with a direct staining assay to identify compounds binding to Tau aggregates and not Abeta plaques using human brain sections derived from late stage Alzheimer's disease donors. The binding of Tau aggregate selective compounds was then quantitatively assessed with human brain derived paired helical filaments utilizing the label-free Back Scattering Interferometry assay. In vivo biodistribution experiments of selected fluorine-18 labeled compounds were performed in mice to assess brain uptake, brain washout, and defluorination. Compound 11 emerged as the most promising candidate, displaying high in vitro binding affinity and selectivity to neurofibrillary tangles. Fluorine-18 labeled compound 11 showed high brain uptake and rapid washout from the mouse brain with no observed bone uptake. Furthermore, compound 11 was able to detect Tau aggregates in tauopathy brain sections from corticobasal degeneration, progressive supranuclear palsy, and Pick's disease donors. Thus, 2-(4-(2-fluoroethoxy)piperidin-1-yl)-9-methyl-9H-pyrrolo[2,3-b:4,5-c']dipyridine (PI-2014, compound 11) was selected for characterization in a first-in-human study.
Collapse
Affiliation(s)
| | | | - Jerome Molette
- AC Immune SA, EPFL Innovation Park, Building B, 1015, Lausanne, Switzerland
| | | | - Andre Mueller
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353, Berlin, Germany
| | - Mathias Berndt
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353, Berlin, Germany
| | - Hanno Schieferstein
- Formerly Piramal Imaging GmbH, Tegeler Strasse 6-7, 13353, Berlin, Germany; Merck KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
| | - Tanja Juergens
- AC Immune SA, EPFL Innovation Park, Building B, 1015, Lausanne, Switzerland
| | - Yvan Varisco
- AC Immune SA, EPFL Innovation Park, Building B, 1015, Lausanne, Switzerland
| | - Felix Oden
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353, Berlin, Germany
| | | | - David Hickman
- AC Immune SA, EPFL Innovation Park, Building B, 1015, Lausanne, Switzerland
| | - Ludger Dinkelborg
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353, Berlin, Germany
| | - Andrew Stephens
- Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353, Berlin, Germany
| | - Andrea Pfeifer
- AC Immune SA, EPFL Innovation Park, Building B, 1015, Lausanne, Switzerland
| | - Heiko Kroth
- AC Immune SA, EPFL Innovation Park, Building B, 1015, Lausanne, Switzerland.
| |
Collapse
|
15
|
Characterization of MK6240, a tau PET tracer, in autopsy brain tissue from Alzheimer's disease cases. Eur J Nucl Med Mol Imaging 2020; 48:1093-1102. [PMID: 32970217 PMCID: PMC8041708 DOI: 10.1007/s00259-020-05035-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 07/31/2020] [Indexed: 12/21/2022]
Abstract
Purpose MK6240 is a second-generation tau PET tracer designed to detect the neurofibrillary tangles in the brains of patients with Alzheimer’s disease (AD). The aim of the study was to characterize 3H-MK6240 in AD and control brain tissue and to compare its binding properties with those of first-generation tau PET tracers. Methods Saturation binding assays with 3H-MK6240 were carried out in the temporal and parietal cortices of AD brains to determine the maximum number of binding sites (Bmax) and the dissociation constants (Kd) at these sites. Competitive binding assays were carried out between 3H-MK6240 and unlabelled MK6240, AV-1451 (aka T807, flortaucipir) and THK5117, and between 3H-THK5351 and unlabelled MK6240. Regional binding studies with 3H-MK6240 were carried out in homogenates from six AD and seven control brains and, using autoradiography, on large frozen sections from two AD brains and one control brain. Results The saturation binding assays gave Bmax and Kd values of 59.2 fmol/mg and 0.32 nM in the temporal cortex and 154.7 fmol/mg and 0.15 nM in the parietal cortex. The competitive binding assays revealed two binding sites with affinities in the picomolar and nanomolar range shared by 3H-MK6240 and all the tested unlabelled compounds. There were no binding sites in common between 3H-THK5351 and unlabelled MK6240. Regional binding of 3H-MK6240 was significantly higher in AD brain tissue than in controls. Binding in brain tissue from AD patients with early-onset AD was significantly higher than in brain tissue from patients with late-onset AD. Binding of 3H-MK6240 was not observed in off-target regions. Autoradiography showed high regional cortical binding in the two AD brains and very low binding in the control brain. Conclusions 3H-MK6240 has a high binding affinity for tau deposits in AD brain tissue but also has different binding characteristics from those of the first-generation tau tracers. This confirms the complexity of tau tracer binding on tau deposits with different binding affinities for different binding sites. Electronic supplementary material The online version of this article (10.1007/s00259-020-05035-y) contains supplementary material, which is available to authorized users.
