1
|
Panmanee J, Phanchana M, Pearngam P, Petchyam N, Promthep K, Wisomka P, Kutpruek S, Pannengpetch S, Prasertporn T, Mukda S, Govitrapong P, Nopparat C. A Proteomics Profiling Reveals the Neuroprotective Effects of Melatonin on Exogenous β-amyloid-42 Induced Mitochondrial Impairment, Intracellular β-amyloid Accumulation and Tau Hyperphosphorylation in Human SH-SY5Y Cells. Cell Biol Int 2025; 49:659-673. [PMID: 40047119 PMCID: PMC12070025 DOI: 10.1002/cbin.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 11/19/2024] [Accepted: 02/21/2025] [Indexed: 05/14/2025]
Abstract
Alzheimer's disease (AD) is prevalent in the elderly population and characterized by the intracellular accumulation of neurofibrillary tangles (NFTs), composed of tau proteins, and extracellular deposition of beta-amyloid protein (Aβ). The present study aimed to investigate the neuroprotective effects of melatonin on Aβ42-induced AD-like pathology in SH-SY5Y cell lines. To assess the effects of melatonin on Aβ42-exposed cells, we performed a proteomics analysis of altered protein expression in Aβ42-treated cells, with or without melatonin Pretreatment, using label-free nano-LC-MS/MS. Experimental validations of pathways related to the neuroprotective effects of melatonin were carried out using Milliplex amyloid beta and tau magnetic bead assays, Western blot analysis, and measurements of mitochondrial membrane potential and ROS levels. Our results show that Aβ42 exposure led to an increase in an accumulation of intracellular Aβ42/40 and phosphorylated tau (Thr181)/Tau ratios. Pretreatment with melatonin effectively reduced the levels of these pathogenic proteins. Proteomics analysis has revealed protein markers associated with the Alzheimer's disease pathway, neuronal synapses, cellular apoptosis, and mitochondrial functions. Changes in proteins regulating the mitochondrial permeability transition pore, the electron transport chain, and mitochondrial oxidative stress were observed in Aβ42-treated cells. Pretreatment with melatonin protected the cells against Aβ42-induced cellular damages by regulating the expression of several proteins underpinning these biological processes, including the suppression of mitochondrial ROS generation and mitigation of mitochondrial membrane depolarization.
Collapse
Affiliation(s)
- Jiraporn Panmanee
- Research Center for Neuroscience, Institute of Molecular BiosciencesMahidol UniversityNakhon PathomThailand
| | - Matthew Phanchana
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical MedicineMahidol UniversityBangkokThailand
| | - Phorutai Pearngam
- Biological Sciences ProgramMahidol University International College, Mahidol UniversityNakhon PathomThailand
| | - Nopphon Petchyam
- Center for Advanced Therapeutics, Institute of Molecular BiosciencesMahidol UniversityNakhon PathomThailand
| | - Kornkanok Promthep
- Research Center for Neuroscience, Institute of Molecular BiosciencesMahidol UniversityNakhon PathomThailand
| | - Ponlawit Wisomka
- Research Center for Neuroscience, Institute of Molecular BiosciencesMahidol UniversityNakhon PathomThailand
| | - Suchanoot Kutpruek
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, LaksiBangkokThailand
| | - Supitcha Pannengpetch
- Center for Research and Innovation, Faculty of Medical TechnologyMahidol UniversityNakhon PathomThailand
| | - Tanya Prasertporn
- Research Center for Neuroscience, Institute of Molecular BiosciencesMahidol UniversityNakhon PathomThailand
| | - Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular BiosciencesMahidol UniversityNakhon PathomThailand
| | - Piyarat Govitrapong
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, LaksiBangkokThailand
| | | |
Collapse
|
2
|
Matlyuba Jakhonkulovna S, Bahodirova Kamolovna G, Zokirov M, Umida Tajimuratovna B, Yumashev A, Shichiyakh R, Safarova NI, Nargiza Nusratovna A, Esanmuradova N, Muyassar Karimbaevna T, Lazizakhon A, Ishankulov A. Electrochemical biosensors for early detection of Alzheimer's disease. Clin Chim Acta 2025; 572:120278. [PMID: 40185381 DOI: 10.1016/j.cca.2025.120278] [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: 03/15/2025] [Revised: 03/29/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
In recent years, electrochemical biosensors have shown great promise as innovative tools for the early identification of Alzheimer's disease (AD), a neurodegenerative disorder that severely affects cognitive ability and overall quality of life. This comprehensive review aims to consolidate the latest research on the creation and implementation of electrochemical biosensors designed to detect AD-related biomarkers. We examine cutting-edge approaches to surface modification that enhance the attachment of biorecognition molecules, thus enabling the simultaneous identification of multiple biomarkers. This review emphasizes the crucial role that electrochemical biosensors play in the early diagnosis of Alzheimer's disease, highlighting their potential to revolutionize clinical practices by facilitating timely interventions. In the future, research efforts should concentrate on refining these technologies for widespread clinical adoption, ensuring that they meet the needs of both healthcare professionals and patients.
Collapse
Affiliation(s)
| | - Gulnoz Bahodirova Kamolovna
- Department of Scientific Research, Innovations and Scientific and Pedagogical Personnel Training International School of Finance Technology and Science (ISFT Institute), Uzbekistan
| | | | | | - Alexey Yumashev
- Department of Prosthetic Dentistry, Doctor of Medicine, Professor Sechenov First Moscow State Medical University, Russia
| | - Rustem Shichiyakh
- Department of Management, Candidate of Economic Sciences, Associate Professor. Kuban State Agrarian University named after I.T. Trubilin, Krasnodar, Russia
| | - Nasiba I Safarova
- Department of Otorhinolaryngology, Faculty of Postgraduate Education, Samarkand State Medical University, Samarkand, Uzbekistan
| | | | - Nilufar Esanmuradova
- "Tashkent Institute of Irrigation and Agricultural Mechanization Engineers" National Research University, Tashkent, Uzbekistan; Western Caspian University, Scientific Researcher, Baku, Azerbaijan
| | - Tadjibaeva Muyassar Karimbaevna
- Department of Zoology, Human Morphophysiology and Nutrition (PhD), Nukus State Pedagogical Institute Named After Ajiniyaz, Uzbekistan
| | - Alidjanova Lazizakhon
- International Islamic Academy of Uzbekistan, Senior Lecturer of "UNESCO Chair on Religious Studies and the Comparative Study of World Religions", Kadiri st. 11, Tashkent, Uzbekistan
| | - Alisher Ishankulov
- Samarkand State University named after Sharof Rashidov, Uzbekistan; Kimyo International University in Tashkent, Branch Samarkand, Uzbekistan
| |
Collapse
|
3
|
Dhana A, DeCarli CS, Dhana K, Desai P, Krueger K, Evans DA, Rajan KB. Association of subjective memory complaints with serum biomarkers of neurodegeneration and cognition: A population-based study. J Am Geriatr Soc 2025; 73:859-866. [PMID: 39625703 PMCID: PMC11908938 DOI: 10.1111/jgs.19300] [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: 05/21/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 12/08/2024]
Abstract
BACKGROUND Subjective memory complaints (SMCs) refer to memory concerns reported by an individual, regardless of objective test impairment. We conducted a study to evaluate the association of SMCs with serum biomarkers of neurodegeneration, including neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), and total tau (t-tau). In addition, we evaluated the association of SMCs with the rate of cognitive decline. METHODS This study included 1096 individuals participating in the Chicago Health and Aging Project, with data on SMCs, neurodegenerative biomarkers (NfL, GFAP, and t-tau), and global cognitive scores. The SMC score ranges from 0 to 8 and higher scores reflect more memory complaints. Linear regression models were developed to investigate the association of SMCs with serum biomarkers, adjusted by age, sex, race, education, and APOE e4. Linear mixed-effects models were used to examine the independent association of SMCs with cognitive decline in a multivariable model that was additionally adjusted by biomarkers of neurodegeneration. RESULTS Of the 1096 individuals, 665 (60.7%) were female, 643 (58.7%) were African Americans, and the mean (SD) age at the baseline was 78.0 (5.8) years. Compared to individuals with fewer memory complaints (i.e., people in the first tertile of SMCs), those reporting more memory complaints (i.e., people in the third tertile of SMCs) had a 12.0% increase in serum concentrations of NfL and an 9.4% increase in GFAP. In addition, individuals reporting more memory complaints (i.e., those in the 3rd versus the 1st tertile of SMCs) had a faster rate of cognitive decline by 0.029 ( β -0.029, 95% CI -0.046 to -0.013) standardized units per year. CONCLUSIONS Individuals who reported more memory complaints had higher levels of biomarkers of neurodegeneration and a faster rate of cognitive decline, suggesting that SMCs may be valuable for identifying people at high risk of cognitive impairment.
