1
|
El Abiad E, Al-Kuwari A, Al-Aani U, Al Jaidah Y, Chaari A. Navigating the Alzheimer's Biomarker Landscape: A Comprehensive Analysis of Fluid-Based Diagnostics. Cells 2024; 13:1901. [PMID: 39594648 PMCID: PMC11593284 DOI: 10.3390/cells13221901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) affects a significant portion of the aging population, presenting a serious challenge due to the limited availability of effective therapies during its progression. The disease advances rapidly, underscoring the need for early diagnosis and the application of preventative measures. Current diagnostic methods for AD are often expensive and invasive, restricting access for the general public. One potential solution is the use of biomarkers, which can facilitate early detection and treatment through objective, non-invasive, and cost-effective evaluations of AD. This review critically investigates the function and role of biofluid biomarkers in detecting AD, with a specific focus on cerebrospinal fluid (CSF), blood-based, and saliva biomarkers. RESULTS CSF biomarkers have demonstrated potential for accurate diagnosis and valuable prognostic insights, while blood biomarkers offer a minimally invasive and cost-effective approach for diagnosing cognitive issues. However, while current biomarkers for AD show significant potential, none have yet achieved the precision needed to replace expensive PET scans and CSF assays. The lack of a single accurate biomarker underscores the need for further research to identify novel or combined biomarkers to enhance the clinical efficacy of existing diagnostic tests. In this context, artificial intelligence (AI) and deep-learning (DL) tools present promising avenues for improving biomarker analysis and interpretation, enabling more precise and timely diagnoses. CONCLUSIONS Further research is essential to confirm the utility of all AD biomarkers in clinical settings. Combining biomarker data with AI tools offers a promising path toward revolutionizing the personalized characterization and early diagnosis of AD symptoms.
Collapse
Affiliation(s)
| | | | | | | | - Ali Chaari
- Weill Cornell Medicine–Qatar, Qatar Foundation, Education City, Doha P.O. Box 24144, Qatar; (E.E.A.); (A.A.-K.); (U.A.-A.); (Y.A.J.)
| |
Collapse
|
2
|
Kale MB, Bhondge HM, Wankhede NL, Shende PV, Thanekaer RP, Aglawe MM, Rahangdale SR, Taksande BG, Pandit SB, Upaganlawar AB, Umekar MJ, Kopalli SR, Koppula S. Navigating the intersection: Diabetes and Alzheimer's intertwined relationship. Ageing Res Rev 2024; 100:102415. [PMID: 39002642 DOI: 10.1016/j.arr.2024.102415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/06/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Alzheimer's disease (AD) and Diabetes mellitus (DM) exhibit comparable pathophysiological pathways. Genetic abnormalities in APP, PS-1, and PS-2 are linked to AD, with diagnostic aid from CSF and blood biomarkers. Insulin dysfunction, termed "type 3 diabetes mellitus" in AD, involves altered insulin signalling and neuronal shrinkage. Insulin influences beta-amyloid metabolism, exacerbating neurotoxicity in AD and amyloid production in DM. Both disorders display impaired glucose transporter expression, hastening cognitive decline. Mitochondrial dysfunction and Toll-like receptor 4-mediated inflammation worsen neurodegeneration in both diseases. ApoE4 raises disease risk, especially when coupled with dyslipidemia common in DM. Targeting shared pathways like insulin-degrading enzyme activation and HSP60 holds promise for therapeutic intervention. Recognizing these interconnected mechanisms underscores the imperative for developing tailored treatments addressing the overlapping pathophysiology of AD and DM, offering potential avenues for more effective management of both conditions.
Collapse
Affiliation(s)
- Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | | | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Rushikesh P Thanekaer
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Manish M Aglawe
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sandip R Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sunil B Pandit
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
3
|
Kim AY, Al Jerdi S, MacDonald R, Triggle CR. Alzheimer's disease and its treatment-yesterday, today, and tomorrow. Front Pharmacol 2024; 15:1399121. [PMID: 38868666 PMCID: PMC11167451 DOI: 10.3389/fphar.2024.1399121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/25/2024] [Indexed: 06/14/2024] Open
Abstract
Alois Alzheimer described the first patient with Alzheimer's disease (AD) in 1907 and today AD is the most frequently diagnosed of dementias. AD is a multi-factorial neurodegenerative disorder with familial, life style and comorbidity influences impacting a global population of more than 47 million with a projected escalation by 2050 to exceed 130 million. In the USA the AD demographic encompasses approximately six million individuals, expected to increase to surpass 13 million by 2050, and the antecedent phase of AD, recognized as mild cognitive impairment (MCI), involves nearly 12 million individuals. The economic outlay for the management of AD and AD-related cognitive decline is estimated at approximately 355 billion USD. In addition, the intensifying prevalence of AD cases in countries with modest to intermediate income countries further enhances the urgency for more therapeutically and cost-effective treatments and for improving the quality of life for patients and their families. This narrative review evaluates the pathophysiological basis of AD with an initial focus on the therapeutic efficacy and limitations of the existing drugs that provide symptomatic relief: acetylcholinesterase inhibitors (AChEI) donepezil, galantamine, rivastigmine, and the N-methyl-D-aspartate receptor (NMDA) receptor allosteric modulator, memantine. The hypothesis that amyloid-β (Aβ) and tau are appropriate targets for drugs and have the potential to halt the progress of AD is critically analyzed with a particular focus on clinical trial data with anti-Aβ monoclonal antibodies (MABs), namely, aducanumab, lecanemab and donanemab. This review challenges the dogma that targeting Aβ will benefit the majority of subjects with AD that the anti-Aβ MABs are unlikely to be the "magic bullet". A comparison of the benefits and disadvantages of the different classes of drugs forms the basis for determining new directions for research and alternative drug targets that are undergoing pre-clinical and clinical assessments. In addition, we discuss and stress the importance of the treatment of the co-morbidities, including hypertension, diabetes, obesity and depression that are known to increase the risk of developing AD.
Collapse
Affiliation(s)
- A. Y. Kim
- Medical Education, Weill Cornell Medicine—Qatar, Doha, Qatar
| | | | - R. MacDonald
- Health Sciences Library, Weill Cornell Medicine—Qatar, Doha, Qatar
| | - C. R. Triggle
- Department of Pharmacology and Medical Education, Weill Cornell Medicine—Qatar, Doha, Qatar
| |
Collapse
|
4
|
Kavoosi S, Shahraki A, Sheervalilou R. Identification of microRNA-mRNA Regulatory Networks with Therapeutic Values in Alzheimer's Disease by Bioinformatics Analysis. J Alzheimers Dis 2024; 98:671-689. [PMID: 38427479 DOI: 10.3233/jad-230966] [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] [Indexed: 03/03/2024]
Abstract
Background Alzheimer's disease (AD) is the most prevalent neurological disorder worldwide, affecting approximately 24 million individuals. Despite more than a century of research on AD, its pathophysiology is still not fully understood. Objective Recently, genetic studies of AD have focused on analyzing the general expression profile by employing high-throughput genomic techniques such as microarrays. Current research has leveraged bioinformatics advancements in genetic science to build upon previous efforts. Methods Data from the GSE118553 dataset used in this investigation, and the analyses carried out using programs such as Limma and BioBase. Differentially expressed genes (DEGs) and differentially expressed microRNAs (DEmiRs) associated with AD identified in the studied areas of the brain. Target genes of the DEmiRs identified using the MultiMiR package. Gene ontology (GO) completed using the Enrichr website, and the protein-protein interaction (PPI) network for these genes drawn using STRING and Cytoscape software. Results The findings introduced DEGs including CTNNB1, PAK2, MAP2K1, PNPLA6, IGF1R, FOXL2, DKK3, LAMA4, PABPN1, and GDPD5, and DEmiRs linked to AD (miR-106A, miR-1826, miR-1253, miR-10B, miR-18B, miR-101-2, miR-761, miR-199A1, miR-379 and miR-668), (miR-720, miR-218-2, miR-25, miR-602, miR-1226, miR-548K, miR-H1, miR-410, miR-548F2, miR-181A2), (miR-1470, miR-651, miR-544, miR-1826, miR-195, miR-610, miR-599, miR-323, miR-587 and miR-340), and (miR-1282, miR-1914, miR-642, miR-1323, miR-373, miR-323, miR-1322, miR-612, miR-606 and miR-758) in cerebellum, frontal cortex, temporal cortex, and entorhinal cortex, respectively. Conclusions The majority of the genes and miRNAs identified by our findings may be employed as biomarkers for prediction, diagnosis, or therapy response monitoring.
Collapse
Affiliation(s)
- Sakine Kavoosi
- Department of Biology, Faculty of Science, University of Sistan and Baluchestan, Zahedan, Iran
| | - Ali Shahraki
- Department of Biology, Faculty of Science, University of Sistan and Baluchestan, Zahedan, Iran
| | | |
Collapse
|
5
|
Wheeler KV, Irimia A, Braskie MN. Using Neuroimaging to Study Cerebral Amyloid Angiopathy and Its Relationship to Alzheimer's Disease. J Alzheimers Dis 2024; 97:1479-1502. [PMID: 38306032 DOI: 10.3233/jad-230553] [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] [Indexed: 02/03/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by amyloid-β aggregation in the media and adventitia of the leptomeningeal and cortical blood vessels. CAA is one of the strongest vascular contributors to Alzheimer's disease (AD). It frequently co-occurs in AD patients, but the relationship between CAA and AD is incompletely understood. CAA may drive AD risk through damage to the neurovascular unit and accelerate parenchymal amyloid and tau deposition. Conversely, early AD may also drive CAA through cerebrovascular remodeling that impairs blood vessels from clearing amyloid-β. Sole reliance on autopsy examination to study CAA limits researchers' ability to investigate CAA's natural disease course and the effect of CAA on cognitive decline. Neuroimaging allows for in vivo assessment of brain function and structure and can be leveraged to investigate CAA staging and explore its associations with AD. In this review, we will discuss neuroimaging modalities that can be used to investigate markers associated with CAA that may impact AD vulnerability including hemorrhages and microbleeds, blood-brain barrier permeability disruption, reduced cerebral blood flow, amyloid and tau accumulation, white matter tract disruption, reduced cerebrovascular reactivity, and lowered brain glucose metabolism. We present possible areas for research inquiry to advance biomarker discovery and improve diagnostics.
Collapse
Affiliation(s)
- Koral V Wheeler
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina Del Rey, CA, USA
| | - Andrei Irimia
- Ethel Percy Andrus Gerontology Center, USC Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Biomedical Engineering, Corwin D. Denney Research Center, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Meredith N Braskie
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina Del Rey, CA, USA
| |
Collapse
|
6
|
Cisterna-García A, Beric A, Ali M, Pardo JA, Chen HH, Fernandez MV, Norton J, Gentsch J, Bergmann K, Budde J, Perlmutter JS, Morris JC, Cruchaga C, Botia JA, Ibanez L. Cell-free RNA signatures predict Alzheimer's disease. iScience 2023; 26:108534. [PMID: 38089583 PMCID: PMC10711471 DOI: 10.1016/j.isci.2023.108534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/09/2023] [Accepted: 11/20/2023] [Indexed: 02/01/2024] Open
Abstract
There is a need for affordable, scalable, and specific blood-based biomarkers for Alzheimer's disease that can be applied to a population level. We have developed and validated disease-specific cell-free transcriptomic blood-based biomarkers composed by a scalable number of transcripts that capture AD pathobiology even in the presymptomatic stages of the disease. Accuracies are in the range of the current CSF and plasma biomarkers, and specificities are high against other neurodegenerative diseases.