Collapse
|
16
|
Kramer V, Brooks AF, Haeger A, Kuljis RO, Rafique W, Koeppe RA, Raffel DM, Frey KA, Amaral H, Scott PJH, Riss PJ. Evaluation of [ 18F]- N-Methyl lansoprazole as a Tau PET Imaging Agent in First-in-Human Studies. ACS Chem Neurosci 2020; 11:427-435. [PMID: 31898886 DOI: 10.1021/acschemneuro.9b00639] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Development of positron emission tomography (PET) imaging agents capable of quantifying tau aggregates in neurodegenerative disorders such as Alzheimer's disease (AD) is of enormous importance in the field of dementia research. The aim of the present study was to conduct first-in-man imaging studies with the potential novel tau imaging agent [18F]N-methyl lansoprazole ([18F]NML). Herein we report validation of the synthesis of [18F]NML for clinical use by labeling the trifluoromethyl group via radiofluorination of the corresponding gem-difluoro enol ether precursor. This is the first use of this method for clinical production of PET radiotracers and confirmed that it can be readily implemented at multiple production facilities to provide [18F]NML in good noncorrected radiochemical yield (3.4 ± 1.5 GBq, 4.6% ± 2.6%) and molar activity (120.1 ± 186.3 GBq/μmol), excellent radiochemical purity (>97%), and suitable for human use (n = 15). With [18F]NML in hand, we conducted rodent biodistribution, estimates of human dosimetry, and preliminary evaluation of [18F]NML in human subjects at two imaging sites. Healthy controls (n = 4) and mildly cognitively impaired (MCI) AD patients (n = 6) received [18F]NML (tau), [18F]AV1451 (tau), and [18F]florbetaben or [18F]florbetapir (amyloid) PET scans. A single progressive supranuclear palsy (PSP) patient also received [18F]NML and [18F]AV1451 PET scans. [18F]NML showed good brain uptake, reasonable pharmacokinetics, and appropriate imaging characteristics in healthy controls. The mean ± SD of the administered mass of [18F/19F]NML was 2.01 ± 2.17 μg (range, 0.16-8.27 μg) and the mean administered activity was 350 ± 62 MBq (range, 199-403 MBq). There were no adverse or clinically detectable pharmacologic effects in any of the 11 subjects, and no significant changes in vital signs were observed. However, despite high affinity for tau in vitro, brain retention in MCI/AD and PSP patients was low, and there was no evidence of specific signals in vivo that corresponded to tau. Although it is still unclear why clinical translation of the radiotracer was unsuccessful, we nevertheless conclude that further development of [18F]NML as a tau PET imaging agent is not warranted at this time.
Collapse
Affiliation(s)
- Vasko Kramer
- Center for Nuclear Medicine & PET/CT Positronmed, Providencia, 7501068 Santiago, Chile
- Positronpharma SA, Providencia, 7500921 Santiago Chile
| | - Allen F. Brooks
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Arlette Haeger
- Center for Nuclear Medicine & PET/CT Positronmed, Providencia, 7501068 Santiago, Chile
| | - Rodrigo O. Kuljis
- Center for Nuclear Medicine & PET/CT Positronmed, Providencia, 7501068 Santiago, Chile
| | - Waqas Rafique
- realomics SRI, Kjemisk Institutt, Universitetet i Oslo, Sem Sælands vei 26, Kjemibygningen, 0371 Oslo, Norway
| | - Robert A. Koeppe
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - David M. Raffel
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Kirk A. Frey
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Horacio Amaral
- Center for Nuclear Medicine & PET/CT Positronmed, Providencia, 7501068 Santiago, Chile
- Positronpharma SA, Providencia, 7500921 Santiago Chile
| | - Peter J. H. Scott
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Patrick J. Riss
- realomics SRI, Kjemisk Institutt, Universitetet i Oslo, Sem Sælands vei 26, Kjemibygningen, 0371 Oslo, Norway
- Klinik for Kirurgi og Nevrofag, Oslo Universitets Sykehus HF−Rikshospitalet, Postboks
4950 Nydalen, 0424 Oslo, Norway
- Norsk Medisinsk Syklotronsenter AS, Gaustad, Postboks
4950 Nydalen, 0424 Oslo, Norway
| |
Collapse
|
17
|
Beaurain M, Salabert AS, Ribeiro MJ, Arlicot N, Damier P, Le Jeune F, Demonet JF, Payoux P. Innovative Molecular Imaging for Clinical Research, Therapeutic Stratification, and Nosography in Neuroscience. Front Med (Lausanne) 2019; 6:268. [PMID: 31828073 PMCID: PMC6890558 DOI: 10.3389/fmed.2019.00268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 11/01/2019] [Indexed: 01/06/2023] Open
Abstract