Collapse
Affiliation(s)
- Anisa Dhana
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL
| | - Charles S. DeCarli
- Department of Neurology, University of California at Davis, Sacramento, CA, USA
| | - Klodian Dhana
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL
| | - Pankaja Desai
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL
| | - Kristin Krueger
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL
| | - Denis A. Evans
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL
| | - Kumar B. Rajan
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL
- Department of Neurology, University of California at Davis, Sacramento, CA, USA
| |
Collapse
|
4
|
Song J, Cho E, Lee H, Lee S, Kim S, Kim J. Development of Neurodegenerative Disease Diagnosis and Monitoring from Traditional to Digital Biomarkers. BIOSENSORS 2025; 15:102. [PMID: 39997004 PMCID: PMC11852611 DOI: 10.3390/bios15020102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/03/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
Monitoring and assessing the progression of symptoms in neurodegenerative diseases, including Alzheimer's and Parkinson's disease, are critical for improving patient outcomes. Traditional biomarkers, such as cerebrospinal fluid analysis and brain imaging, are widely used to investigate the underlying mechanisms of disease and enable early diagnosis. In contrast, digital biomarkers derived from phenotypic changes-such as EEG, eye movement, gait, and speech analysis-offer a noninvasive and accessible alternative. Leveraging portable and widely available devices, such as smartphones and wearable sensors, digital biomarkers are emerging as a promising tool for ND diagnosis and monitoring. This review highlights the comprehensive developments in digital biomarkers, emphasizing their unique advantages and integration potential alongside traditional biomarkers.
Collapse
Affiliation(s)
| | | | | | | | | | - Jinsik Kim
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul 04620, Republic of Korea; (J.S.); (E.C.); (H.L.); (S.L.); (S.K.)
| |
Collapse
|
5
|
Barker RM, Chambers A, Kehoe PG, Rowe E, Perks CM. Untangling the role of tau in sex hormone responsive cancers: lessons learnt from Alzheimer's disease. Clin Sci (Lond) 2024; 138:1357-1369. [PMID: 39469929 PMCID: PMC11522895 DOI: 10.1042/cs20230317] [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: 05/22/2024] [Revised: 09/20/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Tubulin associated unit has been extensively studied in neurodegenerative diseases including Alzheimer's disease (AD), whereby its hyperphosphorylation and accumulation contributes to disease pathogenesis. Tau is abundantly expressed in the central nervous system but is also present in non-neuronal tissues and in tumours including sex hormone responsive cancers such as breast and prostate. Curiously, hormonal effects on tau also exist in an AD context from numerous studies on menopause, hormone replacement therapy, and androgen deprivation therapy. Despite sharing some risk factors, most importantly advancing age, there are numerous reports from population studies of, currently poorly explained inverse associations between cancer and Alzheimer's disease. We previously reviewed important components of the phosphoinositide-3-kinase/protein kinase B (PI3K/Akt) signalling pathway and their differential modulation in relation to the two diseases. Similarly, receptor tyrosine kinases, estrogen receptor and androgen receptor have all been implicated in the pathogenesis of both cancer and AD. In this review, we focus on tau and its effects in hormone responsive cancer in terms of development, progression, and treatment and in relation to sex hormones and PI3K/Akt signalling molecules including IRS-1, PTEN, Pin1, and p53.
Collapse
Affiliation(s)
- Rachel M. Barker
- Cancer Endocrinology Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| | - Alfie Chambers
- Cancer Endocrinology Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| | - Patrick G. Kehoe
- Department of Urology, Bristol Urological Institute, Southmead Hospital, Bristol BS10 5NB, UK
| | - Edward Rowe
- Dementia Research Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| | - Claire M. Perks
- Cancer Endocrinology Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| |
Collapse
|
6
|
Kamatham PT, Shukla R, Khatri DK, Vora LK. Pathogenesis, diagnostics, and therapeutics for Alzheimer's disease: Breaking the memory barrier. Ageing Res Rev 2024; 101:102481. [PMID: 39236855 DOI: 10.1016/j.arr.2024.102481] [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: 05/06/2024] [Revised: 06/28/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and accounts for 60-70 % of all cases. It affects millions of people worldwide. AD poses a substantial economic burden on societies and healthcare systems. AD is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired daily functioning. As the prevalence of AD continues to increase, understanding its pathogenesis, improving diagnostic methods, and developing effective therapeutics have become paramount. This comprehensive review delves into the intricate mechanisms underlying AD, explores the current state of diagnostic techniques, and examines emerging therapeutic strategies. By revealing the complexities of AD, this review aims to contribute to the growing body of knowledge surrounding this devastating disease.
Collapse
Affiliation(s)
- Pushpa Tryphena Kamatham
- Molecular and Cellular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Rashi Shukla
- Molecular and Cellular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology, Nims Institute of Pharmacy, Nims University Rajasthan, Jaipur, India.
| | - Lalitkumar K Vora
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland BT9 7BL, UK.
| |
Collapse
|
7
|
Friberg S, Lindblad C, Zeiler FA, Zetterberg H, Granberg T, Svenningsson P, Piehl F, Thelin EP. Fluid biomarkers of chronic traumatic brain injury. Nat Rev Neurol 2024; 20:671-684. [PMID: 39363129 DOI: 10.1038/s41582-024-01024-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2024] [Indexed: 10/05/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability across the world. Evidence for the usefulness of imaging and fluid biomarkers to predict outcomes and screen for the need to monitor complications in the acute stage is steadily increasing. Still, many people experience symptoms such as fatigue and cognitive and motor dysfunction in the chronic phase of TBI, where objective assessments for brain injury are lacking. Consensus criteria for traumatic encephalopathy syndrome, a clinical syndrome possibly associated with the neurodegenerative disease chronic traumatic encephalopathy, which is commonly associated with sports concussion, have been defined only recently. However, these criteria do not fit all individuals living with chronic consequences of TBI. The pathophysiology of chronic TBI shares many similarities with other neurodegenerative and neuroinflammatory conditions, such as Alzheimer disease. As with Alzheimer disease, advancements in fluid biomarkers represent one of the most promising paths for unravelling the chain of pathophysiological events to enable discrimination between these conditions and, with time, provide prediction modelling and therapeutic end points. This Review summarizes fluid biomarker findings in the chronic phase of TBI (≥6 months after injury) that demonstrate the involvement of inflammation, glial biology and neurodegeneration in the long-term complications of TBI. We explore how the biomarkers associate with outcome and imaging findings and aim to establish mechanistic differences in biomarker patterns between types of chronic TBI and other neurodegenerative conditions. Finally, current limitations and areas of priority for future fluid biomarker research are highlighted.