Collapse
Affiliation(s)
- Alejandro Cisterna-García
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Departamento de Ingeniería de la Información y las Comunicaciones, Universidad de Murcia, Murcia, Spain
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Aleksandra Beric
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Muhammad Ali
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Jose Adrian Pardo
- Departamento de Ingeniería de la Información y las Comunicaciones, Universidad de Murcia, Murcia, Spain
| | - Hsiang-Han Chen
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Maria Victoria Fernandez
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Joanne Norton
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Jen Gentsch
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Kristy Bergmann
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - John Budde
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Joel S. Perlmutter
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Department of Radiology, Neuroscience, Physical Therapy, and Occupational Therapy, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - John C. Morris
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in Saint Louis, Saint Louis, MO, USA
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Department of Pathology and Immunology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in Saint Louis, Saint Louis, MO, USA
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Department of Genetics, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Juan A. Botia
- Departamento de Ingeniería de la Información y las Comunicaciones, Universidad de Murcia, Murcia, Spain
| | - Laura Ibanez
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
7
|
Gold D, Wisialowski C, Piryatinsky I, Malloy P, Correia S, Salloway S, Klinge P, Gaudet CE, Niermeyer M, Lee A. Longitudinal post-shunt outcomes in idiopathic normal pressure hydrocephalus with and without comorbid Alzheimer's disease. J Int Neuropsychol Soc 2023; 29:751-762. [PMID: 36515069 DOI: 10.1017/s1355617722000868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Alzheimer's disease (AD) is highly comorbid with idiopathic normal pressure hydrocephalus (iNPH) and may diminish the benefits of shunting; however, findings in this area are mixed. We examined postoperative outcomes, with emphases on cognition and utilization of novel scoring procedures to enhance sensitivity. METHODS Using participant data from an iNPH outcome study at Butler Hospital, a mixed effect model examined main and interaction effects of time since surgery (baseline, 3 months, 12 months, and 24-60 months) and AD comorbidity (20 iNPH and 11 iNPH+AD) on activities of daily living (ADLs) and iNPH symptoms. Regression modeling explored whether baseline variables predicted improvements 3 months postoperatively. RESULTS There were no group differences in gait, incontinence, and global cognition over time, and neither group showed changes in ADLs. Cognitive differences were observed postoperatively; iNPH patients showed stable improvements in working memory (p = 0.012) and response inhibition (p = 0.010), while iNPH + AD patients failed to maintain initial gains. Regarding predicting postoperative outcomes, baseline AD biomarkers did not predict shunt response at 3 months; however, older age at surgery predicted poorer cognitive outcomes (p = 0.04), and presurgical Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) (p = 0.035) and Mini-Mental Status Examination (MMSE) scores (p = 0.009) predicted improvements incontinence. CONCLUSION iNPH + AD may be linked with greater declines in aspects of executive functioning postoperatively relative to iNPH alone. While baseline AD pathology may not prognosticate shunt response, younger age appears linked with postsurgical cognitive improvement, and utilizing both brief and comprehensive cognitive measures may help predict improved incontinence. These results illustrate the potential benefits of surgery and inform postoperative expectations for those with iNPH + AD.
Collapse
Affiliation(s)
- Dov Gold
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Memory and Aging Program, Butler Hospital, Providence, RI, USA
- Department of Clinical Psychology, William James College, Newton, MA, USA
| | | | | | - Paul Malloy
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Memory and Aging Program, Butler Hospital, Providence, RI, USA
| | - Stephen Correia
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Memory and Aging Program, Butler Hospital, Providence, RI, USA
| | - Stephen Salloway
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Memory and Aging Program, Butler Hospital, Providence, RI, USA
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Petra Klinge
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Lifespan Physician Group, Rhode Island Hospital, Providence, RI, USA
| | - Charles E Gaudet
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Memory and Aging Program, Butler Hospital, Providence, RI, USA
| | - Madison Niermeyer
- Department of Physical Medicine & Rehabilitation, University of Utah, Salt Lake City, UT, USA
| | - Athene Lee
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Memory and Aging Program, Butler Hospital, Providence, RI, USA
| |
Collapse
|
8
|
Lepinay E, Cicchetti F. Tau: a biomarker of Huntington's disease. Mol Psychiatry 2023; 28:4070-4083. [PMID: 37749233 DOI: 10.1038/s41380-023-02230-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 09/27/2023]
Abstract
Developing effective treatments for patients with Huntington's disease (HD)-a neurodegenerative disorder characterized by severe cognitive, motor and psychiatric impairments-is proving extremely challenging. While the monogenic nature of this condition enables to identify individuals at risk, robust biomarkers would still be extremely valuable to help diagnose disease onset and progression, and especially to confirm treatment efficacy. If measurements of cerebrospinal fluid neurofilament levels, for example, have demonstrated use in recent clinical trials, other proteins may prove equal, if not greater, relevance as biomarkers. In fact, proteins such as tau could specifically be used to detect/predict cognitive affectations. We have herein reviewed the literature pertaining to the association between tau levels and cognitive states, zooming in on Alzheimer's disease, Parkinson's disease and traumatic brain injury in which imaging, cerebrospinal fluid, and blood samples have been interrogated or used to unveil a strong association between tau and cognition. Collectively, these areas of research have accrued compelling evidence to suggest tau-related measurements as both diagnostic and prognostic tools for clinical practice. The abundance of information retrieved in this niche of study has laid the groundwork for further understanding whether tau-related biomarkers may be applied to HD and guide future investigations to better understand and treat this disease.
Collapse
Affiliation(s)
- Eva Lepinay
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada
| | - Francesca Cicchetti
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada.
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada.
| |
Collapse
|
9
|
Neuroprotective Effect of Artichoke-Based Nanoformulation in Sporadic Alzheimer’s Disease Mouse Model: Focus on Antioxidant, Anti-Inflammatory, and Amyloidogenic Pathways. Pharmaceuticals (Basel) 2022; 15:ph15101202. [PMID: 36297313 PMCID: PMC9610800 DOI: 10.3390/ph15101202] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
The vast socio-economic impact of Alzheimer’s disease (AD) has prompted the search for new neuroprotective agents with good tolerability and safety profile. With its outstanding role as antioxidant and anti-inflammatory, alongside its anti-acetylcholinesterase activity, the artichoke can be implemented in a multi-targeted approach in AD therapy. Moreover, artichoke agricultural wastes can represent according to the current United Nations Sustainable Development goals an opportunity to produce medicinally valuable phenolic-rich extracts. In this context, the UPLC-ESI-MS/MS phytochemical characterization of artichoke bracts extract revealed the presence of mono- and di-caffeoylquinic acids and apigenin, luteolin, and kaempferol O-glycosides with remarkable total phenolics and flavonoids contents. A broad antioxidant spectrum was established in vitro. Artichoke-loaded, chitosan-coated, solid lipid nanoparticles (SLNs) were prepared and characterized for their size, zeta potential, morphology, entrapment efficiency, release, and ex vivo permeation and showed suitable colloidal characteristics, a controlled release profile, and promising ex vivo permeation, indicating possibly better physicochemical and biopharmaceutical parameters than free artichoke extract. The anti-Alzheimer potential of the extract and prepared SLNs was assessed in vivo in streptozotocin-induced sporadic Alzheimer mice. A great improvement in cognitive functions and spatial memory recovery, in addition to a marked reduction of the inflammatory biomarker TNF-α, β-amyloid, and tau protein levels, were observed. Significant neuroprotective efficacy in dentate Gyrus sub-regions was achieved in mice treated with free artichoke extract and to a significantly higher extent with artichoke-loaded SLNs. The results clarify the strong potential of artichoke bracts extract as a botanical anti-AD drug and will contribute to altering the future medicinal outlook of artichoke bracts previously regarded as agro-industrial waste.
Collapse
|
10
|
Seitkazina A, Kim KH, Fagan E, Sung Y, Kim YK, Lim S. The Fate of Tau Aggregates Between Clearance and Transmission. Front Aging Neurosci 2022; 14:932541. [PMID: 35923541 PMCID: PMC9339952 DOI: 10.3389/fnagi.2022.932541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/22/2022] [Indexed: 11/30/2022] Open
Abstract
Neuronal accumulation of mis-folded tau is the pathological hallmark of multiple neurodegenerative disorders, including Alzheimer’s disease. Distinct from amyloid plaques, which appear simultaneously throughout the brain, tau pathology develops first in a specific brain region and then propagates to neuroanatomically connected brain regions, exacerbating the disease. Due to the implication in disease progression, prevention of tau transmission is recognized as an important therapeutic strategy that can halt disease progression in the brain. Recently, accumulating studies have demonstrated diverse cellular mechanisms associated with cell-to-cell transmission of tau. Once transmitted, mis-folded tau species act as a prion-like seed for native tau aggregation in the recipient neuron. In this review, we summarize the diverse cellular mechanisms associated with the secretion and uptake of tau, and highlight tau-trafficking receptors, which mediate tau clearance or cell-to-cell tau transmission.
Collapse
Affiliation(s)
- Assel Seitkazina
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) School, University of Science and Technology (UST), Seoul, South Korea
| | - Kyu Hyeon Kim
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) School, University of Science and Technology (UST), Seoul, South Korea
| | - Erin Fagan
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, United States
| | - Yoonsik Sung
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) School, University of Science and Technology (UST), Seoul, South Korea
| | - Yun Kyung Kim
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) School, University of Science and Technology (UST), Seoul, South Korea
- *Correspondence: Yun Kyung Kim,
| | - Sungsu Lim
- Convergence Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Sungsu Lim,
| |
Collapse
|
11
|
Landry RL, Embers ME. Does Dementia Have a Microbial Cause? NEUROSCI 2022; 3:262-283. [PMID: 39483362 PMCID: PMC11523730 DOI: 10.3390/neurosci3020019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/11/2022] [Indexed: 11/03/2024] Open
Abstract
The potential contribution of pathogenic microbes to dementia-inducing disease is a subject of considerable importance. Alzheimer's disease (AD) is a neurocognitive disease that slowly destroys brain function, leading to cognitive decline and behavioral and psychiatric disorders. The histopathology of AD is associated with neuronal loss and progressive synaptic dysfunction, accompanied by the deposition of amyloid-β (Aβ) peptide in the form of parenchymal plaques and abnormal aggregated tau protein in the form of neurofibrillary tangles. Observational, epidemiological, experimental, and pathological studies have generated evidence for the complexity and possible polymicrobial causality in dementia-inducing diseases. The AD pathogen hypothesis states that pathogens and microbes act as triggers, interacting with genetic factors to initiate the accumulation of Aβ, hyperphosphorylated tau protein (p-tau), and inflammation in the brain. Evidence indicates that Borrelia sp., HSV-1, VZV (HHV-2), HHV-6/7, oral pathogens, Chlamydophila pneumoniae, and Candida albicans can infect the central nervous system (CNS), evade the immune system, and consequently prevail in the AD brain. Researchers have made significant progress in understanding the multifactorial and overlapping factors that are thought to take part in the etiopathogenesis of dementia; however, the cause of AD remains unclear.
Collapse
Affiliation(s)
- Remi L Landry
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA;
- Division of Immunology, Tulane National Primate Research Center, Tulane University Health Sciences, Covington, LA 70433, USA
| | - Monica E Embers
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA;
- Division of Immunology, Tulane National Primate Research Center, Tulane University Health Sciences, Covington, LA 70433, USA
| |
Collapse
|
12
|
Bjorkli C, Louet C, Flo TH, Hemler M, Sandvig A, Sandvig I. In Vivo Microdialysis in Mice Captures Changes in Alzheimer's Disease Cerebrospinal Fluid Biomarkers Consistent with Developing Pathology. J Alzheimers Dis 2021; 84:1781-1794. [PMID: 34719495 DOI: 10.3233/jad-210715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Preclinical models of Alzheimer's disease (AD) can provide valuable insights into the onset and progression of the disease, such as changes in concentrations of amyloid-β (Aβ) and tau in cerebrospinal fluid (CSF). However, such models are currently underutilized due to limited advancement in techniques that allow for longitudinal CSF monitoring. OBJECTIVE An elegant way to understand the biochemical environment in the diseased brain is intracerebral microdialysis, a method that has until now been limited to short-term observations, or snapshots, of the brain microenvironment. Here we draw upon patient-based findings to characterize CSF biomarkers in a commonly used preclinical mouse model for AD. METHODS Our modified push-pull microdialysis method was first validated ex vivo with human CSF samples, and then in vivo in an AD mouse model, permitting assessment of dynamic changes of CSF Aβ and tau and allowing for better translational understanding of CSF biomarkers. RESULTS We demonstrate that CSF biomarker changes in preclinical models capture what is observed in the brain; with a decrease in CSF Aβ observed when plaques are deposited, and an increase in CSF tau once tau pathology is present in the brain parenchyma. We found that a high molecular weight cut-off membrane allowed for simultaneous sampling of Aβ and tau, comparable to CSF collection by lumbar puncture in patients. CONCLUSION Our approach can further advance AD and other neurodegenerative research by following evolving neuropathology along the disease cascade via consecutive sampling from the same animal and can additionally be used to administer pharmaceutical compounds and assess their efficacy (Bjorkli, unpublished data).