Over the past few decades, several radiotracers have been developed for neuroimaging applications, especially in PET. Because of their low steric hindrance, PET radionuclides can be used to label molecules that are small enough to cross the blood brain barrier, without modifying their biological properties. As the use of 11C is limited by its short physical half-life (20 min), there has been an increasing focus on developing tracers labeled with 18F for clinical use. The first such tracers allowed cerebral blood flow and glucose metabolism to be measured, and the development of molecular imaging has since enabled to focus more closely on specific targets such as receptors, neurotransmitter transporters, and other proteins. Hence, PET and SPECT biomarkers have become indispensable for innovative clinical research. Currently, the treatment options for a number of pathologies, notably neurodegenerative diseases, remain only supportive and symptomatic. Treatments that slow down or reverse disease progression are therefore the subject of numerous studies, in which molecular imaging is proving to be a powerful tool. PET and SPECT biomarkers already make it possible to diagnose several neurological diseases in vivo and at preclinical stages, yielding topographic, and quantitative data about the target. As a result, they can be used for assessing patients' eligibility for new treatments, or for treatment follow-up. The aim of the present review was to map major innovative radiotracers used in neuroscience, and explain their contribution to clinical research. We categorized them according to their target: dopaminergic, cholinergic or serotoninergic systems, β-amyloid plaques, tau protein, neuroinflammation, glutamate or GABA receptors, or α-synuclein. Most neurological disorders, and indeed mental disorders, involve the dysfunction of one or more of these targets. Combinations of molecular imaging biomarkers can afford us a better understanding of the mechanisms underlying disease development over time, and contribute to early detection/screening, diagnosis, therapy delivery/monitoring, and treatment follow-up in both research and clinical settings.
Collapse
Affiliation(s)
- Marie Beaurain
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| | - Anne-Sophie Salabert
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| | - Maria Joao Ribeiro
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Inserm CIC 1415, University Hospital, Tours, France.,CHRU Tours, Tours, France
| | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Inserm CIC 1415, University Hospital, Tours, France.,CHRU Tours, Tours, France
| | - Philippe Damier
- Inserm U913, Neurology Department, University Hospital, Nantes, France
| | | | - Jean-François Demonet
- Leenards Memory Centre, Department of Clinical Neuroscience, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Pierre Payoux
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| |
Collapse
|
18
|
Rankovic M, Zweckstetter M. Upregulated levels and pathological aggregation of abnormally phosphorylated Tau-protein in children with neurodevelopmental disorders. Neurosci Biobehav Rev 2019; 98:1-9. [DOI: 10.1016/j.neubiorev.2018.12.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/06/2018] [Accepted: 12/10/2018] [Indexed: 02/06/2023]
|
19
|
Katako A, Shelton P, Goertzen AL, Levin D, Bybel B, Aljuaid M, Yoon HJ, Kang DY, Kim SM, Lee CS, Ko JH. Machine learning identified an Alzheimer's disease-related FDG-PET pattern which is also expressed in Lewy body dementia and Parkinson's disease dementia. Sci Rep 2018; 8:13236. [PMID: 30185806 PMCID: PMC6125295 DOI: 10.1038/s41598-018-31653-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 08/23/2018] [Indexed: 01/17/2023] Open
Abstract
Utilizing the publicly available neuroimaging database enabled by Alzheimer's disease Neuroimaging Initiative (ADNI; http://adni.loni.usc.edu/ ), we have compared the performance of automated classification algorithms that differentiate AD vs. normal subjects using Positron Emission Tomography (PET) with fluorodeoxyglucose (FDG). General linear model, scaled subprofile modeling and support vector machines were examined. Among the tested classification methods, support vector machine with Iterative Single Data Algorithm produced the best performance, i.e., sensitivity (0.84) × specificity (0.95), by 10-fold cross-validation. We have applied the same classification algorithm to four different datasets from ADNI, Health Science Centre (Winnipeg, Canada), Dong-A University Hospital (Busan, S. Korea) and Asan Medical Centre (Seoul, S. Korea). Our data analyses confirmed that the support vector machine with Iterative Single Data Algorithm showed the best performance in prediction of future development of AD from the prodromal stage (mild cognitive impairment), and that it was also sensitive to other types of dementia such as Parkinson's Disease Dementia and Dementia with Lewy Bodies, and that perfusion imaging using single photon emission computed tomography may achieve a similar accuracy to that of FDG-PET.