Collapse
Affiliation(s)
- Susanna Friberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Lindblad
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Neurosurgery, Uppsala University Hospital, Uppsala, Sweden
| | - Frederick A Zeiler
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Section of Neurosurgery, Department of Surgery, University of Manitoba, Rady Faculty of Health Sciences, Winnipeg, Manitoba, Canada
- Department of Biomedical Engineering, Price Faculty of Engineering, University of Manitoba, Winnipeg, Manitoba, Canada
- Pan Am Clinic Foundation, Winnipeg, Manitoba, Canada
- Division of Anaesthesia, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Henrik Zetterberg
- UK Dementia Research Institute, University College London, London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Tobias Granberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Department of Basic and Clinical Neuroscience, King's College London, London, UK
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Eric P Thelin
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
8
|
Cayir S, Volpi T, Toyonaga T, Gallezot JD, Yang Y, Sadabad FE, Mulnix T, Mecca AP, Fesharaki-Zadeh A, Matuskey D. Relationship between neuroimaging and cognition in frontotemporal dementia: An FDG-PET and structural MRI study. J Neuroimaging 2024; 34:627-634. [PMID: 38676301 PMCID: PMC11511789 DOI: 10.1111/jon.13206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/14/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND AND PURPOSE Frontotemporal dementia (FTD) is a clinically and pathologically heterogeneous neurodegenerative condition with a prevalence comparable to Alzheimer's disease for patients under 65 years of age. Limited studies have examined the association between cognition and neuroimaging in FTD using different imaging modalities. METHODS We examined the association of cognition using Montreal Cognitive Assessment (MoCA) with both gray matter (GM) volume and glucose metabolism using magnetic resonance imaging and fluorodeoxyglucose (FDG)-PET in 21 patients diagnosed with FTD. Standardized uptake value ratio (SUVR) using the brainstem as a reference region was the primary outcome measure for FDG-PET. Partial volume correction was applied to PET data to account for disease-related atrophy. RESULTS Significant positive associations were found between whole-cortex GM volume and MoCA scores (r = 0.46, p = .04). The association between whole-cortex FDG SUVR and MoCA scores was not significant (r = 0.37, p = .09). GM volumes of the frontal cortex (r = 0.54, p = .01), caudate (r = 0.62, p<.01), and insula (r = 0.57, p<.01) were also significantly correlated with MoCA, as were SUVR values of the insula (r = 0.51, p = .02), thalamus (r = 0.48, p = .03), and posterior cingulate cortex (PCC) (r = 0.47, p = .03). CONCLUSIONS Whole-cortex atrophy is associated with cognitive dysfunction, and this association is larger than for whole-cortex hypometabolism as measured with FDG-PET. At the regional level, focal atrophy and/or hypometabolism in the frontal cortex, insula, PCC, thalamus, and caudate seem to be important for the decline of cognitive function in FTD. Furthermore, these results highlight how functional and structural changes may not overlap and might contribute to cognitive dysfunction in FTD in different ways.
Collapse
Affiliation(s)
- Salih Cayir
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tommaso Volpi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jean-Dominique Gallezot
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yanghong Yang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Faranak Ebrahimian Sadabad
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tim Mulnix
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Adam P Mecca
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Arman Fesharaki-Zadeh
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - David Matuskey
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
9
|
Ahmad S, Yang W, Orellana A, Frölich L, de Rojas I, Cano A, Boada M, Hernández I, Hausner L, Harms AC, Bakker MHM, Cabrera-Socorro A, Amin N, Ramírez A, Ruiz A, Van Duijn CM, Hankemeier T. Association of oxidative stress and inflammatory metabolites with Alzheimer's disease cerebrospinal fluid biomarkers in mild cognitive impairment. Alzheimers Res Ther 2024; 16:171. [PMID: 39080778 PMCID: PMC11287840 DOI: 10.1186/s13195-024-01542-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024]
Abstract
BACKGROUND Isoprostanes and prostaglandins are biomarkers for oxidative stress and inflammation. Their role in Alzheimer's disease (AD) pathophysiology is yet unknown. In the current study, we aim to identify the association of isoprostanes and prostaglandins with the Amyloid, Tau, Neurodegeneration (ATN) biomarkers (Aβ-42, p-tau, and t-tau) of AD pathophysiology in mild cognitive impairment (MCI) subjects. METHODS Targeted metabolomics profiling was performed using liquid chromatography-mass spectrometry (LCMS) in 147 paired plasma-CSF samples from the Ace Alzheimer Center Barcelona and 58 CSF samples of MCI patients from the Mannheim/Heidelberg cohort. Linear regression was used to evaluate the association of metabolites with CSF levels of ATN biomarkers in the overall sample and stratified by Aβ-42 pathology and APOE genotype. We further evaluated the role of metabolites in MCI to AD dementia progression. RESULTS Increased CSF levels of PGF2α, 8,12-iso-iPF2α VI, and 5-iPF2α VI were significantly associated (False discovery rate (FDR) < 0.05) with higher p-tau levels. Additionally, 8,12-iso-iPF2α VI was associated with increased total tau levels in CSF. In MCI due to AD, PGF2α was associated with both p-tau and total tau, whereases 8,12-iso-iPF2α VI was specifically associated with p-tau levels. In APOE stratified analysis, association of PGF2α with p-tau and t-tau was observed in only APOE ε4 carriers while 5-iPF2α VI showed association with both p-tau and t-tau in APOE ε33 carriers. CSF levels of 8,12- iso-iPF2α VI showed association with p-tau and t-tau in APOE ε33/APOE ε4 carriers and with t-tau in APOE ε3 carriers. None of the metabolites showed evidence of association with MCI to AD progression. CONCLUSIONS Oxidative stress (8,12-iso-iPF2α VI) and inflammatory (PGF2α) biomarkers are correlated with biomarkers of AD pathology during the prodromal stage of AD and relation of PGF2α with tau pathology markers may be influenced by APOE genotype.
Collapse
Affiliation(s)
- Shahzad Ahmad
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
- Oxford-GSK Institute of Molecular and Computational Medicine (IMCM), Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Wei Yang
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Adelina Orellana
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Amanda Cano
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Hernández
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Lucrezia Hausner
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany
| | - Amy C Harms
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Margot H M Bakker
- Discovery Research, AbbVie Deutschland GmbH & Co. KG, 67061, KnollstrasseLudwigshafen, Germany
| | | | - Najaf Amin
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands
- Nuffield Department of Population Health, University of Oxford, Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Old Road Campus, , Headington-Oxford, OX3 7FZ, UK
| | - Alfredo Ramírez
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Excellence Cluster On Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931, Cologne, Germany
- Department of Psychiatry and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, USA
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Cornelia M Van Duijn
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands.
- Nuffield Department of Population Health, University of Oxford, Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Old Road Campus, , Headington-Oxford, OX3 7FZ, UK.
| | - Thomas Hankemeier
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands.