Collapse
Affiliation(s)
- Christiana Bjorkli
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Claire Louet
- Center for Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Trude Helen Flo
- Center for Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mary Hemler
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Axel Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Clinical Neuroscience, Neuro, Head and Neck, Umeå University Hospital, Umeå, Sweden.,Department of Community Medicine and Rehabilitation, Neuro, Head and Neck, Umeå University Hospital, Umeå, Sweden
| | - Ioanna Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
13
|
Abbas H, Refai H, El Sayed N, Rashed LA, Mousa MR, Zewail M. Superparamagnetic iron oxide loaded chitosan coated bilosomes for magnetic nose to brain targeting of resveratrol. Int J Pharm 2021; 610:121244. [PMID: 34737114 DOI: 10.1016/j.ijpharm.2021.121244] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 01/16/2023]
Abstract
The objective of this study was to improve effectiveness of resveratrol (RES) through brain targeting by the intranasal olfactory mucosa for the treatment Alzheimer's disease (AD). To attain this, chitosan coated bilosomes (non ionic surfactant vesicles stabilized by bile salts, loaded with RES and superparamagnetic iron oxide nanoparticles (SPIONs) were prepared and incorporated into sodium alginate/PVP wafers. In vitro characterization of bilosomes including colloidal characteristics, entrapment efficiency and in vitro release was carried out. Hydration capacity, porosity percentage, morphology and in vitro release for selected wafer formulation were also investigated. Particle size of selected bilosomes, CS coated bilosome and SPION bilosomes was 208, 238 and 243 nm, respectively and they provided sustained RES release for 24 h. Both formulations were loaded in wafers and intra-nasally administered in mice with lipopolysaccharide induced AD model. Neurobehavioral tests, AD markers analysis, RT-PCR, western blotting and histopathological evaluation of the dissected brains were carried out. Results revealed the superiority of SPION bilosomes over conventional bilosomes and RES suspension in improving cognitive and memory functions, reduction of pro-inflammatory markers levels and down regulation of expression of NF-κB and P38. This may be attributed to enhanced RES therapeutic effects upon nanoencapsulation, loading into wafers, nasal administration and enhanced targeting the application of an external magnetic field.
Collapse
Affiliation(s)
- Haidy Abbas
- Department of Pharmaceutics, Damanhour University, Damanhour, Egypt.
| | - Hanan Refai
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th October City, Egypt.
| | - Nesrine El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Laila Ahmed Rashed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine Cairo University, Egypt
| | - Mohamed R Mousa
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Mariam Zewail
- Department of Pharmaceutics, Damanhour University, Damanhour, Egypt
| |
Collapse
|
14
|
Liu L, Liu L, Lu Y, Zhang T, Zhao W. Serum aberrant expression of miR-24-3p and its diagnostic value in Alzheimer's disease. Biomark Med 2021; 15:1499-1507. [PMID: 34668391 DOI: 10.2217/bmm-2021-0098] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aim: This study aimed to evaluate the effect of miR-24-3p in Alzheimer's disease (AD). Materials & methods: A total of 198 participants were recruited in this study, including 104 AD patients and 94 healthy controls. Expression of miR-24-3p was detected using quantitative real-time PCR. Receiver-operating characteristic curve was used to assess the diagnostic value of miR-24-3p. In vitro AD model was established to evaluate the effect of miR-24-3p. The downstream target was detected by luciferase reporter gene assay. Results: Expression of miR-24-3p showed 1.6-fold increase in AD group compared with healthy controls, and a negative correlation of miR-24-3p with mini-mental state examination score was obtained. Receiver-operating characteristic curve showed satisfactory diagnostic accuracy. Downregulation of miR-24-3p promoted cell proliferation and inhibited cell apoptosis. KLF8 is a target gene of miR-24-3p. Conclusion: MiR-24-3p has a certain value in the diagnosis of AD and may be a potential biomarker.
Collapse
Affiliation(s)
- Lina Liu
- Department of Neurology, Science & Technology Innovation Park of The Fourth Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, 150028, China
| | - Luran Liu
- Department of Neurology, Science & Technology Innovation Park of The Fourth Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, 150028, China
| | - Yunting Lu
- Department of Neurology, Science & Technology Innovation Park of The Fourth Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, 150028, China
| | - Tianyuan Zhang
- Department of Neurology, Science & Technology Innovation Park of The Fourth Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, 150028, China
| | - Wenting Zhao
- Department of Neurology, Science & Technology Innovation Park of The Fourth Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, 150028, China
| |
Collapse
|
15
|
Lussier FZ, Benedet AL, Therriault J, Pascoal TA, Tissot C, Chamoun M, Mathotaarachchi S, Savard M, Ashton NJ, Karikari TK, Rodriguez JL, Snellman A, Bezgin G, Kang MS, Fernandez Arias J, Wang YT, Gauthier S, Zetterberg H, Blennow K, Rosa-Neto P. Plasma levels of phosphorylated tau 181 are associated with cerebral metabolic dysfunction in cognitively impaired and amyloid-positive individuals. Brain Commun 2021; 3:fcab073. [PMID: 33959711 PMCID: PMC8088291 DOI: 10.1093/braincomms/fcab073] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease biomarkers are primarily evaluated through MRI, PET and CSF methods in order to diagnose and monitor disease. Recently, advances in the assessment of blood-based biomarkers have shown promise for simple, inexpensive, accessible and minimally invasive tools with diagnostic and prognostic value for Alzheimer's disease. Most recently, plasma phosphorylated tau181 has shown excellent performance. The relationship between plasma phosphorylated tau181 and cerebral metabolic dysfunction assessed by [18F]fluorodeoxyglucose PET in Alzheimer's disease is still unknown. This study was performed on 892 older individuals (297 cognitively unimpaired; 595 cognitively impaired) from the Alzheimer's Disease Neuroimaging Initiative cohort. Plasma phosphorylated tau181 was assessed using single molecular array technology and metabolic dysfunction was indexed by [18F]fluorodeoxyglucose PET. Cross-sectional associations between plasma and CSF phosphorylated tau181 and [18F]fluorodeoxyglucose were assessed using voxelwise linear regression models, with individuals stratified by diagnostic group and by β-amyloid status. Associations between baseline plasma phosphorylated tau181 and longitudinal (24 months) rate of brain metabolic decline were also assessed in 389 individuals with available data using correlations and voxelwise regression models. Plasma phosphorylated tau181 was elevated in β-amyloid positive and cognitively impaired individuals as well as in apolipoprotein E ε4 carriers and was significantly associated with age, worse cognitive performance and CSF phosphorylated tau181. Cross-sectional analyses showed strong associations between plasma phosphorylated tau181 and [18F]fluorodeoxyglucose PET in cognitively impaired and β-amyloid positive individuals. Voxelwise longitudinal analyses showed that baseline plasma phosphorylated tau181 concentrations were significantly associated with annual rates of metabolic decline in cognitively impaired individuals, bilaterally in the medial and lateral temporal lobes. The associations between plasma phosphorylated tau181 and reduced brain metabolism, primarily in cognitively impaired and in β-amyloid positive individuals, supports the use of plasma phosphorylated tau181 as a simple, low-cost, minimally invasive and accessible tool to both assess current and predict future metabolic dysfunction associated with Alzheimer's disease, comparatively to PET, MRI and CSF methods.
Collapse
Affiliation(s)
- Firoza Z Lussier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
| | - Andréa L Benedet
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
| | - Tharick A Pascoal
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
| | - Melissa Savard
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Juan Lantero Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Anniina Snellman
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Turku PET Centre, University of Turku, Turku, Finland
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
| | - Jaime Fernandez Arias
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
- Alzheimer’s Disease Research Unit, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montréal, QC, Canada
- Montréal Neurological Institute, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | | |
Collapse
|
16
|
Medala VK, Gollapelli B, Dewanjee S, Ogunmokun G, Kandimalla R, Vallamkondu J. Mitochondrial dysfunction, mitophagy, and role of dynamin-related protein 1 in Alzheimer's disease. J Neurosci Res 2021; 99:1120-1135. [PMID: 33465841 DOI: 10.1002/jnr.24781] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia and progressive neurodegenerative disease. The presence of β-amyloid (Aβ) plaques and phosphorylated Tau tangles are considered to be the two main hallmarks of AD. Recent findings have shown that different changes in the structure and dynamics of mitochondria play an important role in AD pathology progression. Mitochondrial changes in AD are expressed as enhanced mitochondrial fragmentation, altered mitochondrial dynamics, and changes in the expression of mitochondrial biogenesis genes in vitro and in vivo models. Therefore, targeting mitochondria and associated mitochondrial proteins seems to be a promising alternative instead of targeting Aβ and Tau in the prevention of Alzheimer's disease. The dynamin-related protein (Drp1) is one such protein that plays an important role in the regulation of mitochondrial division and maintenance of mitochondrial structures. Few researchers have shown that inhibition of Drp1 GTPase activity in neuronal cells rescues excessive mitochondrial fragmentation. In addition, the growing evidence revealed that Drp1 can interact with both Aβ and Tau protein in human brain tissues and mouse models. In this review, we would like to update existing knowledge about various changes in and around mitochondria related to the pathogenesis of Alzheimer's disease, with particular emphasis on mitophagy and autophagy.
Collapse
Affiliation(s)
| | - Buchaiah Gollapelli
- Department of Physics, National Institute of Technology-Warangal, Warangal, India
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | | | - Ramesh Kandimalla
- Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Department of Biochemistry, Kakatiya Medical College, Warangal, India
| | | |
Collapse
|
17
|
Khan MI, Hasan F, Hasan Al Mahmud KA, Adnan A. Domain focused and residue focused phosphorylation effect on tau protein: A molecular dynamics simulation study. J Mech Behav Biomed Mater 2020; 113:104149. [PMID: 33125954 DOI: 10.1016/j.jmbbm.2020.104149] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/13/2020] [Accepted: 10/17/2020] [Indexed: 11/19/2022]
Abstract
Phosphorylation has been hypothesized to alter the ability of tau protein to bind with microtubules (MT), and pathological level of phosphorylation can incorporate formation of Paired Helical Filaments (PHF) in affected tau. Study of the effect of phosphorylation on different domains of tau (projection domain, microtubule binding sites and N-terminus tail) is important to obtain insight about tau neuropathology. In an earlier study, we have already obtained the mechanical properties and behavior of single tau and dimerized tau and observed tau-MT interaction for normal level of phosphorylation. This study attempts to obtain insights on the effect of phosphorylation on different domains of tau, using molecular dynamics (MD) simulation with the aid of CHARMM force field under high strain rate. It also determines the effect of residue focused phosphorylation on tau-MT interaction and tau accumulation tendency. The results show that for single tau protein, unfolding stiffness does not differ significantly due to phosphorylation, but stretching stiffness can be much higher than the normally phosphorylated protein. For dimerized tau protein, the stretching required to separate the protein forming the dimer is similar for phosphorylation in individual domains but is significantly less in case of phosphorylation in all domains. For tau-MT interaction simulations, it is found that for normal phosphorylation, the tau separation from MT occurs at higher strain for phosphorylation in projection domain and N-terminus tail, and earlier for phosphorylation in all domains altogether than the normal phosphorylation state. The residue focused phosphorylation study also shows that tau separates earlier from MT and shows stronger accumulation tendency at the phosphorylated state, while preserving the highly stretchable and flexible characteristic of tau. This study provides important insight on mechanochemical phenomena relevant to traumatic brain injury (TBI) scenario, where the result of mechanical loading and posttranslational modification as well as conformation decides the mechanical behavior.
Collapse
Affiliation(s)
- Md Ishak Khan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| | - Fuad Hasan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| | | | - Ashfaq Adnan
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX, 76019, USA.
| |
Collapse
|
18
|
Fang WQ, Hwu WL, Chien YH, Yang SY, Chieh JJ, Chang LM, Huang AC, Lee NC, Chiu MJ. Composite Scores of Plasma Tau and β-Amyloids Correlate with Dementia in Down Syndrome. ACS Chem Neurosci 2020; 11:191-196. [PMID: 31799825 DOI: 10.1021/acschemneuro.9b00585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Dementia frequently occurs in Down syndrome (DS) patients, and early intervention is important in its management. We have previously demonstrated a positive correlation of plasma β-amyloid Aβ42 levels and negative correlations of Aβ40 and tau levels with dementia in DS. In this study, we examined more cases and constructed composite scores with both tau and amyloids to correlate with dementia in DS. Plasma Aβ42, Aβ40, and tau proteins were measured by an immunomagnetic reduction assay in DS patients. Data were randomly and repeatedly split into training and validating sets, and logistic regression was applied to calculate the area under the curve (AUC) for each biomarker. A total of 73 DS patients (among them, 23 had neurodegeneration) and 77 controls were recruited. In DS patients without dementia, plasma Aβ40 and tau levels were highly elevated, but Aβ42 levels were lower than those of the healthy controls. DS patients with dementia, compared with DS patients with no dementia, had a large decline in Aβ40 and tau but a rise in Aβ42. For biomarker scores correlating with dementia, Aβ40 revealed an AUC of 0.912; the composite score of Aβ40 × tau revealed an AUC of 0.953; and a combined composite score of 0.1 for Aβ40 × Tau +0.9 Tau × Aβ40/Aβ42 achieved the highest AUC of 0.965. Therefore, composite biomarker scores including both plasma tau and β-amyloid levels correlate with dementia in DS better than using individual biomarker scores. The pattern of tau decline and Aβ42 rise in DS patients with dementia are also different from previous findings in Alzheimer's disease.