Collapse
Affiliation(s)
- Audrey Katako
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Manitoba, Canada
| | - Paul Shelton
- Section of Neurology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrew L Goertzen
- Section of Nuclear Medicine, Department of Radiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Daniel Levin
- Section of Nuclear Medicine, Department of Radiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Bohdan Bybel
- Section of Nuclear Medicine, Department of Radiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Maram Aljuaid
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Manitoba, Canada
| | - Hyun Jin Yoon
- Department of Nuclear Medicine, College of Medicine, Dong-A University, Busan, South Korea
| | - Do Young Kang
- Department of Nuclear Medicine, College of Medicine, Dong-A University, Busan, South Korea
| | - Seok Min Kim
- Institute of Parkinson's Clinical Research, Ulsan University College of Medicine, Seoul, South Korea
| | - Chong Sik Lee
- Institute of Parkinson's Clinical Research, Ulsan University College of Medicine, Seoul, South Korea.,Department of Neurology, Asan Medical Center, Seoul, South Korea
| | - Ji Hyun Ko
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. .,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Manitoba, Canada.
| |
Collapse
|
20
|
A Closer Look into the Role of Protein Tau in the Identification of Promising Therapeutic Targets for Alzheimer's Disease. Brain Sci 2018; 8:brainsci8090162. [PMID: 30149687 PMCID: PMC6162660 DOI: 10.3390/brainsci8090162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 08/24/2018] [Accepted: 08/24/2018] [Indexed: 01/09/2023] Open
Abstract
One of the most commonly known chronic neurodegenerative disorders, Alzheimer's disease (AD), manifests the common type of dementia in 60⁻80% of cases. From a clinical standpoint, a patent cognitive decline and a severe change in personality, as caused by a loss of neurons, is usually evident in AD with about 50 million people affected in 2016. The disease progression in patients is distinguished by a gradual plummet in cognitive functions, eliciting symptoms such as memory loss, and eventually requiring full-time medical care. From a histopathological standpoint, the defining characteristics are intracellular aggregations of hyper-phosphorylated tau protein, known as neurofibrillary tangles (NFT), and depositions of amyloid β-peptides (Aβ) in the brain. The abnormal phosphorylation of tau protein is attributed to a wide gamut of neurological disorders known as tauopathies. In addition to the hyperphosphorylated tau lesions, neuroinflammatory processes could occur in a sustained manner through astro-glial activation, resulting in the disease progression. Recent findings have suggested a strong interplay between the mechanism of Tau phosphorylation, disruption of microtubules, and synaptic loss and pathology of AD. The mechanisms underlying these interactions along with their respective consequences in Tau pathology are still ill-defined. Thus, in this review: (1) we highlight the interplays existing between Tau pathology and AD; and (2) take a closer look into its role while identifying some promising therapeutic advances including state of the art imaging techniques.