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
10
|
Bhatt V, Shukla H, Tiwari AK. Parkinson's Disease and Mitotherapy-Based Approaches towards α-Synucleinopathies. J Integr Neurosci 2024; 23:109. [PMID: 38940084 DOI: 10.31083/j.jin2306109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/25/2024] [Accepted: 02/05/2024] [Indexed: 06/29/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta region of the midbrain and the formation of intracellular protein aggregates known as Lewy bodies, of which a major component is the protein α-synuclein. Several studies have suggested that mitochondria play a central role in the pathogenesis of PD, encompassing both familial and sporadic forms of the disease. Mitochondrial dysfunction is attributed to bioenergetic impairment, increased oxidative stress, damage to mitochondrial DNA, and alteration in mitochondrial morphology. These alterations may contribute to improper functioning of the central nervous system and ultimately lead to neurodegeneration. The perturbation of mitochondrial function makes it a potential target, worthy of exploration for neuroprotective therapies and to improve mitochondrial health in PD. Thus, in the current review, we provide an update on mitochondria-based therapeutic approaches toward α-synucleinopathies in PD.
Collapse
Affiliation(s)
- Vidhi Bhatt
- Genetics & Developmental Biology Laboratory, Department of Biotechnology & Bioengineering, Institute of Advanced Research (IAR), 382426 Gandhinagar, Gujarat, India
| | - Halak Shukla
- Genetics & Developmental Biology Laboratory, Department of Biotechnology & Bioengineering, Institute of Advanced Research (IAR), 382426 Gandhinagar, Gujarat, India
| | - Anand Krishna Tiwari
- Genetics & Developmental Biology Laboratory, Department of Biotechnology & Bioengineering, Institute of Advanced Research (IAR), 382426 Gandhinagar, Gujarat, India
| |
Collapse
|
11
|
Andrés-Benito P, Vázquez-Costa JF, Ñungo Garzón NC, Colomina MJ, Marco C, González L, Terrafeta C, Domínguez R, Ferrer I, Povedano M. Neurodegeneration Biomarkers in Adult Spinal Muscular Atrophy (SMA) Patients Treated with Nusinersen. Int J Mol Sci 2024; 25:3810. [PMID: 38612621 PMCID: PMC11011665 DOI: 10.3390/ijms25073810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
The objective of this study is to evaluate biomarkers for neurodegenerative disorders in adult SMA patients and their potential for monitoring the response to nusinersen. Biomarkers for neurodegenerative disorders were assessed in plasma and CSF samples obtained from a total of 30 healthy older adult controls and 31 patients with adult SMA type 2 and 3. The samples were collected before and during nusinersen treatment at various time points, approximately at 2, 6, 10, and 22 months. Using ELISA technology, the levels of total tau, pNF-H, NF-L, sAPPβ, Aβ40, Aβ42, and YKL-40 were evaluated in CSF samples. Additionally, plasma samples were used to measure NF-L and total tau levels using SIMOA technology. SMA patients showed improvements in clinical outcomes after nusinersen treatment, which were statistically significant only in walkers, in RULM (p = 0.04) and HFMSE (p = 0.05) at 24 months. A reduction in sAPPβ levels was found after nusinersen treatment, but these levels did not correlate with clinical outcomes. Other neurodegeneration biomarkers (NF-L, pNF-H, total tau, YKL-40, Aβ40, and Aβ42) were not found consistently changed with nusinersen treatment. The slow progression rate and mild treatment response of adult SMA types 2 and 3 may not lead to detectable changes in common markers of axonal degradation, inflammation, or neurodegeneration, since it does not involve large pools of damaged neurons as observed in pediatric forms. However, changes in biomarkers associated with the APP processing pathway might be linked to treatment administration. Further studies are warranted to better understand these findings.
Collapse
Affiliation(s)
- Pol Andrés-Benito
- Neurologic Diseases and Neurogenetics Group, Institute of Biomedical Research (IDIBELL), 08907 Barcelona, Spain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, 08907 Barcelona, Spain
| | - Juan Francisco Vázquez-Costa
- Neuromuscular Unit and ERN-NMD Group, Department of Neurology, Hospital Universitario y Politécnico La Fe and IIS La Fe, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46026 Valencia, Spain
- Department of Medicine, University of Valencia, 46021 Valencia, Spain
| | - Nancy Carolina Ñungo Garzón
- Neuromuscular Unit and ERN-NMD Group, Department of Neurology, Hospital Universitario y Politécnico La Fe and IIS La Fe, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46026 Valencia, Spain
| | - María J. Colomina
- Anesthesia and Critical Care Department, Bellvitge University Hospital-University of Barcelona, 08907 Barcelona, Spain
| | - Carla Marco
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELA), Department of Neurology, Bellvitge University Hospital, 08907 Barcelona, Spain
| | - Laura González
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELA), Department of Neurology, Bellvitge University Hospital, 08907 Barcelona, Spain
| | - Cristina Terrafeta
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELA), Department of Neurology, Bellvitge University Hospital, 08907 Barcelona, Spain
| | - Raúl Domínguez
- Neurologic Diseases and Neurogenetics Group, Institute of Biomedical Research (IDIBELL), 08907 Barcelona, Spain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, 08907 Barcelona, Spain
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELA), Department of Neurology, Bellvitge University Hospital, 08907 Barcelona, Spain
| | - Isidro Ferrer
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, 08907 Barcelona, Spain
- Neuropathology Group, Institute of Biomedical Research (IDIBELL), 08907 Barcelona, Spain
- Department of Pathology and Experimental Therapeutics, University of Barcelona, 08907 Barcelona, Spain
| | - Mónica Povedano
- Neurologic Diseases and Neurogenetics Group, Institute of Biomedical Research (IDIBELL), 08907 Barcelona, Spain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, 08907 Barcelona, Spain
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELA), Department of Neurology, Bellvitge University Hospital, 08907 Barcelona, Spain
| |
Collapse
|
12
|
Bisi N, Pinzi L, Rastelli G, Tonali N. Early Diagnosis of Neurodegenerative Diseases: What Has Been Undertaken to Promote the Transition from PET to Fluorescence Tracers. Molecules 2024; 29:722. [PMID: 38338465 PMCID: PMC10856728 DOI: 10.3390/molecules29030722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Alzheimer's Disease (AD) and Parkinson's Disease (PD) represent two among the most frequent neurodegenerative diseases worldwide. A common hallmark of these pathologies is the misfolding and consequent aggregation of amyloid proteins into soluble oligomers and insoluble β-sheet-rich fibrils, which ultimately lead to neurotoxicity and cell death. After a hundred years of research on the subject, this is the only reliable histopathological feature in our hands. Since AD and PD are diagnosed only once neuronal death and the first symptoms have appeared, the early detection of these diseases is currently impossible. At present, there is no effective drug available, and patients are left with symptomatic and inconclusive therapies. Several reasons could be associated with the lack of effective therapeutic treatments. One of the most important factors is the lack of selective probes capable of detecting, as early as possible, the most toxic amyloid species involved in the onset of these pathologies. In this regard, chemical probes able to detect and distinguish among different amyloid aggregates are urgently needed. In this article, we will review and put into perspective results from ex vivo and in vivo studies performed on compounds specifically interacting with such early species. Following a general overview on the three different amyloid proteins leading to insoluble β-sheet-rich amyloid deposits (amyloid β1-42 peptide, Tau, and α-synuclein), a list of the advantages and disadvantages of the approaches employed to date is discussed, with particular attention paid to the translation of fluorescence imaging into clinical applications. Furthermore, we also discuss how the progress achieved in detecting the amyloids of one neurodegenerative disease could be leveraged for research into another amyloidosis. As evidenced by a critical analysis of the state of the art, substantial work still needs to be conducted. Indeed, the early diagnosis of neurodegenerative diseases is a priority, and we believe that this review could be a useful tool for better investigating this field.
Collapse
Affiliation(s)
- Nicolò Bisi
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17, Av. des Sciences, 91400 Orsay, France
| | - Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy; (L.P.); (G.R.)