Collapse
Affiliation(s)
- Wei-Quan Fang
- Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Wuh-Liang Hwu
- Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yin-Hsiu Chien
- Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | | | - Jen-Jie Chieh
- Institute of Electro-optical Science and Technology, National Taiwan Normal University, Taipei 116, Taiwan
| | - Lih-Maan Chang
- Department of Clinical Psychology, National Taiwan University Hospital, National Taiwan University, Taipei 100, Taiwan
| | - Ai-Chu Huang
- Department of Medical Genetics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ni-Chung Lee
- Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department of Psychology, National Taiwan University, Taipei 106, Taiwan
- Graduate Institute of Biomedical Engineering and Bioinformatics, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
19
|
Tsai CL, Liang CS, Lee JT, Su MW, Lin CC, Chu HT, Tsai CK, Lin GY, Lin YK, Yang FC. Associations between Plasma Biomarkers and Cognition in Patients with Alzheimer's Disease and Amnestic Mild Cognitive Impairment: A Cross-Sectional and Longitudinal Study. J Clin Med 2019; 8:jcm8111893. [PMID: 31698867 PMCID: PMC6912664 DOI: 10.3390/jcm8111893] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 01/16/2023] Open
Abstract
Brain degeneration in patients with Alzheimer's disease (AD) results from the accumulation of pathological amyloid- (Aβ) plaques and tau protein tangles, leading to altered plasma levels of biomarkers. However, few studies have investigated the association between plasma biomarkers and cognitive impairment in patients with AD. In this cross-sectional study, we investigated correlations between mini-mental state examination (MMSE) scores and levels of plasma biomarkers in patients with amnestic mild cognitive impairment (aMCI) and AD. Thirteen individuals with normal cognition, 40 patients with aMCI, and 37 patients with AD were enrolled. Immunomagnetic reduction was used to assess the levels of plasma biomarkers, including amyloid A1-40, A1-42, total tau protein (t-Tau), and phosphorylated tau protein (threonine 181, p-Tau181). Our analysis revealed a significant negative correlation between MMSE and both measures of tau, and a trend toward negative correlation between MMSE and A1-42. In a longitudinal study involving three patients with aMCI and two patients with AD, we observed strong negative correlations (r < -0.8) between changes in MMSE scores and plasma levels of t-Tau. Our results suggest that plasma levels of t-Tau and p-Tau181 can be used to assess the severity of cognitive impairment in patients with AD. Furthermore, the results of our preliminary longitudinal study suggest that levels of t-Tau can be used to monitor the progression of cognitive decline in patients with aMCI/AD.
Collapse
Affiliation(s)
- Chia-Lin Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chih-Sung Liang
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei 112, Taiwan
| | - Jiunn-Tay Lee
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Ming-Wei Su
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Chun-Chieh Lin
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Hsuan-Te Chu
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei 112, Taiwan
| | - Chia-Kuang Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Guan-Yu Lin
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Yu-Kai Lin
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Fu-Chi Yang
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Correspondence: ; Tel.: +886-2-879-233-11; Fax: +886-2-879-271-74
| |
Collapse
|
20
|
Guzman-Martinez L, Maccioni RB, Andrade V, Navarrete LP, Pastor MG, Ramos-Escobar N. Neuroinflammation as a Common Feature of Neurodegenerative Disorders. Front Pharmacol 2019; 10:1008. [PMID: 31572186 PMCID: PMC6751310 DOI: 10.3389/fphar.2019.01008] [Citation(s) in RCA: 488] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 08/08/2019] [Indexed: 12/26/2022] Open
Abstract
Neurodegenerative diseases share the fact that they derive from altered proteins that undergo an unfolding process followed by formation of β-structures and a pathological tendency to self-aggregate in neuronal cells. This is a characteristic of tau protein in Alzheimer’s disease and several tauopathies associated with tau unfolding, α-synuclein in Parkinson’s disease, and huntingtin in Huntington disease. Usually, the self-aggregation products are toxic to these cells, and toxicity spreads all over different brain areas. We have postulated that these protein unfolding events are the molecular alterations that trigger several neurodegenerative disorders. Most interestingly, these events occur as a result of neuroinflammatory cascades involving alterations in the cross-talks between glial cells and neurons as a consequence of the activation of microglia and astrocytes. The model we have hypothesized for Alzheimer’s disease involves damage signals that promote glial activation, followed by nuclear factor NF-kβ activation, synthesis, and release of proinflammatory cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-1, IL-6, and IL-12 that affect neuronal receptors with an overactivation of protein kinases. These patterns of pathological events can be applied to several neurodegenerative disorders. In this context, the involvement of innate immunity seems to be a major paradigm in the pathogenesis of these diseases. This is an important element for the search for potential therapeutic approaches for all these brain disorders.
Collapse
Affiliation(s)
- Leonardo Guzman-Martinez
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile
| | - Ricardo B Maccioni
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile.,Department of Neurological Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Víctor Andrade
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile
| | - Leonardo Patricio Navarrete
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile
| | - María Gabriela Pastor
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile.,Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Nicolas Ramos-Escobar
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile
| |
Collapse
|
21
|
Guzman-Martinez L, Maccioni RB, Farías GA, Fuentes P, Navarrete LP. Biomarkers for Alzheimer’s Disease. Curr Alzheimer Res 2019; 16:518-528. [DOI: 10.2174/1567205016666190517121140] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/08/2019] [Accepted: 04/25/2019] [Indexed: 12/15/2022]
Abstract
Alzheimer´s disease (AD) and related forms of dementia are increasingly affecting the aging population throughout the world, at an alarming rate. The World Alzheimer´s Report indicates a prevalence of 46.8 million people affected by AD worldwide. As population ages, this number is projected to triple by 2050 unless effective interventions are developed and implemented. Urgent efforts are required for an early detection of this disease. The ultimate goal is the identification of viable targets for the development of molecular markers and validation of their use for early diagnosis of AD that may improve treatment and the disease outcome in patients. The diagnosis of AD has been difficult to resolve since approaches for early and accurate detection and follow-up of AD patients at the clinical level have been reported only recently. Some proposed AD biomarkers include the detection of pathophysiological processes in the brain in vivo with new imaging techniques and novel PET ligands, and the determination of pathogenic proteins in cerebrospinal fluid showing anomalous levels of hyperphosphorylated tau and low Aβ peptide. These biomarkers have been increasingly accepted by AD diagnostic criteria and are important tools for the design of clinical trials, but difficulties in accessibility to costly and invasive procedures have not been completely addressed in clinical settings. New biomarkers are currently being developed to allow determinations of multiple pathological processes including neuroinflammation, synaptic dysfunction, metabolic impairment, protein aggregation and neurodegeneration. Highly specific and sensitive blood biomarkers, using less-invasive procedures to detect AD, are derived from the discoveries of peripheric tau oligomers and amyloid variants in human plasma and platelets. We have also developed a blood tau biomarker that correlates with a cognitive decline and also with neuroimaging determinations of brain atrophy.
Collapse
|
22
|
Majd S, Power J, Majd Z. Alzheimer's Disease and Cancer: When Two Monsters Cannot Be Together. Front Neurosci 2019; 13:155. [PMID: 30881282 PMCID: PMC6407038 DOI: 10.3389/fnins.2019.00155] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 02/11/2019] [Indexed: 12/29/2022] Open
Abstract
Alzheimer's disease (AD) and cancer are among the leading causes of human death around the world. While neurodegeneration is the main feature of AD, the most important characteristic of malignant tumors is cell proliferation, placing these two diseases in opposite sides of cell division spectrum. Interestingly, AD and cancer's pathologies consist of a remarkable common feature and that is the presence of active cell cycle in both conditions. In an in vitro model of primary adult neuronal culture, we previously showed that treating cell with beta amyloid forced neurons to start a cell cycle. Instead of cell division, however, neuronal cell cycle was aborted and a massive neurodegeneration was left behind as the consequence. A high level of cell cycle entry, which is a requirement for cancer pathogenesis, was reported in clinically diagnosed cases of AD, leading to neurodegeneration. The diverse clinical manifestation of a similar etiology, have puzzled researchers for many years. In fact, the evidence showed an inverse association between AD and cancer prevalence, suggesting that switching pathogenesis toward AD protects patients against cancer and vice versa. In this mini review, we discussed the possibility of involvement of cell proliferation and survival dysregulation as the underlying mechanism of neurodegeneration in AD, and the leading event to develop both disorders' pathology. As examples, the role of phosphoinositide 3 kinase/Akt/ mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway in cell cycle re-entry and blocking autophagy are discussed as potential common intracellular components between AD and cancer pathogenesis, with diverse clinical diagnosis.
Collapse
Affiliation(s)
- Shohreh Majd
- Neuronal Injury and Repair Laboratory, Centre for Neuroscience, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - John Power
- Neuronal Injury and Repair Laboratory, Centre for Neuroscience, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - Zohreh Majd
- Psychosomatische Tagesklinik, Passau, Germany
| |
Collapse
|
23
|
Ayob F, Lam E, Ho G, Chung F, El-Beheiry H, Wong J. Pre-operative biomarkers and imaging tests as predictors of post-operative delirium in non-cardiac surgical patients: a systematic review. BMC Anesthesiol 2019; 19:25. [PMID: 30797230 PMCID: PMC6387490 DOI: 10.1186/s12871-019-0693-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/08/2019] [Indexed: 02/06/2023] Open
Abstract
Background Post-operative delirium (POD) is a common post-operative complication in elderly individuals and imposes a significant health and financial burden. Identifying predictive biomarkers may help understand the pathophysiology of POD. Our objective is to summarize the evidence of pre-operative biomarkers and imaging tests to predict POD in patients undergoing non-cardiac surgery. Methods A systematic search of English language articles in MEDLINE, EMBASE, Cochrane Database, PsychINFO, PubMed and ClinicalTrials. Gov up to January 2018 was performed. Studies that used biomarkers or imaging tests to predict POD and a validated POD assessment tool were included. Animal studies, paediatric, cardiac and intracranial surgery were excluded. Risk of bias was assessed using the Quality In Prognosis Study tool. Results Thirty-four prospective cohort studies involving 4424 patients were included. Nineteen studies described serum tests [Interleukin-6, Insulin-like Growth Factor 1, C-Reactive Protein (CRP), cholinesterases, apolipoprotein-E genotype, leptin, hypovitaminosis, hypoalbuminaemia, gamma-amino butyric acid], 10 described cerebral-spinal fluid tests (monoamine precursor, melatonin, acute phase proteins, S100B and neurofibrillary tangles), and 5 described imaging tests. Two studies had high risk of bias due to unclear outcome measurement and study participation. CRP was significantly associated with POD in 5 studies. Other biomarkers were either examined by only a single study or two or more studies with conflicting results. Conclusion CRP is the most promising biomarker associated with POD. However, we are still in the early stages in identifying biomarkers and imaging tests that may further understanding of the pathophysiology of POD. Electronic supplementary material The online version of this article (10.1186/s12871-019-0693-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Farrah Ayob
- Department of Anesthesia, Toronto Western Hospital, University Health Network, 2-434 McLaughlin Wing, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada
| | - Enoch Lam
- Department of Anesthesia, Toronto Western Hospital, University Health Network, 2-434 McLaughlin Wing, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada
| | - George Ho
- Faculty of Medicine, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 2109, Toronto, ON, M5S 1A8, Canada
| | - Frances Chung
- Department of Anesthesia, Toronto Western Hospital, University Health Network, 2-434 McLaughlin Wing, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada
| | - Hossam El-Beheiry
- Department of Anesthesia, Trillium Health Partners, Mississauga Hospital, 100 Queensway, West, Mississauga, ON, L5B 1B, Canada
| | - Jean Wong
- Department of Anesthesia, Toronto Western Hospital, University Health Network, 2-434 McLaughlin Wing, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada. .,Women's College Hospital, Toronto, Ontario, 76 Grenville St, Toronto, ON, M5S 1B2, Canada.
| |
Collapse
|
24
|
Abstract
The pathological propagation of Tau protein is a hallmark of multiple neurodegenerative disorders, collectively referred to tauopathies with Alzheimer's disease (AD) being most prevalent, but including a range of frontotemporal dementias (FTDs). The extracellular Tau is important during the progression of tauopathies, although Tau is mainly expressed intracellularly for physiological functions. Extracellular Tau could be actively secreted by one cell then taken up by adjacent cells, leading to the cell-to-cell transmission of Tau. Accumulating evidence has demonstrated that Tau propagation is not only by the trans-synaptic spreading but also via exo-synaptic spreading pathways especially under the pathological conditions. Among these, exosomes, microvesicles and tunneling nanotubes (TNTs) are proposed exo-synaptic pathways for the spread of Tau pathology. These findings have led to the idea that extracellular Tau could be a novel therapeutic target to halt the propagation of Tau pathology. From this perspective, this charter focuses on recent advances in understanding the mechanisms of Tau secretion and discusses the role of such mechanisms in the development of Tau pathology.