Collapse
|
21
|
Hampel H, Toschi N, Babiloni C, Baldacci F, Black KL, Bokde AL, Bun RS, Cacciola F, Cavedo E, Chiesa PA, Colliot O, Coman CM, Dubois B, Duggento A, Durrleman S, Ferretti MT, George N, Genthon R, Habert MO, Herholz K, Koronyo Y, Koronyo-Hamaoui M, Lamari F, Langevin T, Lehéricy S, Lorenceau J, Neri C, Nisticò R, Nyasse-Messene F, Ritchie C, Rossi S, Santarnecchi E, Sporns O, Verdooner SR, Vergallo A, Villain N, Younesi E, Garaci F, Lista S. Revolution of Alzheimer Precision Neurology. Passageway of Systems Biology and Neurophysiology. J Alzheimers Dis 2018; 64:S47-S105. [PMID: 29562524 PMCID: PMC6008221 DOI: 10.3233/jad-179932] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The Precision Neurology development process implements systems theory with system biology and neurophysiology in a parallel, bidirectional research path: a combined hypothesis-driven investigation of systems dysfunction within distinct molecular, cellular, and large-scale neural network systems in both animal models as well as through tests for the usefulness of these candidate dynamic systems biomarkers in different diseases and subgroups at different stages of pathophysiological progression. This translational research path is paralleled by an "omics"-based, hypothesis-free, exploratory research pathway, which will collect multimodal data from progressing asymptomatic, preclinical, and clinical neurodegenerative disease (ND) populations, within the wide continuous biological and clinical spectrum of ND, applying high-throughput and high-content technologies combined with powerful computational and statistical modeling tools, aimed at identifying novel dysfunctional systems and predictive marker signatures associated with ND. The goals are to identify common biological denominators or differentiating classifiers across the continuum of ND during detectable stages of pathophysiological progression, characterize systems-based intermediate endophenotypes, validate multi-modal novel diagnostic systems biomarkers, and advance clinical intervention trial designs by utilizing systems-based intermediate endophenotypes and candidate surrogate markers. Achieving these goals is key to the ultimate development of early and effective individualized treatment of ND, such as Alzheimer's disease. The Alzheimer Precision Medicine Initiative (APMI) and cohort program (APMI-CP), as well as the Paris based core of the Sorbonne University Clinical Research Group "Alzheimer Precision Medicine" (GRC-APM) were recently launched to facilitate the passageway from conventional clinical diagnostic and drug development toward breakthrough innovation based on the investigation of the comprehensive biological nature of aging individuals. The APMI movement is gaining momentum to systematically apply both systems neurophysiology and systems biology in exploratory translational neuroscience research on ND.
Collapse
Affiliation(s)
- Harald Hampel
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
- Department of Radiology, “Athinoula A. Martinos” Center for Biomedical Imaging, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Claudio Babiloni
- Department of Physiology and Pharmacology “Vittorio Erspamer”, University of Rome “La Sapienza”, Rome, Italy
- Institute for Research and Medical Care, IRCCS “San Raffaele Pisana”, Rome, Italy
| | - Filippo Baldacci
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Keith L. Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Arun L.W. Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience (TCIN), Trinity College Dublin, Dublin, Ireland
| | - René S. Bun
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Francesco Cacciola
- Unit of Neurosurgery, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Enrica Cavedo
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
- IRCCS “San Giovanni di Dio-Fatebenefratelli”, Brescia, Italy
| | - Patrizia A. Chiesa
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Olivier Colliot
- Inserm, U1127, Paris, France; CNRS, UMR 7225 ICM, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Paris, France; Institut du Cerveau et de la Moelle Épinière (ICM) Paris, France; Inria, Aramis project-team, Centre de Recherche de Paris, France; Department of Neuroradiology, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France; Department of Neurology, AP-HP, Hôpital de la Pitié-Salpêtrière, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Paris, France
| | - Cristina-Maria Coman
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Bruno Dubois
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
| | - Andrea Duggento
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Stanley Durrleman
- Inserm, U1127, Paris, France; CNRS, UMR 7225 ICM, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Paris, France; Institut du Cerveau et de la Moelle Épinière (ICM) Paris, France; Inria, Aramis project-team, Centre de Recherche de Paris, France
| | - Maria-Teresa Ferretti
- IREM, Institute for Regenerative Medicine, University of Zurich, Zürich, Switzerland
- ZNZ Neuroscience Center Zurich, Zürich, Switzerland
| | - Nathalie George
- Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Inserm U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle Épinière, ICM, Ecole Normale Supérieure, ENS, Centre MEG-EEG, F-75013, Paris, France
| | - Remy Genthon
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
| | - Marie-Odile Habert
- Département de Médecine Nucléaire, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
- Laboratoire d’Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, Inserm U 1146, CNRS UMR 7371, Paris, France
| | - Karl Herholz
- Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Foudil Lamari
- AP-HP, UF Biochimie des Maladies Neuro-métaboliques, Service de Biochimie Métabolique, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | | | - Stéphane Lehéricy