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy; (L.P.); (G.R.)
| | - Nicolò Tonali
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17, Av. des Sciences, 91400 Orsay, France
| |
Collapse
|
13
|
Wu YW, HuangFu WC, Lin TE, Peng CH, Tu HJ, Sung TY, Sung TY, Yen SC, Pan SL, Hsu KC. Identification of selective dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) inhibitors and their effects on tau and microtubule. Int J Biol Macromol 2024; 259:129074. [PMID: 38163507 DOI: 10.1016/j.ijbiomac.2023.129074] [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: 04/28/2023] [Revised: 11/17/2023] [Accepted: 12/25/2023] [Indexed: 01/03/2024]
Abstract
The overexpression of dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A), commonly observed in neurodegenerative diseases like Alzheimer's disease (AD) and Down syndrome (DS), can induce the formation of neurofibrillary tangles (NFTs) and amyloid plaques. Hence, designing a selective DYRK1A inhibitor would result in a promising small molecule for treating neurodegenerative diseases. Developing selective inhibitors for DYRK1A has been a difficult challenge due to the highly preserved ATP-binding site of protein kinases. In this study, we employed a structure-based virtual screening (SBVS) campaign targeting DYRK1A from a database containing 1.6 million compounds. Enzymatic assays were utilized to verify inhibitory properties, confirming that Y020-3945 and Y020-3957 showed inhibitory activity towards DYRK1A. In particular, the compounds exhibited high selectivity for DYRK1A over a panel of 120 kinases, reduced the phosphorylation of tau, and reversed the tubulin polymerization for microtubule stability. Additionally, treatment with the compounds significantly reduced the secretion of inflammatory cytokines IL-6 and TNF-α activated by DYRK1A-assisted NFTs and Aβ oligomers. These identified inhibitors possess promising therapeutic potential for conditions associated with DYRK1A in neurodegenerative diseases. The results showed that Y020-3945 and Y020-3957 demonstrated structural novelty compared to known DYRK1A inhibitors, making them a valuable addition to developing potential treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Wen Wu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Wei-Chun HuangFu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tony Eight Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chao-Hsiang Peng
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Huang-Ju Tu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Ting-Yi Sung
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Ying Sung
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Shih-Chung Yen
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong (Shenzhen), Shenzhen, Guangdong, People's Republic of China
| | - Shiow-Lin Pan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, Taiwan; Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
14
|
Cayir S, Volpi T, Toyonaga T, Gallezot JD, Yanghong Y, Sadabad FE, Mulnix T, Mecca AP, Fesharaki-Zadeh A, Matuskey D. Relationship between Neuroimaging and Cognition in Frontotemporal Dementia: A [18 F]FDG PET and Structural MRI Study. RESEARCH SQUARE 2024:rs.3.rs-3846125. [PMID: 38313264 PMCID: PMC10836106 DOI: 10.21203/rs.3.rs-3846125/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Background Frontotemporal dementia (FTD) is a clinically and pathologically heterogeneous condition with a prevalence comparable to Alzheimer's Disease for patients under sixty-five years of age. Gray matter (GM) atrophy and glucose hypometabolism are important biomarkers for the diagnosis and evaluation of disease progression in FTD. However, limited studies have systematically examined the association between cognition and neuroimaging in FTD using different imaging modalities in the same patient group. Methods We examined the association of cognition using Montreal Cognitive Assessment (MoCA) with both GM volume and glucose metabolism using structural magnetic resonance imaging (MRI) and 18F-fluorodeoxyglucose positron emission tomography scanning ([18F]FDG PET) in 21 patients diagnosed with FTD. Standardized uptake value ratio (SUVR) using the brainstem as a reference region was the primary outcome measure for [18F]FDG PET. Partial volume correction was applied to PET data to account for disease-related atrophy. Results Significant positive associations were found between whole-cortex GM volume and MoCA scores (r = 0.461, p = 0.035). The association between whole-cortex [18F]FDG SUVR and MoCA scores was not Significant (r = 0.374, p = 0.094). GM volumes of the frontal cortex (r = 0.540, p = 0.011), caudate (r = 0.616, p = 0.002), and insula (r = 0.568, p = 0.007) were also Significantly correlated with MoCA, as were SUVR values of the insula (r = 0.508, p = 0.018), thalamus (r = 0.478, p = 0.028), and posterior cingulate cortex (PCC) (r = 0.472, p = 0.030). Discussion Whole-cortex atrophy is associated with cognitive dysfunction, and this effect is larger than for cortical hypometabolism as measured with [18F]FDG PET. At the regional level, focal atrophy and/or hypometabolism in the frontal lobe, insula, PCC, thalamus, and caudate seem to imply the importance of these regions for the decline of cognitive function in FTD. Furthermore, these results highlight how functional and structural changes may not overlap and might contribute to cognitive dysfunction in FTD in different ways. Our findings provide insight into the relationships between structural, metabolic, and cognitive changes due to FTD.
Collapse
|
15
|
Krishnamurthy K, Pradhan RK. Emerging perspectives of synaptic biomarkers in ALS and FTD. Front Mol Neurosci 2024; 16:1279999. [PMID: 38249293 PMCID: PMC10796791 DOI: 10.3389/fnmol.2023.1279999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 12/01/2023] [Indexed: 01/23/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD) are debilitating neurodegenerative diseases with shared pathological features like transactive response DNA-binding protein of 43 kDa (TDP-43) inclusions and genetic mutations. Both diseases involve synaptic dysfunction, contributing to their clinical features. Synaptic biomarkers, representing proteins associated with synaptic function or structure, offer insights into disease mechanisms, progression, and treatment responses. These biomarkers can detect disease early, track its progression, and evaluate therapeutic efficacy. ALS is characterized by elevated neurofilament light chain (NfL) levels in cerebrospinal fluid (CSF) and blood, correlating with disease progression. TDP-43 is another key ALS biomarker, its mislocalization linked to synaptic dysfunction. In FTD, TDP-43 and tau proteins are studied as biomarkers. Synaptic biomarkers like neuronal pentraxins (NPs), including neuronal pentraxin 2 (NPTX2), and neuronal pentraxin receptor (NPTXR), offer insights into FTD pathology and cognitive decline. Advanced technologies, like machine learning (ML) and artificial intelligence (AI), aid biomarker discovery and drug development. Challenges in this research include technological limitations in detection, variability across patients, and translating findings from animal models. ML/AI can accelerate discovery by analyzing complex data and predicting disease outcomes. Synaptic biomarkers offer early disease detection, personalized treatment strategies, and insights into disease mechanisms. While challenges persist, technological advancements and interdisciplinary efforts promise to revolutionize the understanding and management of ALS and FTD. This review will explore the present comprehension of synaptic biomarkers in ALS and FTD and discuss their significance and emphasize the prospects and obstacles.