Collapse
Affiliation(s)
- Zhi Ruan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Tsuneya Ikezu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
25
|
Chen Y, Fu AKY, Ip NY. Synaptic dysfunction in Alzheimer's disease: Mechanisms and therapeutic strategies. Pharmacol Ther 2018; 195:186-198. [PMID: 30439458 DOI: 10.1016/j.pharmthera.2018.11.006] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD), the most prevalent neurodegenerative disease in the elderly population, is characterized by progressive cognitive decline and pathological hallmarks of amyloid plaques and neurofibrillary tangles. However, its pathophysiological mechanisms are poorly understood, and diagnostic tools and interventions are limited. Here, we review recent research on the amyloid hypothesis and beta-amyloid-induced dysfunction of neuronal synapses through distinct cell surface receptors. We also review how tau protein leads to synaptotoxicity through pathological modification, localization, and propagation. Finally, we discuss experimental therapeutics for AD and propose potential applications of disease-modifying strategies targeting synaptic failure for improved treatment of AD.
Collapse
Affiliation(s)
- Yu Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China.
| | - Amy K Y Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Nancy Y Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China.
| |
Collapse
|
26
|
Waragai M, Moriya M, Nojo T. Decreased N-Acetyl Aspartate/Myo-Inositol Ratio in the Posterior Cingulate Cortex Shown by Magnetic Resonance Spectroscopy May Be One of the Risk Markers of Preclinical Alzheimer's Disease: A 7-Year Follow-Up Study. J Alzheimers Dis 2018; 60:1411-1427. [PMID: 28968236 PMCID: PMC5676849 DOI: 10.3233/jad-170450] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although molecular positron emission tomography imaging of amyloid and tau proteins can facilitate the detection of preclinical Alzheimer’s disease (AD) pathology, it is not useful in clinical practice. More practical surrogate markers for preclinical AD would provide valuable tools. Thus, we sought to validate the utility of conventional magnetic resonance spectroscopy (MRS) as a screening method for preclinical AD. A total of 289 older participants who were cognitively normal at baseline were clinically followed up for analysis of MRS metabolites, including N-acetyl aspartate (NAA) and myo-inositol (MI) in the posterior cingulate cortex (PCC) for 7 years. The 289 participants were retrospectively divided into five groups 7 years after baseline: 200 (69%) remained cognitively normal; 53 (18%) developed mild cognitive impairment (MCI); 21 (7%) developed AD; eight (2%) developed Parkinson’s disease with normal cognition, and seven (2%) developed dementia with Lewy bodies (DLB). The NAA/MI ratios of the PCC in the AD, MCI, and DLB groups were significantly decreased compared with participants who maintained normal cognition from baseline to 7 years after baseline. MMSE scores 7 years after baseline were significantly correlated with MI/Cr and NAA/MI ratios in the PCC. These results suggest that cognitively normal elderly subjects with low NAA/MI ratios in the PCC might be at risk of progression to clinical AD. Thus, the NAA/MI ratio in the PCC measured with conventional 1H MRS should be reconsidered as a possible adjunctive screening marker of preclinical AD in clinical practice.
Collapse
Affiliation(s)
- Masaaki Waragai
- Department of Neurology, Higashi Matsudo Municipal Hospital, Matsudo, Japan
| | - Masaru Moriya
- Division of Radiology, Higashi Matsudo Municipal Hospital, Matsudo, Japan
| | - Takeshi Nojo
- Department of Radiology, New Tokyo Hospital, Matsudo, Chiba, Japan
| |
Collapse
|
27
|
Yang CC, Chiu MJ, Chen TF, Chang HL, Liu BH, Yang SY. Assay of Plasma Phosphorylated Tau Protein (Threonine 181) and Total Tau Protein in Early-Stage Alzheimer’s Disease. J Alzheimers Dis 2018; 61:1323-1332. [DOI: 10.3233/jad-170810] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Che-Chuan Yang
- MagQu Co., Ltd., Xindian District, New Taipei City, Taiwan
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Psychology, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Biomedical Engineering and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hui-Ling Chang
- MagQu Co., Ltd., Xindian District, New Taipei City, Taiwan
| | - Bing-Hsien Liu
- MagQu Co., Ltd., Xindian District, New Taipei City, Taiwan
| | | |
Collapse
|
28
|
Higham JP. Tangled tau: Active pathology or footprint of disease? ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2017; 3:658-659. [PMID: 29255792 PMCID: PMC5725285 DOI: 10.1016/j.trci.2017.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- James P Higham
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
29
|
Lv F, Yang X, Cui C, Su C. Exogenous expression of Drp1 plays neuroprotective roles in the Alzheimer's disease in the Aβ42 transgenic Drosophila model. PLoS One 2017; 12:e0176183. [PMID: 28531191 PMCID: PMC5439651 DOI: 10.1371/journal.pone.0176183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 04/06/2017] [Indexed: 12/23/2022] Open
Abstract
Background Alzheimer's disease (AD) is one of the most common neurodegenerative disorders. Recent studies have shown that mitochondrial dysfunction is a causative factor of AD. Drp1 (Dynamin-related protein 1), a regulator of mitochondrial fission, shows neuroprotective effects on Parkinson’s disease. In this study, we investigate the effect and mechanism of Drp1 on Aβ42 transgenic Drosophila. Methods Elav-gal4/UAS>Aβ42 transgenic Drosophila model was constructed using Elav-gal4 promoter. The effects of Drp1 on the lifespan, motor ability and neuronal degeneration of the transgenic Drosophila were explored by over-expressing Drp1 in the Aβ42 transgenic Drosophila. ATP levels in the brain tissues of Aβ42 transgenic Drosophila were detected using high performance liquid chromatography (HPLC). Results Exogenous expression of Drp1 promoted crawling ability, reduced the levels of ATP in Drosophila brain and suppressed the neuronal degeneration. Conclusion The protective effect of Drp1 on the Aβ42 transgenic Drosophila was achieved by protecting the mitochondrial function, suggesting that Drp1 may be a potential therapeutic strategies for AD.
Collapse
Affiliation(s)
- Fengshou Lv
- Department of Pathology, Henan Medical College, Zhengzhou, China
| | - Xiaopeng Yang
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- * E-mail:
| | - Chuanju Cui
- Department of Neurology, Zhengzhou First People's Hospital, Zhengzhou, China
| | - Chunhe Su
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
30
|
Guo S, Lai C, Wu C, Cen G. Conversion Discriminative Analysis on Mild Cognitive Impairment Using Multiple Cortical Features from MR Images. Front Aging Neurosci 2017; 9:146. [PMID: 28572766 PMCID: PMC5435825 DOI: 10.3389/fnagi.2017.00146] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 05/01/2017] [Indexed: 01/18/2023] Open
Abstract
Neuroimaging measurements derived from magnetic resonance imaging provide important information required for detecting changes related to the progression of mild cognitive impairment (MCI). Cortical features and changes play a crucial role in revealing unique anatomical patterns of brain regions, and further differentiate MCI patients from normal states. Four cortical features, namely, gray matter volume, cortical thickness, surface area, and mean curvature, were explored for discriminative analysis among three groups including the stable MCI (sMCI), the converted MCI (cMCI), and the normal control (NC) groups. In this study, 158 subjects (72 NC, 46 sMCI, and 40 cMCI) were selected from the Alzheimer's Disease Neuroimaging Initiative. A sparse-constrained regression model based on the l2-1-norm was introduced to reduce the feature dimensionality and retrieve essential features for the discrimination of the three groups by using a support vector machine (SVM). An optimized strategy of feature addition based on the weight of each feature was adopted for the SVM classifier in order to achieve the best classification performance. The baseline cortical features combined with the longitudinal measurements for 2 years of follow-up data yielded prominent classification results. In particular, the cortical thickness produced a classification with 98.84% accuracy, 97.5% sensitivity, and 100% specificity for the sMCI–cMCI comparison; 92.37% accuracy, 84.78% sensitivity, and 97.22% specificity for the cMCI–NC comparison; and 93.75% accuracy, 92.5% sensitivity, and 94.44% specificity for the sMCI–NC comparison. The best performances obtained by the SVM classifier using the essential features were 5–40% more than those using all of the retained features. The feasibility of the cortical features for the recognition of anatomical patterns was certified; thus, the proposed method has the potential to improve the clinical diagnosis of sub-types of MCI and predict the risk of its conversion to Alzheimer's disease.
Collapse
Affiliation(s)
- Shengwen Guo
- Department of Biomedical Engineering, South China University of TechnologyGuangzhou, China
| | - Chunren Lai
- Department of Biomedical Engineering, South China University of TechnologyGuangzhou, China
| | - Congling Wu
- Department of Biomedical Engineering, South China University of TechnologyGuangzhou, China
| | - Guiyin Cen
- Guangdong General HospitalGuangzhou, China
| | | |
Collapse
|
31
|
Almdahl IS, Lauridsen C, Selnes P, Kalheim LF, Coello C, Gajdzik B, Møller I, Wettergreen M, Grambaite R, Bjørnerud A, Bråthen G, Sando SB, White LR, Fladby T. Cerebrospinal Fluid Levels of Amyloid Beta 1-43 Mirror 1-42 in Relation to Imaging Biomarkers of Alzheimer's Disease. Front Aging Neurosci 2017; 9:9. [PMID: 28223932 PMCID: PMC5293760 DOI: 10.3389/fnagi.2017.00009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/12/2017] [Indexed: 11/24/2022] Open
Abstract
Introduction: Amyloid beta 1-43 (Aβ43), with its additional C-terminal threonine residue, is hypothesized to play a role in early Alzheimer’s disease pathology possibly different from that of amyloid beta 1-42 (Aβ42). Cerebrospinal fluid (CSF) Aβ43 has been suggested as a potential novel biomarker for predicting conversion from mild cognitive impairment (MCI) to dementia in Alzheimer’s disease. However, the relationship between CSF Aβ43 and established imaging biomarkers of Alzheimer’s disease has never been assessed. Materials and Methods: In this observational study, CSF Aβ43 was measured with ELISA in 89 subjects; 34 with subjective cognitive decline (SCD), 51 with MCI, and four with resolution of previous cognitive complaints. All subjects underwent structural MRI; 40 subjects on a 3T and 50 on a 1.5T scanner. Forty subjects, including 24 with SCD and 12 with MCI, underwent 18F-Flutemetamol PET. Seventy-eight subjects were assessed with 18F-fluorodeoxyglucose PET (21 SCD/7 MCI and 11 SCD/39 MCI on two different scanners). Ten subjects with SCD and 39 with MCI also underwent diffusion tensor imaging. Results: Cerebrospinal fluid Aβ43 was both alone and together with p-tau a significant predictor of the distinction between SCD and MCI. There was a marked difference in CSF Aβ43 between subjects with 18F-Flutemetamol PET scans visually interpreted as negative (37 pg/ml, n = 27) and positive (15 pg/ml, n = 9), p < 0.001. Both CSF Aβ43 and Aβ42 were negatively correlated with standardized uptake value ratios for all analyzed regions; CSF Aβ43 average rho -0.73, Aβ42 -0.74. Both CSF Aβ peptides correlated significantly with hippocampal volume, inferior parietal and frontal cortical thickness and axial diffusivity in the corticospinal tract. There was a trend toward CSF Aβ42 being better correlated with cortical glucose metabolism. None of the studied correlations between CSF Aβ43/42 and imaging biomarkers were significantly different for the two Aβ peptides when controlling for multiple testing. Conclusion: Cerebrospinal fluid Aβ43 appears to be strongly correlated with cerebral amyloid deposits in the same way as Aβ42, even in non-demented patients with only subjective cognitive complaints. Regarding imaging biomarkers, there is no evidence from the present study that CSF Aβ43 performs better than the classical CSF biomarker Aβ42 for distinguishing SCD and MCI.