- Centre de NeuroImagerie de Recherche - CENIR, Institut du Cerveau et de la Moelle Épinière - ICM, F-75013, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Inserm U 1127, CNRS UMR 7225, ICM, F-75013, Paris, France
| | - Jean Lorenceau
- Institut de la Vision, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR_S968, CNRS UMR7210, Paris, France
| | - Christian Neri
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, CNRS UMR 8256, Institut de Biologie Paris-Seine (IBPS), Place Jussieu, F-75005, Paris, France
| | - Robert Nisticò
- Department of Biology, University of Rome “Tor Vergata” & Pharmacology of Synaptic Disease Lab, European Brain Research Institute (E.B.R.I.), Rome, Italy
| | - Francis Nyasse-Messene
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
| | - Craig Ritchie
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Simone Rossi
- Department of Medicine, Surgery and Neurosciences, Unit of Neurology and Clinical Neurophysiology, Brain Investigation & Neuromodulation Lab. (Si-BIN Lab.), University of Siena, Siena, Italy
- Department of Medicine, Surgery and Neurosciences, Section of Human Physiology University of Siena, Siena, Italy
| | - Emiliano Santarnecchi
- Department of Medicine, Surgery and Neurosciences, Unit of Neurology and Clinical Neurophysiology, Brain Investigation & Neuromodulation Lab. (Si-BIN Lab.), University of Siena, Siena, Italy
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Olaf Sporns
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
- IU Network Science Institute, Indiana University, Bloomington, IN, USA
| | | | - Andrea Vergallo
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Nicolas Villain
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | | | - Francesco Garaci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
- Casa di Cura “San Raffaele Cassino”, Cassino, Italy
| | - Simone Lista
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| |
Collapse
|
22
|
Castrillo JI, Lista S, Hampel H, Ritchie CW. Systems Biology Methods for Alzheimer’s Disease Research Toward Molecular Signatures, Subtypes, and Stages and Precision Medicine: Application in Cohort Studies and Trials. Methods Mol Biol 2018; 1750:31-66. [PMID: 29512064 DOI: 10.1007/978-1-4939-7704-8_3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Juan I Castrillo
- Genetadi Biotech S.L. Parque Tecnológico de Bizkaia, Derio, Bizkaia, Spain.
| | - Simone Lista
- AXA Research Fund & UPMC Chair, F-75013, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l'hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l'hôpital, F-75013, Paris, France
| | - Harald Hampel
- AXA Research Fund & UPMC Chair, F-75013, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l'hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l'hôpital, F-75013, Paris, France
| | - Craig W Ritchie
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
23
|
Abstract
Although dementia has been described in ancient texts over many centuries (e.g., "Be kind to your father, even if his mind fail him." - Old Testament: Sirach 3:12), our knowledge of its underlying causes is little more than a century old. Alzheimer published his now famous case study only 110 years ago, and our modern understanding of the disease that bears his name, and its neuropsychological consequences, really only began to accelerate in the 1980s. Since then we have witnessed an explosion of basic and translational research into the causes, characterizations, and possible treatments for Alzheimer's disease (AD) and other dementias. We review this lineage of work beginning with Alzheimer's own writings and drawings, then jump to the modern era beginning in the 1970s and early 1980s and provide a sampling of neuropsychological and other contextual work from each ensuing decade. During the 1980s our field began its foundational studies of profiling the neuropsychological deficits associated with AD and its differentiation from other dementias (e.g., cortical vs. subcortical dementias). The 1990s continued these efforts and began to identify the specific cognitive mechanisms affected by various neuropathologic substrates. The 2000s ushered in a focus on the study of prodromal stages of neurodegenerative disease before the full-blown dementia syndrome (i.e., mild cognitive impairment). The current decade has seen the rise of imaging and other biomarkers to characterize preclinical disease before the development of significant cognitive decline. Finally, we suggest future directions and predictions for dementia-related research and potential therapeutic interventions. (JINS, 2017, 23, 818-831).
Collapse
Affiliation(s)
- Mark W. Bondi
- Department of Psychiatry, University of California San Diego, School of Medicine, La Jolla, California
- Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Emily C. Edmonds
- Department of Psychiatry, University of California San Diego, School of Medicine, La Jolla, California
- Veterans Affairs San Diego Healthcare System, San Diego, California
| | - David P. Salmon
- Department of Neurosciences, University of California San Diego, School of Medicine, La Jolla, California
| |
Collapse
|
24
|
Husain M. Alzheimer's disease: time to focus on the brain, not just molecules. Brain 2017; 140:251-253. [DOI: 10.1093/brain/aww353] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 12/20/2016] [Indexed: 01/01/2023] Open
|
25
|
Shefner JM, Sabbagh MN. An Appraisal of Novel Biomarkers for Evaluating and Monitoring Neurologic Diseases: Editorial Introduction. Neurotherapeutics 2017; 14:1-3. [PMID: 27933486 PMCID: PMC5233637 DOI: 10.1007/s13311-016-0502-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Jeremy M. Shefner
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ USA
| | - Marwan N. Sabbagh
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ USA
| |
Collapse
|