Collapse
Affiliation(s)
- Karrthik Krishnamurthy
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | | |
Collapse
|
16
|
El Hajjar L, Bridot C, Nguyen M, Cantrelle FX, Landrieu I, Smet-Nocca C. Phosphorylation of Tau Protein by CDK2/cyclin A and GSK3β Recombinant Kinases: Analysis of Phosphorylation Patterns by Nuclear Magnetic Resonance Spectroscopy. Methods Mol Biol 2024; 2754:271-306. [PMID: 38512672 DOI: 10.1007/978-1-0716-3629-9_15] [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: 03/23/2024]
Abstract
Posttranslational modifications (PTMs) of proteins can be investigated by Nuclear Magnetic Resonance (NMR) spectroscopy as a powerful analytical tool to define modification sites, their relative stoichiometry, and crosstalk between modifications. As a Structural Biology method, NMR provides important additional information on changes in protein conformation and dynamics upon modification as well as a mapping of binding sites upon biomolecular interactions. Indeed, PTMs not only mediate functional modulation in protein-protein interactions, but can also induce diverse structural responses with different biological outcomes. Here we present protocols that have been developed for the production and phosphorylation of the neuronal tau protein. Under its aggregated form, tau is a hallmark of Alzheimer's disease and other neurodegenerative diseases named tauopathies involving tau dysfunction and/or mutations. As a common feature shared by various tauopathies, tau aggregates are found into a form displaying an increased, abnormal phosphorylation, also referred to hyperphosphorylation. We have used NMR to investigate the phosphorylation patterns of tau induced by several kinases or cell extracts, how phosphorylation affects the local and overall conformation of tau, its interactions with partners (proteins, DNA, small-molecules, etc.) including tubulin and microtubules, and its capacity to form insoluble fibrillar aggregates. We present here detailed protocols for in vitro phosphorylation of tau by the recombinant kinases CDK2/cyclin A and GSK3β, the production of the recombinant kinases thereof, as well as the analytical characterization of phosphorylated tau by NMR spectroscopy.
Collapse
Affiliation(s)
- Léa El Hajjar
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
- CNRS EMR9002 Integrative Structural Biology, Lille, France
| | - Clarisse Bridot
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
- CNRS EMR9002 Integrative Structural Biology, Lille, France
| | - Marine Nguyen
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
- CNRS EMR9002 Integrative Structural Biology, Lille, France
| | - François-Xavier Cantrelle
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
- CNRS EMR9002 Integrative Structural Biology, Lille, France
| | - Isabelle Landrieu
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
- CNRS, EMR9002 BSI Integrative Structural Biology, Lille, France
- LabEx (Laboratory of Excellence) DISTALZ (Development of Innovative Strategies for a Transdisciplinary Approach to Alzheimer's Disease ANR-11-LABX-01), Lille, France
| | - Caroline Smet-Nocca
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France.
- CNRS EMR9002 Integrative Structural Biology, Lille, France.
| |
Collapse
|
17
|
Grari O, Elmoujtahide D, Sebbar E, Choukri M. The Biochemistry Behind Cognitive Decline: Biomarkers of Alzheimer's Disease. EJIFCC 2023; 34:276-283. [PMID: 38303754 PMCID: PMC10828533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent type of dementia. Pathologically, the disease is marked by neurofibrillary tangles (NFT), which are aberrant accumulations of the tau protein that develop inside neurons, and extracellular plaque deposits of the amyloid β peptide (Aβ). These pathological lesions are present in the brain before the beginning of clinical manifestations. However, despite advancements in the comprehension of AD pathophysiology, timely and accurate clinical diagnosis remains challenging. Therefore, developing biomarkers capable of detecting AD during the preclinical phase holds enormous promise for precise diagnosis since detecting the disease early is crucial because it enables interventions when treatments may be more effective. This article intends to provide a comprehensive review of AD biomarkers, discussing their significance, classification, and recent developments in the field.
Collapse
Affiliation(s)
- O. Grari
- : Faculty of Medicine and Pharmacy, Mohammed I University, Oujda, Morocco
- : Department of Biochemistry, Mohammed VI University Hospital, Oujda, Morocco
| | - D. Elmoujtahide
- : Faculty of Medicine and Pharmacy, Mohammed I University, Oujda, Morocco
- : Department of Biochemistry, Mohammed VI University Hospital, Oujda, Morocco
| | - E. Sebbar
- : Faculty of Medicine and Pharmacy, Mohammed I University, Oujda, Morocco
- : Department of Biochemistry, Mohammed VI University Hospital, Oujda, Morocco
| | - M. Choukri
- : Faculty of Medicine and Pharmacy, Mohammed I University, Oujda, Morocco
- : Department of Biochemistry, Mohammed VI University Hospital, Oujda, Morocco
| |
Collapse
|
18
|
Petrozziello T, Huntress SS, Castillo-Torres AL, Quinn JP, Connors TR, Auger CA, Mills AN, Kim SE, Liu S, Mahmood F, Boudi A, Wu M, Sapp E, Kivisäkk P, Sunderesh SR, Pouladi MA, Arnold SE, Hyman BT, Rosas HD, DiFiglia M, Pinto RM, Kegel-Gleason K, Sadri-Vakili G. Age-dependent increase in tau phosphorylation at serine 396 in Huntington's disease pre-frontal cortex. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.03.23290851. [PMID: 37333415 PMCID: PMC10274990 DOI: 10.1101/2023.06.03.23290851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Background To date, it is still controversial whether tau phosphorylation plays a role in Huntington's disease (HD), as previous studies demonstrated either no alterations or increases in phosphorylated tau (pTau) in HD post-mortem brain and mouse models. Objectives The goal of this study was to determine whether total tau and pTau levels are altered in HD. Methods Immunohistochemistry, cellular fractionations, and western blots were used to measure tau and pTau levels in a large cohort of HD and control post-mortem prefrontal cortex (PFC). Furthermore, western blots were performed to assess tau, and pTau levels in HD and control isogenic embryonic stem cell (ESC)-derived cortical neurons and neuronal stem cells (NSCs). Similarly, western blots were used to assess tau and pTau in Htt Q111 and transgenic R6/2 mice. Lastly, total tau levels were assessed in HD and healthy control plasma using Quanterix Simoa assay. Results Our results revealed that, while there was no difference in tau or pTau levels in HD PFC compared to controls, tau phosphorylated at S396 levels were increased in PFC samples from HD patients 60 years or older at time of death. Additionally, tau and pTau levels were not changed in HD ESC-derived cortical neurons and NSCs. Similarly, tau or pTau levels were not altered in Htt Q111 and transgenic R6/2 mice compared to wild-type littermates. Lastly, tau levels were not changed in plasma from a small cohort of HD patients compared to controls. Conclusion Together these findings demonstrate that pTau-S396 levels increase significantly with age in HD PFC.
Collapse
|
19
|
Salvador CL, Oppebøen M, Vassli AØ, Pfeiffer HCV, Varhaug KN, Elgstøen KBP, Yazdani M. Increased Sphingomyelin and Free Sialic Acid in Cerebrospinal Fluid of Kearns-Sayre Syndrome: New Findings Using Untargeted Metabolomics. Pediatr Neurol 2023; 143:68-76. [PMID: 37018879 DOI: 10.1016/j.pediatrneurol.2023.02.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/10/2023] [Accepted: 02/25/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Kearns-Sayre syndrome (KSS) is caused by duplications and/or deletions of mitochondrial DNA (mtDNA) and is typically diagnosed based on a classic triad of symptoms with chronic progressive external ophthalmoplegia (CPEO), retinitis pigmentosa, and onset before age 20 years. The present study aimed to diagnose two patients, on suspicion of KSS. METHODS One of the patients went through a diagnostic odyssey, with normal results from several mtDNA analyses, both in blood and muscle, before the diagnosis was confirmed genetically. RESULTS Two patients presented increased tau protein and low 5-methyltetrahydrofolate (5-MTHF) levels in the cerebrospinal fluid (CSF). Untargeted metabolomics on CSF samples also showed an increase in the levels of free sialic acid and sphingomyelin C16:0 (d18:1/C16:0), compared with four control groups (patients with mitochondrial disorders, nonmitochondrial disorders, low 5-MTHF, or increased tau proteins). CONCLUSIONS It is the first time that elevated sphingomyelin C16:0 (d18:1/C16:0) and tau protein in KSS are reported. Using an untargeted metabolomics approach and standard laboratory methods, the study could shed new light on metabolism in KSS to better understand its complexity. In addition, the findings may suggest the combination of elevated free sialic acid, sphingomyelin C16:0 (d18:1/C16:0), and tau protein as well as low 5-MTHF as new biomarkers in the diagnostics of KSS.