Collapse
Affiliation(s)
- Ina S Almdahl
- Division of Medicine and Laboratory Sciences, Institute of Clinical Medicine, Faculty of Medicine, University of OsloOslo, Norway; Department of Neurology, Akershus University HospitalLørenskog, Norway
| | - Camilla Lauridsen
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and Technology Trondheim, Norway
| | - Per Selnes
- Division of Medicine and Laboratory Sciences, Institute of Clinical Medicine, Faculty of Medicine, University of OsloOslo, Norway; Department of Neurology, Akershus University HospitalLørenskog, Norway
| | - Lisa F Kalheim
- Division of Medicine and Laboratory Sciences, Institute of Clinical Medicine, Faculty of Medicine, University of OsloOslo, Norway; Department of Neurology, Akershus University HospitalLørenskog, Norway
| | - Christopher Coello
- Preclinical PET/CT, Institute of Basic Medical Sciences, University of Oslo Oslo, Norway
| | | | - Ina Møller
- Department of Neurology and Clinical Neurophysiology, University Hospital of Trondheim Trondheim, Norway
| | - Marianne Wettergreen
- Department of Neurology, Akershus University HospitalLørenskog, Norway; Department of Clinical Molecular Biology (EpiGen), Institute of Clinical Medicine, University of Oslo - Akershus University HospitalLørenskog, Norway
| | - Ramune Grambaite
- Department of Neurology, Akershus University Hospital Lørenskog, Norway
| | - Atle Bjørnerud
- The Intervention Centre, Oslo University Hospital Oslo, Norway
| | - Geir Bråthen
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and TechnologyTrondheim, Norway; Department of Neurology and Clinical Neurophysiology, University Hospital of TrondheimTrondheim, Norway
| | - Sigrid B Sando
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and TechnologyTrondheim, Norway; Department of Neurology and Clinical Neurophysiology, University Hospital of TrondheimTrondheim, Norway
| | - Linda R White
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and TechnologyTrondheim, Norway; Department of Neurology and Clinical Neurophysiology, University Hospital of TrondheimTrondheim, Norway
| | - Tormod Fladby
- Division of Medicine and Laboratory Sciences, Institute of Clinical Medicine, Faculty of Medicine, University of OsloOslo, Norway; Department of Neurology, Akershus University HospitalLørenskog, Norway
| |
Collapse
|
32
|
Lee NC, Yang SY, Chieh JJ, Huang PT, Chang LM, Chiu YN, Huang AC, Chien YH, Hwu WL, Chiu MJ. Blood Beta-Amyloid and Tau in Down Syndrome: A Comparison with Alzheimer's Disease. Front Aging Neurosci 2017; 8:316. [PMID: 28144219 PMCID: PMC5239773 DOI: 10.3389/fnagi.2016.00316] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 12/08/2016] [Indexed: 11/13/2022] Open
Abstract
Background: Changes in β-amyloids (Aβ) and tau proteins have been noted in patients with Alzheimer's disease (AD) and patients with both Down syndrome (DS) and AD. However, reports of changes in the early stage of regression, such as behavioral and psychological symptoms of dementia (BPSD), in DS are sparse. Methods: Seventy-eight controls, 62 patients with AD, 35 with DS and 16 with DS with degeneration (DS_D), including 9 with BPSD and 7 with dementia, were enrolled. The levels of β-amyloids 40 and 42 (Aβ-40, Aβ-42) and tau protein in the blood were analyzed using immunomagnetic reduction (IMR). The Adaptive Behavior Dementia Questionnaire (ABDQ) was used to evaluate the clinical status of the degeneration. Results: The Aβ-40 and tau levels were higher and the Aβ-42 level and Aβ-42/Aβ-40 ratio were lower in DS than in the controls (all p < 0.001). Decreased Aβ-40 and increased Aβ-42 levels and Aβ-42/40 ratios were observed in DS_D compared with DS without degeneration (all p < 0.001). The ABDQ score was negatively correlated with the Aβ-40 level (ρ = -0.556) and the tau protein level (ρ = -0.410) and positively associated with the Aβ-42 level (ρ = 0.621) and the Aβ-42/40 ratio (ρ = 0.544; all p < 0.05). Conclusions: The Aβ-40 and Aβ-42 levels and the Aβ-42/Aβ-40 ratio are considered possible biomarkers for the early detection of degeneration in DS. The elevated Aβ-40 and tau levels in DS may indicate early neurodegeneration. The increased Aβ-42 in DS_D may reflect the neurotoxicity of Aβ-42. The paradox of the tau decreases in DS_D could be explained by a burnout phenomenon during long-term neurodegeneration. The different patterns of the plasma beta amyloids and tau protein may imply a different pathogenesis between DS with degeneration and AD in the general population, in spite of their common key pathological features.
Collapse
Affiliation(s)
- Ni-Chung Lee
- Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan UniversityTaipei, Taiwan; Department of Medical Genetics, National Taiwan University Hospital, College of Medicine, National Taiwan UniversityTaipei, Taiwan
| | | | - Jen-Jie Chieh
- Institute of Electro-Optical Science and Technology, National Taiwan Normal University Taipei, Taiwan
| | - Po-Tsang Huang
- Department of Clinical Psychology Center, National Taiwan University Hospital, College of Medicine, National Taiwan University Taipei, Taiwan
| | - Lih-Maan Chang
- Department of Clinical Psychology Center, National Taiwan University Hospital, College of Medicine, National Taiwan University Taipei, Taiwan
| | - Yen-Nan Chiu
- Department of Psychiatry, National Taiwan University Hospital, College of Medicine, National Taiwan University Taipei, Taiwan
| | - Ai-Chiu Huang
- Department of Medical Genetics, National Taiwan University Hospital, College of Medicine, National Taiwan University Taipei, Taiwan
| | - Yin-Hsiu Chien
- Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan UniversityTaipei, Taiwan; Department of Medical Genetics, National Taiwan University Hospital, College of Medicine, National Taiwan UniversityTaipei, Taiwan
| | - Wuh-Liang Hwu
- Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan UniversityTaipei, Taiwan; Department of Medical Genetics, National Taiwan University Hospital, College of Medicine, National Taiwan UniversityTaipei, Taiwan
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan UniversityTaipei, Taiwan; Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan UniversityTaipei, Taiwan; Department of Psychology, National Taiwan UniversityTaipei, Taiwan; Graduate Institute of Biomedical Engineering and Bioinformatics, National Taiwan UniversityTaipei, Taiwan
| |
Collapse
|
33
|
Neto E, Biessmann F, Aurlien H, Nordby H, Eichele T. Regularized Linear Discriminant Analysis of EEG Features in Dementia Patients. Front Aging Neurosci 2016; 8:273. [PMID: 27965568 PMCID: PMC5127828 DOI: 10.3389/fnagi.2016.00273] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 10/31/2016] [Indexed: 10/24/2022] Open
Abstract
The present study explores if EEG spectral parameters can discriminate between healthy elderly controls (HC), Alzheimer's disease (AD) and vascular dementia (VaD) using. We considered EEG data recorded during normal clinical routine with 114 healthy controls (HC), 114 AD, and 114 VaD patients. The spectral features extracted from the EEG were the absolute delta power, decay from lower to higher frequencies, amplitude, center and dispersion of the alpha power and baseline power of the entire frequency spectrum. For discrimination, we submitted these EEG features to regularized linear discriminant analysis algorithm with a 10-fold cross-validation. To check the consistency of the results obtained by our classifiers, we applied bootstrap statistics. Four binary classifiers were used to discriminate HC from AD, HC from VaD, AD from VaD, and HC from dementia patients (AD or VaD). For each model, we measured the discrimination performance using the area under curve (AUC) and the accuracy of the cross-validation (cv-ACC). We applied this procedure using two different sets of predictors. The first set considered all the features extracted from the 22 channels. For the second set of features, we automatically rejected features poorly correlated with their labels. Fairly good results were obtained when discriminating HC from dementia patients with AD or VaD (AUC = 0.84). We also obtained AUC = 0.74 for discrimination of AD from HC, AUC = 0.77 for discrimination of VaD from HC, and finally AUC = 0.61 for discrimination of AD from VaD. Our models were able to separate HC from dementia patients, and also and to discriminate AD from VaD above chance. Our results suggest that these features may be relevant for the clinical assessment of patients with dementia.
Collapse
Affiliation(s)
- Emanuel Neto
- Section for Clinical Neurophysiology, Haukeland University HospitalBergen, Norway; Institute of Biological and Medical Psychology, University of BergenBergen, Norway
| | | | - Harald Aurlien
- Section for Clinical Neurophysiology, Haukeland University Hospital Bergen, Norway
| | - Helge Nordby
- Institute of Biological and Medical Psychology, University of Bergen Bergen, Norway
| | - Tom Eichele
- Section for Clinical Neurophysiology, Haukeland University HospitalBergen, Norway; Institute of Biological and Medical Psychology, University of BergenBergen, Norway; K.G. Jebsen Center for Neuropsychiatric DisordersBergen, Norway
| |
Collapse
|
34
|
Qin Y, Gu JW. A Surgical Method to Improve the Homeostasis of CSF for the Treatment of Alzheimer's Disease. Front Aging Neurosci 2016; 8:261. [PMID: 27853433 PMCID: PMC5090002 DOI: 10.3389/fnagi.2016.00261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 10/19/2016] [Indexed: 11/13/2022] Open
Abstract
Reduced cerebrospinal fluid (CSF) production and increased resistance to CSF outflow are considered to be associated with aging, and are also characteristics of Alzheimer's disease (AD). These changes probably result in a decrease in the efficiency of the mechanism by which CSF removes toxic molecules such as amyloid-β (Aβ) and tau from the interstitial fluid space. Soluble Aβ is potently neurotoxic and dysfunctional in CSF circulation and can accelerate the progression of AD. Current therapies for AD exhibit poor efficiency; therefore, a surgical method to improve the homeostasis of CSF is worthy of investigation. To achieve this, we conceived a novel device, which consists of a ventriculo-peritoneal shunt, an injection port and a portable infusion pump. Artificial CSF (ACSF) is pumped into the ventricles and the ACSF composition, infusion modes and pressure threshold of shunting can be adjusted according to the intracranial pressure and CSF contents. We hypothesize that this active treatment for CSF circulation dysfunction will significantly retard the progression of AD.
Collapse
Affiliation(s)
- Yang Qin
- Department of Neurosurgery, Chengdu Military General Hospital Chengdu, China
| | - Jian W Gu
- Department of Neurosurgery, Chengdu Military General HospitalChengdu, China; Department of Neurosurgery, The 306th Hospital of PLABeijing, China
| |
Collapse
|
35
|
Kandimalla R, Manczak M, Fry D, Suneetha Y, Sesaki H, Reddy PH. Reduced dynamin-related protein 1 protects against phosphorylated Tau-induced mitochondrial dysfunction and synaptic damage in Alzheimer's disease. Hum Mol Genet 2016; 25:4881-4897. [PMID: 28173111 PMCID: PMC6078590 DOI: 10.1093/hmg/ddw312] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 08/31/2016] [Accepted: 09/02/2016] [Indexed: 11/13/2022] Open
Abstract
The purpose of our study was to understand the protective effects of a partial reduction of dynamin-related protein 1 (Drp1) in Alzheimer’s disease (AD) progression and pathogenesis. Increasing evidence suggests that phosphorylated Tau and mitochondrial abnormalities are involved in the loss of synapses, defective axonal transport and cognitive decline, in patients with AD. In the current study, we investigated whether a partial reduction of Drp1 protect neurons from phosphorylated Tau-induced mitochondrial and synaptic toxicities in AD progression. We crossed Drp1+/− mice with Tau transgenic mice (P301L line) and created double mutant (TauXDrp1+/−) mice. Using real-time RT-PCR, immunoblotting and immunostaining analyses, we measured mRNA expressions and protein levels of genes related to the mitochondrial dynamics—Drp1 and Fis1 (fission), Mfn1, Mfn2 and Opa1 (fusion), CypD (matrix), mitochondrial biogenesis—Nrf1, Nrf2, PGC1α and TFAM and synaptic—synaptophysin, PSD95, synapsin 1, synaptobrevin 1, neurogranin, GAP43 and synaptopodin in brain tissues from 6-month-old Drp1+/−, Tau, TauXDrp1+/− and wild-type mice. Using biochemical and immunoblotting methods, mitochondrial function and phosphorylated Tau were measured. Decreased mRNA and protein levels of fission and matrix and increased levels of fusion, mitochondrial biogenesis, and synaptic genes were found in 6-month-old TauXDrp1+/− mice relative to Tau mice. Mitochondrial dysfunction was reduced in TauXDrp1+/− mice relative to Tau mice. Phosphorylated Tau found to be reduced in TauXDrp1+/− mice relative to Tau mice. These findings suggest that a partial reduction of Drp1 decreases the production of phosphorylated Tau, reduces mitochondrial dysfunction, and maintains mitochondrial dynamics, enhances mitochondrial biogenesis and synaptic activity in Tau mice. Findings of this study may have implications for the development of Drp1 based therapeutics for patients with AD and other tauopathies.