Collapse
Affiliation(s)
| | - Mari Oppebøen
- Department of Pediatrics, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Anja Østeby Vassli
- Department of Medical Biochemistry, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Helle Cecilie Viekilde Pfeiffer
- Department of Pediatrics, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Department of Pediatrics, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Kristin Nielsen Varhaug
- The Mitochondrial Medicine and Neurogenetics (MMN) Group, Department of Clinical Medicine, University of Bergen, Bergen, Norway; Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | | | - Mazyar Yazdani
- Department of Medical Biochemistry, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
20
|
Musco H, Beecher K, Chand KK, Colditz PB, Wixey JA. Blood Biomarkers in the Fetally Growth Restricted and Small for Gestational Age Neonate: Associations with Brain Injury. Dev Neurosci 2023; 46:84-97. [PMID: 37231871 DOI: 10.1159/000530492] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 03/29/2023] [Indexed: 05/27/2023] Open
Abstract
Fetal growth restriction (FGR) and small for gestational age (SGA) infants have increased risk of mortality and morbidity. Although both FGR and SGA infants have low birthweights for gestational age, a diagnosis of FGR also requires assessments of umbilical artery Doppler, physiological determinants, neonatal features of malnutrition, and in utero growth retardation. Both FGR and SGA are associated with adverse neurodevelopmental outcomes ranging from learning and behavioral difficulties to cerebral palsy. Up to 50% of FGR, newborns are not diagnosed until around the time of birth, yet this diagnosis lacks further indication of the risk of brain injury or adverse neurodevelopmental outcomes. Blood biomarkers may be a promising tool. Defining blood biomarkers indicating an infant's risk of brain injury would provide the opportunity for early detection and therefore earlier support. The aim of this review was to summarize the current literature to assist in guiding the future direction for the early detection of adverse brain outcomes in FGR and SGA neonates. The studies investigated potential diagnostic blood biomarkers from cord and neonatal blood or serum from FGR and SGA human neonates. Results were often conflicting with heterogeneity common in the biomarkers examined, timepoints, gestational age, and definitions of FGR and SGA used. Due to these variations, it was difficult to draw strong conclusions from the results. The search for blood biomarkers of brain injury in FGR and SGA neonates should continue as early detection and intervention is critical to improve outcomes for these neonates.
Collapse
Affiliation(s)
- Hannah Musco
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Kate Beecher
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Kirat K Chand
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Paul B Colditz
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
- Perinatal Research Centre, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Julie A Wixey
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
21
|
Negi D, Granak S, Shorter S, O'Leary VB, Rektor I, Ovsepian SV. Molecular Biomarkers of Neuronal Injury in Epilepsy Shared with Neurodegenerative Diseases. Neurotherapeutics 2023; 20:767-778. [PMID: 36884195 PMCID: PMC10275849 DOI: 10.1007/s13311-023-01355-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2023] [Indexed: 03/09/2023] Open
Abstract
In neurodegenerative diseases, changes in neuronal proteins in the cerebrospinal fluid and blood are viewed as potential biomarkers of the primary pathology in the central nervous system (CNS). Recent reports suggest, however, that level of neuronal proteins in fluids also alters in several types of epilepsy in various age groups, including children. With increasing evidence supporting clinical and sub-clinical seizures in Alzheimer's disease, Lewy body dementia, Parkinson's disease, and in other less common neurodegenerative conditions, these findings call into question the specificity of neuronal protein response to neurodegenerative process and urge analysis of the effects of concomitant epilepsy and other comorbidities. In this article, we revisit the evidence for alterations in neuronal proteins in the blood and cerebrospinal fluid associated with epilepsy with and without neurodegenerative diseases. We discuss shared and distinctive characteristics of changes in neuronal markers, review their neurobiological mechanisms, and consider the emerging opportunities and challenges for their future research and diagnostic use.
Collapse
Affiliation(s)
- Deepika Negi
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, ME4 4TB, UK
| | - Simon Granak
- National Institute of Mental Health, Topolova 748, Klecany, 25067, Czech Republic
| | - Susan Shorter
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, ME4 4TB, UK
| | - Valerie B O'Leary
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Ruská 87, Prague, 10000, Czech Republic
| | - Ivan Rektor
- First Department of Neurology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Multimodal and Functional Neuroimaging Research Group, CEITEC-Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Saak V Ovsepian
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, ME4 4TB, UK.
| |
Collapse
|
22
|
Yin C, Harms AC, Hankemeier T, Kindt A, de Lange ECM. Status of Metabolomic Measurement for Insights in Alzheimer's Disease Progression-What Is Missing? Int J Mol Sci 2023; 24:ijms24054960. [PMID: 36902391 PMCID: PMC10003384 DOI: 10.3390/ijms24054960] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/24/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Alzheimer's disease (AD) is an aging-related neurodegenerative disease, leading to the progressive loss of memory and other cognitive functions. As there is still no cure for AD, the growth in the number of susceptible individuals represents a major emerging threat to public health. Currently, the pathogenesis and etiology of AD remain poorly understood, while no efficient treatments are available to slow down the degenerative effects of AD. Metabolomics allows the study of biochemical alterations in pathological processes which may be involved in AD progression and to discover new therapeutic targets. In this review, we summarized and analyzed the results from studies on metabolomics analysis performed in biological samples of AD subjects and AD animal models. Then this information was analyzed by using MetaboAnalyst to find the disturbed pathways among different sample types in human and animal models at different disease stages. We discuss the underlying biochemical mechanisms involved, and the extent to which they could impact the specific hallmarks of AD. Then we identify gaps and challenges and provide recommendations for future metabolomics approaches to better understand AD pathogenesis.
Collapse
Affiliation(s)
- Chunyuan Yin
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Amy C. Harms
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Thomas Hankemeier
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Alida Kindt
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Elizabeth C. M. de Lange
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
- Correspondence:
| |
Collapse
|
23
|
Rossi S, Silvestri G. Fluid Biomarkers of Central Nervous System (CNS) Involvement in Myotonic Dystrophy Type 1 (DM1). Int J Mol Sci 2023; 24:ijms24032204. [PMID: 36768526 PMCID: PMC9917343 DOI: 10.3390/ijms24032204] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/13/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1), commonly known as Steinert's disease (OMIM #160900), is the most common muscular dystrophy among adults, caused by an unstable expansion of a CTG trinucleotide repeat in the 3' untranslated region (UTR) of DMPK. Besides skeletal muscle, central nervous system (CNS) involvement is one of the core manifestations of DM1, whose relevant cognitive, behavioral, and affective symptoms deeply affect quality of life of DM1 patients, and that, together with muscle and heart, may profoundly influence the global disease burden and overall prognosis. Therefore, CNS should be also included among the main targets for future therapeutic developments in DM1, and, in this regard, identifying a cost-effective, easily accessible, and sensitive diagnostic and monitoring biomarker of CNS involvement in DM1 represents a relevant issue to be addressed. In this mini review, we will discuss all the papers so far published exploring the usefulness of both cerebrospinal fluid (CSF) and blood-based biomarkers of CNS involvement in DM1. Globally, the results of these studies are quite consistent on the value of CSF and blood Neurofilament Light Chain (NfL) as a biomarker of CNS involvement, with less robust results regarding levels of tau protein or amyloid-beta.