Collapse
Affiliation(s)
- Ramesh Kandimalla
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, USA
| | - Maria Manczak
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, USA
| | - David Fry
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, USA
| | - Yeguvapalli Suneetha
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, USA
| | - Hiromi Sesaki
- Cell Biology Department, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, 109 Hunterian, Baltimore, MD 21205, USA
| | - P. Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, USA
- Cell Biology & Biochemistry Department
- Neuroscience & Pharmacology Department
- Neurology Department
- Speech, Language and Hearing Sciences Departments, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, USA
- Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, 6630 S. Quaker Suite E, MS 7495, Lubbock, TX 79413, USA
| |
Collapse
|
36
|
Jiang J, Wang H, Li W, Cao X, Li C. Amyloid Plaques in Retina for Diagnosis in Alzheimer's Patients: a Meta-Analysis. Front Aging Neurosci 2016; 8:267. [PMID: 27891091 PMCID: PMC5102884 DOI: 10.3389/fnagi.2016.00267] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/24/2016] [Indexed: 12/20/2022] Open
Abstract
Background: Detection of retinal β-amyloid (Aβ) peptide accumulation is a novel diagnostic method for Alzheimer's disease (AD), but there is, as yet, no conclusive evidence of its accuracy. Aim: To identify the diagnostic accuracy of pathological retinal Aβ detection for AD by a meta-analytic approach. Methods: Electronic and reference searches were conducted to identify studies related to the diagnostic effects of retinal Aβ detection in AD that met pre-defined inclusion criteria. The QUADAS-2 tool was employed to assess the risk of bias, and Review Manager plus the Open Meta-Analyst were used to perform the data analysis. Results: From 493 unduplicated reports, five studies with small sample sizes were included in this review. Six staining methods were employed. The eligible studies showed extremely broad ranges of sensitivity (0-1.00) and specificity (0.50-1.00) with substantial heterogeneity. The estimates of positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR) were also extremely varied (from 0.71 to 11.57 for PLR, from 0.04 to 1.11 for NLR, and from 0.69 to 297.00 for DOR). Conclusions: The limited number of eligible studies and their methodological heterogeneity make it impossible to come to a conclusion whether pathological retinal Aβ detection is an effective diagnostic tool for AD. More studies, especially large surveys investigating retina Aβ load with quantitative methods among consecutive or random samples, are needed to determine the accuracy of Aβ detection for diagnosing AD.
Collapse
Affiliation(s)
- Jiangling Jiang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Hongyan Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Wei Li
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Xinyi Cao
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Chunbo Li
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of MedicineShanghai, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong UniversityShanghai, China
| |
Collapse
|
37
|
Kumar S, Reddy PH. Are circulating microRNAs peripheral biomarkers for Alzheimer's disease? BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:1617-27. [PMID: 27264337 PMCID: PMC5343750 DOI: 10.1016/j.bbadis.2016.06.001] [Citation(s) in RCA: 226] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/13/2016] [Accepted: 06/01/2016] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by memory loss, multiple cognitive abnormalities and intellectual impairments. Currently, there are no drugs or agents that can delay and/or prevent the progression of disease in elderly individuals, and there are no peripheral biomarkers that can detect AD early in its pathogenesis. Research has focused on identifying biomarkers for AD so that treatment can be begun as soon as possible in order to restrict or prevent intellectual impairments, memory loss, and other cognitive abnormalities that are associated with the disease. One such potential biomarker is microRNAs that are found in circulatory biofluids, such as blood and blood components, serum and plasma. Blood and blood components are primary sources where miRNAs are released in either cell-free form and then bind to protein components, or are in an encapsulated form with microvesicle particles. Exosomal miRNAs are known to be stable in biofluids and can be detected by high throughput techniques, like microarray and RNA sequencing. In AD brain, enriched miRNAs encapsulated with exosomes crosses the blood brain barrier and secreted in the CSF and blood circulations. This review summarizes recent studies that have identified miRNAs in the blood, serum, plasma, exosomes, cerebral spinal fluids, and extracellular fluids as potential biomarkers of AD. Recent research has revealed only six miRNAs - miR-9, miR-125b, miR-146a, miR-181c, let-7g-5p, and miR-191-5p - that were reported by multiple investigators. Some studies analyzed the diagnostic potential of these six miRNAs through receiver operating curve analysis which indicates the significant area-under-curve values in different biofluid samples. miR-191-5p was found to have the maximum area-under-curve value (0.95) only in plasma and serum samples while smaller area-under-curve values were found for miR-125, miR-181c, miR-191-5p, miR-146a, and miR-9. This article shortlisted the promising miRNA candidates and discussed their diagnostic properties and cellular functions in order to search for potential biomarker for AD.
Collapse
Affiliation(s)
- Subodh Kumar
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States
| | - P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Neurology Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Speech, Language and Hearing Sciences Departments, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, 6630 S. Quaker Ste. E, MS 7495, Lubbock, TX 79413, United States.
| |
Collapse
|
38
|
Shekhar S, Kumar R, Rai N, Kumar V, Singh K, Upadhyay AD, Tripathi M, Dwivedi S, Dey AB, Dey S. Estimation of Tau and Phosphorylated Tau181 in Serum of Alzheimer's Disease and Mild Cognitive Impairment Patients. PLoS One 2016; 11:e0159099. [PMID: 27459603 PMCID: PMC4961391 DOI: 10.1371/journal.pone.0159099] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/27/2016] [Indexed: 12/15/2022] Open
Abstract
The elevated level of cerebrospinal fluid (CSF) Tau and phosphorylated Tau181 (p-Tau181) proteins are well established hallmarks of Alzheimer's disease (AD). Elevated level of p-Tau181 can differentiate AD from other neurodegenerative disease. However, the expression level of these proteins in serum of AD patient is not well set up. This study sought to evaluate the level of Tau and p-Tau181 in serum of AD, and mild cognitive impairment (MCI) patients for an alternative approach to establish protein-based markers by convenient way. Blood samples were collected from 39 AD patients, 37 MCI patients and 37 elderly individuals as controls. The levels of Tau and p-Tau181 in the serum of the different groups were measured by label free real time Surface Plasmon Resonance technology by using specific antibodies, and were further confirmed by the conventional western blot method. An appropriate statistical analysis, including Receiver Operating Characteristic (ROC), was performed. The concentrations of serum Tau and p-Tau181 were significantly higher (p<0.00001) in AD (Tau; 47.49±9.00ng/μL, p-Tau181; 0.161±0.04 ng/μL) compared to MCI (Tau; 39.26±7.78 ng/μL, p-Tau181; 0.135±0.02 ng/μL) and were further higher compared to elderly controls (Tau; 34.92±6.58 ng/μL, p-Tau181; 0.122±0.01 ng/ μL). A significant (p<0.0001) downhill correlation was found between Tau as well as p-Tau181 levels with HMSE and MoCA score. This study for the first time reports the concentration of Tau and p-Tau181 in serum of AD and MCI patients. The cutoff values of Tau and p-Tau181 of AD and MCI patients with sensitivity and specificity reveal that serum level of these proteins can be used as a predictive marker for AD and MCI.
Collapse
Affiliation(s)
- Shashank Shekhar
- Department of Biophysics of All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Rahul Kumar
- Department of Biophysics of All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Nitish Rai
- Department of Biophysics of All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Vijay Kumar
- Department of Geriatric Medicine of All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Kusum Singh
- Department of Biophysics of All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Ashish Datt Upadhyay
- Department of Biostatistics of All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Manjari Tripathi
- Department of Neurology of All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sadanand Dwivedi
- Department of Biostatistics of All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Aparajit B. Dey
- Department of Geriatric Medicine of All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sharmistha Dey
- Department of Biophysics of All India Institute of Medical Sciences, New Delhi, 110029, India
- * E-mail:
| |
Collapse
|
39
|
Starks EJ, Patrick O'Grady J, Hoscheidt SM, Racine AM, Carlsson CM, Zetterberg H, Blennow K, Okonkwo OC, Puglielli L, Asthana S, Dowling NM, Gleason CE, Anderson RM, Davenport-Sis NJ, DeRungs LM, Sager MA, Johnson SC, Bendlin BB. Insulin Resistance is Associated with Higher Cerebrospinal Fluid Tau Levels in Asymptomatic APOEɛ4 Carriers. J Alzheimers Dis 2016; 46:525-33. [PMID: 25812851 DOI: 10.3233/jad-150072] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Insulin resistance (IR) is linked with the occurrence of pathological features observed in Alzheimer's disease (AD), including neurofibrillary tangles and amyloid plaques. However, the extent to which IR is associated with AD pathology in the cognitively asymptomatic stages of preclinical AD remains unclear. OBJECTIVE To determine the extent to which IR is linked with amyloid and tau pathology in late-middle-age. METHOD Cerebrospinal fluid (CSF) samples collected from 113 participants enrolled in the Wisconsin Registry for Alzheimer's Prevention study (mean age = 60.6 years), were assayed for AD-related markers of interest: Aβ₄₂, P-Tau181, and T-Tau. IR was determined using the Homeostatic Model Assessment for Insulin Resistance (HOMA-IR). Linear regression was used to test the effect of IR, and APOEɛ4, on tau and amyloid pathology. We hypothesized that greater IR would be associated with higher CSF P-Tau181 and T-Tau, and lower CSF Aβ₄₂. RESULTS No significant main effects of HOMA-IR on P-Tau181, T-Tau, or Aβ₄₂ were observed; however, significant interactions were observed between HOMA-IR and APOEɛ4 on CSF markers related to tau. Among APOEɛ4 carriers, higher HOMA-IR was associated with higher P-Tau181 and T-Tau. Among APOEɛ4 non-carriers, HOMA-IR was negatively associated with P-Tau181 and T-Tau. We found no effects of IR on Aβ₄₂ levels in CSF. CONCLUSION IR among asymptomatic APOEɛ4 carriers was associated with higher P-Tau181 and T-Tau in late-middle age. The results suggest that IR may contribute to tau-related neurodegeneration in preclinical AD. The findings may have implications for developing prevention strategies aimed at modifying IR in mid-life.
Collapse
Affiliation(s)
- Erika J Starks
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - J Patrick O'Grady
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Siobhan M Hoscheidt
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Annie M Racine
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Cynthia M Carlsson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research Education and Clinical Center, Wm. S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,UCL Institute of Neurology, Queen Square, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,The Torsten Söderberg Professorship at the Royal Swedish Academy of Sciences, Sweden
| | - Ozioma C Okonkwo
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Luigi Puglielli
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research Education and Clinical Center, Wm. S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research Education and Clinical Center, Wm. S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - N Maritza Dowling
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Carey E Gleason
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research Education and Clinical Center, Wm. S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Rozalyn M Anderson
- Geriatric Research Education and Clinical Center, Wm. S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Nancy J Davenport-Sis
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - LeAnn M DeRungs
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Mark A Sager
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research Education and Clinical Center, Wm. S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
40
|
Alberdi A, Aztiria A, Basarab A. On the early diagnosis of Alzheimer's Disease from multimodal signals: A survey. Artif Intell Med 2016; 71:1-29. [PMID: 27506128 DOI: 10.1016/j.artmed.2016.06.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/23/2016] [Accepted: 06/07/2016] [Indexed: 11/15/2022]
Abstract
INTRODUCTION The number of Alzheimer's Disease (AD) patients is increasing with increased life expectancy and 115.4 million people are expected to be affected in 2050. Unfortunately, AD is commonly diagnosed too late, when irreversible damages have been caused in the patient. OBJECTIVE An automatic, continuous and unobtrusive early AD detection method would be required to improve patients' life quality and avoid big healthcare costs. Thus, the objective of this survey is to review the multimodal signals that could be used in the development of such a system, emphasizing on the accuracy that they have shown up to date for AD detection. Some useful tools and specific issues towards this goal will also have to be reviewed. METHODS An extensive literature review was performed following a specific search strategy, inclusion criteria, data extraction and quality assessment in the Inspec, Compendex and PubMed databases. RESULTS This work reviews the extensive list of psychological, physiological, behavioural and cognitive measurements that could be used for AD detection. The most promising measurements seem to be magnetic resonance imaging (MRI) for AD vs control (CTL) discrimination with an 98.95% accuracy, while electroencephalogram (EEG) shows the best results for mild cognitive impairment (MCI) vs CTL (97.88%) and MCI vs AD distinction (94.05%). Available physiological and behavioural AD datasets are listed, as well as medical imaging analysis steps and neuroimaging processing toolboxes. Some issues such as "label noise" and multi-site data are discussed. CONCLUSIONS The development of an unobtrusive and transparent AD detection system should be based on a multimodal system in order to take full advantage of all kinds of symptoms, detect even the smallest changes and combine them, so as to detect AD as early as possible. Such a multimodal system might probably be based on physiological monitoring of MRI or EEG, as well as behavioural measurements like the ones proposed along the article. The mentioned AD datasets and image processing toolboxes are available for their use towards this goal. Issues like "label noise" and multi-site neuroimaging incompatibilities may also have to be overcome, but methods for this purpose are already available.
Collapse
Affiliation(s)
- Ane Alberdi
- Mondragon University, Electronics and Computing Department, Goiru Kalea, 2, Arrasate 20500, Spain.
| | - Asier Aztiria
- Mondragon University, Electronics and Computing Department, Goiru Kalea, 2, Arrasate 20500, Spain.
| | - Adrian Basarab
- Université de Toulouse, Institut de Recherche en Informatique de Toulouse, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5505, Université Paul Sabatier, 118 Route de Narbonne, 31062 Toulouse, France.
| |
Collapse
|
41
|
Wang T, Shi F, Jin Y, Jiang W, Shen D, Xiao S. Abnormal Changes of Brain Cortical Anatomy and the Association with Plasma MicroRNA107 Level in Amnestic Mild Cognitive Impairment. Front Aging Neurosci 2016; 8:112. [PMID: 27242521 PMCID: PMC4870937 DOI: 10.3389/fnagi.2016.00112] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 04/29/2016] [Indexed: 11/17/2022] Open
Abstract
UNLABELLED MicroRNA107 (Mir107) has been thought to relate to the brain structure phenotype of Alzheimer's disease. In this study, we evaluated the cortical anatomy in amnestic mild cognitive impairment (aMCI) and the relation between cortical anatomy and plasma levels of Mir107 and beta-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1). Twenty aMCI (20 aMCI) and 24 cognitively normal control (NC) subjects were recruited, and T1-weighted MR images were acquired. Cortical anatomical measurements, including cortical thickness (CT), surface area (SA), and local gyrification index (LGI), were assessed. Quantitative RT-PCR was used to examine plasma expression of Mir107, BACE1 mRNA. Thinner cortex was found in aMCI in areas associated with episodic memory and language, but with thicker cortex in other areas. SA decreased in aMCI in the areas associated with working memory and emotion. LGI showed a significant reduction in aMCI in the areas involved in language function. Changes in Mir107 and BACE1 messenger RNA plasma expression were correlated with changes in CT and SA. We found alterations in key left brain regions associated with memory, language, and emotion in aMCI that were significantly correlated with plasma expression of Mir107 and BACE1 mRNA. This combination study of brain anatomical alterations and gene information may shed lights on our understanding of the pathology of AD. CLINICAL TRIAL REGISTRATION http://www.ClinicalTrials.gov, identifier NCT01819545.
Collapse
Affiliation(s)
- Tao Wang
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of MedicineShanghai, China
- Alzheimer’s Disease and Related Disorders Center, Shanghai Jiao Tong UniversityShanghai, China
- IDEA Lab, Department of Radiology and BRIC, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Feng Shi
- IDEA Lab, Department of Radiology and BRIC, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Yan Jin
- IDEA Lab, Department of Radiology and BRIC, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Weixiong Jiang
- Alzheimer’s Disease and Related Disorders Center, Shanghai Jiao Tong UniversityShanghai, China
| | - Dinggang Shen
- IDEA Lab, Department of Radiology and BRIC, University of North Carolina at Chapel HillChapel Hill, NC, USA
- Department of Brain and Cognitive Engineering, Korea UniversitySeoul, South Korea
| | - Shifu Xiao
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of MedicineShanghai, China
- Alzheimer’s Disease and Related Disorders Center, Shanghai Jiao Tong UniversityShanghai, China
| |
Collapse
|
42
|
Liu S, Cai W, Pujol S, Kikinis R, Feng DD. Cross-View Neuroimage Pattern Analysis in Alzheimer's Disease Staging. Front Aging Neurosci 2016; 8:23. [PMID: 26941639 PMCID: PMC4763344 DOI: 10.3389/fnagi.2016.00023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 02/01/2016] [Indexed: 01/24/2023] Open
Abstract
The research on staging of pre-symptomatic and prodromal phase of neurological disorders, e.g., Alzheimer's disease (AD), is essential for prevention of dementia. New strategies for AD staging with a focus on early detection, are demanded to optimize potential efficacy of disease-modifying therapies that can halt or slow the disease progression. Recently, neuroimaging are increasingly used as additional research-based markers to detect AD onset and predict conversion of MCI and normal control (NC) to AD. Researchers have proposed a variety of neuroimaging biomarkers to characterize the patterns of the pathology of AD and MCI, and suggested that multi-view neuroimaging biomarkers could lead to better performance than single-view biomarkers in AD staging. However, it is still unclear what leads to such synergy and how to preserve or maximize. In an attempt to answer these questions, we proposed a cross-view pattern analysis framework for investigating the synergy between different neuroimaging biomarkers. We quantitatively analyzed nine types of biomarkers derived from FDG-PET and T1-MRI, and evaluated their performance in a task of classifying AD, MCI, and NC subjects obtained from the ADNI baseline cohort. The experiment results showed that these biomarkers could depict the pathology of AD from different perspectives, and output distinct patterns that are significantly associated with the disease progression. Most importantly, we found that these features could be separated into clusters, each depicting a particular aspect; and the inter-cluster features could always achieve better performance than the intra-cluster features in AD staging.
Collapse
Affiliation(s)
- Sidong Liu
- The Biomedical and Multimedia Information Technology Research Group, School of Information Technologies, The University of SydneySydney, NSW, Australia
| | - Weidong Cai
- The Biomedical and Multimedia Information Technology Research Group, School of Information Technologies, The University of SydneySydney, NSW, Australia
| | - Sonia Pujol
- The Surgical Planning Laboratory, Harvard Medical School, Brigham and Women's HospitalBoston, MA, USA
| | - Ron Kikinis
- The Surgical Planning Laboratory, Harvard Medical School, Brigham and Women's HospitalBoston, MA, USA
| | - Dagan D. Feng
- The Biomedical and Multimedia Information Technology Research Group, School of Information Technologies, The University of SydneySydney, NSW, Australia
- The Med-X Research Institute, Shanghai Jiao Tong UniversityShanghai, China
| | | |
Collapse
|
43
|
Jiang Y, Huang H, Abner E, Broster LS, Jicha GA, Schmitt FA, Kryscio R, Andersen A, Powell D, Van Eldik L, Gold BT, Nelson PT, Smith C, Ding M. Alzheimer's Biomarkers are Correlated with Brain Connectivity in Older Adults Differentially during Resting and Task States. Front Aging Neurosci 2016; 8:15. [PMID: 26903858 PMCID: PMC4744860 DOI: 10.3389/fnagi.2016.00015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 01/19/2016] [Indexed: 12/18/2022] Open
Abstract
β-amyloid (Aβ) plaques and tau-related neurodegeneration are pathologic hallmarks of Alzheimer’s disease (AD). The utility of AD biomarkers, including those measured in cerebrospinal fluid (CSF), in predicting future AD risk and cognitive decline is still being refined. Here, we explored potential relationships between functional connectivity (FC) patterns within the default-mode network (DMN), age, CSF biomarkers (Aβ42 and pTau181), and cognitive status in older adults. Multiple measures of FC were explored, including a novel time series-based measure [total interdependence (TI)]. In our sample of 27 cognitively normal older adults, no significant associations were found between levels of Aβ42 or pTau181 and cognitive scores or regional brain volumes. However, we observed several novel relationships between these biomarkers and measures of FC in DMN during both resting-state and a short-term memory task. First, increased connectivity between bilateral anterior middle temporal gyri was associated with higher levels of CSF Aβ42 and Aβ42/pTau181 ratio (reflecting lower AD risk) during both rest and task. Second, increased bilateral parietal connectivity during the short-term memory task, but not during rest, was associated with higher levels of CSF pTau181 (reflecting higher AD risk). Third, increased connectivity between left middle temporal and left parietal cortices during the active task was associated with decreased global cognitive status but not CSF biomarkers. Lastly, we found that our new TI method was more sensitive to the CSF Aβ42-connectivity relationship whereas the traditional cross-correlation method was more sensitive to levels of CSF pTau181 and cognitive status. With further refinement, resting-state connectivity and task-driven connectivity measures hold promise as non-invasive neuroimaging markers of Aβ and pTau burden in cognitively normal older adults.
Collapse
Affiliation(s)
- Yang Jiang
- Department of Behavioral Science, University of Kentucky College of Medicine, Lexington, KY, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; The Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky, Lexington, KY, USA
| | - Haiqing Huang
- J Crayton Pruitt Family Department of Biomedical Engineering, University of Florida , Gainesville, FL , USA
| | - Erin Abner
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; Department of Epidemiology, University of Kentucky College of Public Health, Lexington, KY, USA
| | - Lucas S Broster
- Department of Behavioral Science, University of Kentucky College of Medicine , Lexington, KY , USA
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; Department of Neurology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Frederick A Schmitt
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; Department of Neurology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Richard Kryscio
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; Department of Biostatistics, University of Kentucky College of Public Health, Lexington, KY, USA
| | - Anders Andersen
- The Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky, Lexington, KY, USA; Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - David Powell
- The Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky, Lexington, KY, USA; Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Linda Van Eldik
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Brian T Gold
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; The Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky, Lexington, KY, USA; Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky , Lexington, KY , USA
| | - Charles Smith
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; The Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky, Lexington, KY, USA; Department of Neurology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Mingzhou Ding
- J Crayton Pruitt Family Department of Biomedical Engineering, University of Florida , Gainesville, FL , USA
| |
Collapse
|
44
|
Proteome-wide characterization of signalling interactions in the hippocampal CA4/DG subfield of patients with Alzheimer's disease. Sci Rep 2015; 5:11138. [PMID: 26059363 PMCID: PMC4462342 DOI: 10.1038/srep11138] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 04/27/2015] [Indexed: 12/02/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia; however, mechanisms and biomarkers remain unclear. Here, we examined hippocampal CA4 and dentate gyrus subfields, which are less studied in the context of AD pathology, in post-mortem AD and control tissue to identify possible biomarkers. We performed mass spectrometry-based proteomic analysis combined with label-free quantification for identification of differentially expressed proteins. We identified 4,328 proteins, of which 113 showed more than 2-fold higher or lower expression in AD hippocampi than in control tissues. Five proteins were identified as putative AD biomarkers (MDH2, PCLO, TRRAP, YWHAZ, and MUC19 isoform 5) and were cross-validated by immunoblotting, selected reaction monitoring, and MALDI imaging. We also used a bioinformatics approach to examine upstream signalling interactions of the 113 regulated proteins. Five upstream signalling (IGF1, BDNF, ZAP70, MYC, and cyclosporin A) factors showed novel interactions in AD hippocampi. Taken together, these results demonstrate a novel platform that may provide new strategies for the early detection of AD and thus its diagnosis.
Collapse
|
45
|
Huded CB, Bharath S, Chandra SR, Sivakumar PT, Varghese M, Subramanian S. Supportive CSF biomarker evidence to enhance the National Institute on Aging-Alzheimer's Association criteria for diagnosis of Alzheimer's type dementia--a study from Southern India. Asian J Psychiatr 2015; 13:44-7. [PMID: 25468261 DOI: 10.1016/j.ajp.2014.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/28/2014] [Accepted: 10/29/2014] [Indexed: 01/03/2023]
Abstract
The present study was undertaken to validate the measurement of biomarkers as a supplement to the latest diagnostic criteria for Alzheimer disease (AD) dementia by National Institute on Aging-Alzheimer's Association (NIA-AA) work group using a sample attending a tertiary care center in Southern India. A total of 20 subjects diagnosed clinically as Alzheimer's dementia according to the NIA-AA criteria for AD were included in the study. The CSF biomarkers Aβ42, t-tau, and p-tau181 were assessed. The biomarker results were compared among mild and moderate to severe AD as defined in the NIA-AA work group guidelines. The results revealed that the amount of Aβ42 was very low in all the 20 samples (<50pg/ml) collected from mild AD cases with CDR score of 1 (n=8), and moderate to severe AD cases with CDR >1 (n=12). t-tau and p-tau levels were in the range of 39.45±5.09pg/ml and 13.06±7.32pg/ml for CDR 1 group. t-tau and p-tau levels were in the range of 49.9±11.28pg/ml and 33.94±15.13pg/ml for moderate to severe cases. Analysis of the data revealed statistically significant differences in the p-tau/t-tau ratio and p-tau/Aβ ratio between CDR 1and CDR >1 AD cases (p<0.001) suggesting that p-tau/t-tau and p-tau/Aβ ratio are good indicators of severity of dementia with discriminative value in differentiating mild AD from moderate to severe AD.
Collapse
Affiliation(s)
- Chandrashekar B Huded
- Department of Psychiatry, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - Srikala Bharath
- Department of Psychiatry, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - S R Chandra
- Department of Neurology, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - P T Sivakumar
- Department of Psychiatry, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - Mathew Varghese
- Department of Psychiatry, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - Sarada Subramanian
- Department of Neurochemistry, National Institute of Mental Health & Neurosciences, Bangalore, India.
| |
Collapse
|