Collapse
Affiliation(s)
- Salvatore Rossi
- Department of Neuroscience, Università Cattolica del Sacro Cuore–Sede di Roma, Largo F. Vito 1, 00168 Rome, Italy
| | - Gabriella Silvestri
- Department of Neuroscience, Università Cattolica del Sacro Cuore–Sede di Roma, Largo F. Vito 1, 00168 Rome, Italy
- Neurology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
- Correspondence:
| |
Collapse
|
24
|
Harris G, Rickard JJS, Butt G, Kelleher L, Blanch RJ, Cooper J, Oppenheimer PG. Review: Emerging Eye-Based Diagnostic Technologies for Traumatic Brain Injury. IEEE Rev Biomed Eng 2023; 16:530-559. [PMID: 35320105 PMCID: PMC9888755 DOI: 10.1109/rbme.2022.3161352] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 02/11/2022] [Accepted: 03/15/2022] [Indexed: 11/06/2022]
Abstract
The study of ocular manifestations of neurodegenerative disorders, Oculomics, is a growing field of investigation for early diagnostics, enabling structural and chemical biomarkers to be monitored overtime to predict prognosis. Traumatic brain injury (TBI) triggers a cascade of events harmful to the brain, which can lead to neurodegeneration. TBI, termed the "silent epidemic" is becoming a leading cause of death and disability worldwide. There is currently no effective diagnostic tool for TBI, and yet, early-intervention is known to considerably shorten hospital stays, improve outcomes, fasten neurological recovery and lower mortality rates, highlighting the unmet need for techniques capable of rapid and accurate point-of-care diagnostics, implemented in the earliest stages. This review focuses on the latest advances in the main neuropathophysiological responses and the achievements and shortfalls of TBI diagnostic methods. Validated and emerging TBI-indicative biomarkers are outlined and linked to ocular neuro-disorders. Methods detecting structural and chemical ocular responses to TBI are categorised along with prospective chemical and physical sensing techniques. Particular attention is drawn to the potential of Raman spectroscopy as a non-invasive sensing of neurological molecular signatures in the ocular projections of the brain, laying the platform for the first tangible path towards alternative point-of-care diagnostic technologies for TBI.
Collapse
Affiliation(s)
- Georgia Harris
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical SciencesUniversity of BirminghamB15 2TTBirminghamU.K.
| | - Jonathan James Stanley Rickard
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical SciencesUniversity of BirminghamB15 2TTBirminghamU.K.
- Department of Physics, Cavendish LaboratoryUniversity of CambridgeCB3 0HECambridgeU.K.
| | - Gibran Butt
- Ophthalmology DepartmentUniversity Hospitals Birmingham NHS Foundation TrustB15 2THBirminghamU.K.
| | - Liam Kelleher
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical SciencesUniversity of BirminghamB15 2TTBirminghamU.K.
| | - Richard James Blanch
- Department of Military Surgery and TraumaRoyal Centre for Defence MedicineB15 2THBirminghamU.K.
- Neuroscience and Ophthalmology, Department of Ophthalmology, University Hospitals Birmingham NHS Foundation TrustcBirminghamU.K.
| | - Jonathan Cooper
- School of Biomedical EngineeringUniversity of GlasgowG12 8LTGlasgowU.K.
| | - Pola Goldberg Oppenheimer
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical SciencesUniversity of BirminghamB15 2TTBirminghamU.K.
- Healthcare Technologies Institute, Institute of Translational MedicineB15 2THBirminghamU.K.
| |
Collapse
|
25
|
Sangalli L, Boggero IA. The impact of sleep components, quality and patterns on glymphatic system functioning in healthy adults: A systematic review. Sleep Med 2023; 101:322-349. [PMID: 36481512 DOI: 10.1016/j.sleep.2022.11.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/04/2022] [Accepted: 11/13/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The glymphatic system is thought to be responsible for waste clearance in the brain. As it is primarily active during sleep, different components of sleep, subjective sleep quality, and sleep patterns may contribute to glymphatic functioning. This systematic review aimed at exploring the effect of sleep components, sleep quality, and sleep patterns on outcomes associated with the glymphatic system in healthy adults. METHODS PubMed®, Scopus, and Web of Science were searched for studies published in English until December 2021. Articles subjectively or objectively investigating sleep components (total sleep time, time in bed, sleep efficiency, sleep onset latency, wake-up after sleep onset, sleep stage, awakenings), sleep quality, or sleep pattern in healthy individuals, on outcomes associated with glymphatic system (levels of amyloid-β, tau, α-synuclein; cerebrospinal fluid, perivascular spaces; apolipoprotein E) were selected. RESULTS Out of 8359 records screened, 51 studies were included. Overall, contradictory findings were observed according to different sleep assessment method. The most frequently assessed sleep parameters were total sleep time, sleep quality, and sleep efficiency. No association was found between sleep efficiency and amyloid-β, and between slow-wave activity and tau. Most of the studies did not find any correlation between total sleep time and amyloid-β nor tau level. Opposing results correlated sleep quality with amyloid-β and tau. CONCLUSIONS This review highlighted inconsistent results across the studies; as such, the specific association between the glymphatic system and sleep parameters in healthy adults remains poorly understood. Due to the heterogeneity of sleep assessment methods and the self-reported data representing the majority of the observations, future studies with universal study design and sleep methodology in healthy individuals are advocated.
Collapse
Affiliation(s)
- L Sangalli
- Department of Oral Health Science, Division of Orofacial Pain, University of Kentucky, College of Dentistry, Lexington, Kentucky, USA; College of Dental Medicine - Illinois, Downers Grove, Illinois, USA.
| | - I A Boggero
- Department of Oral Health Science, Division of Orofacial Pain, University of Kentucky, College of Dentistry, Lexington, Kentucky, USA; Department of Psychology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
26
|
Panikkar D, Vivek S, Crimmins E, Faul J, Langa KM, Thyagarajan B. Pre-Analytical Variables Influencing Stability of Blood-Based Biomarkers of Neuropathology. J Alzheimers Dis 2023; 95:735-748. [PMID: 37574735 PMCID: PMC11520930 DOI: 10.3233/jad-230384] [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: 08/15/2023]
Abstract
BACKGROUND Sample collection and preanalytical protocols may significantly impact the results of large-scale epidemiological studies incorporating blood-based biomarkers of neuropathology. OBJECTIVE To evaluate the stability and assay variability of several blood-based biomarkers of neuropathology for common preanalytical conditions. METHODS We collected serum and plasma samples from 41 participants and evaluated the effect of processing delay of up to 72 h when stored at 4∘C, three freeze-thaw cycles, and a combination of 48-h processing delay when stored at 4∘C and three freeze-thaw cycles on biomarker stability. Using the Simoa assay (Quanterix Inc.), we measured amyloid-β 40 (Aβ40), amyloid-β 42 (Aβ42), neurofilament light (NfL), glial fibrillary acidic protein (GFAP), and phosphorylated tau 181 (p-tau-181). RESULTS We found that Aβ40 and Aβ42 levels significantly decreased after a 24-h processing delay in both plasma and serum samples, and a single freeze-thaw cycle (p < 0.0001). Nevertheless, serum Aβ42/40 ratio remained stable with a processing delay up to 48 h while plasma Aβ42/40 ratio showed only small but significant increase with a delay up to 72 h. Both plasma and serum GFAP and NfL levels were only modestly affected by processing delay and freeze-thaw cycles. Plasma p-tau-181 levels notably increased with a 24-, 48-, and 72-h processing delay, but remained stable in serum. Intra-individual variation over two weeks was minimal for all biomarkers and their levels were substantially lower in serum when compared with plasma. CONCLUSION These results suggest that standardizing preanalytical variables will allow robust measurements of biomarkers of neuropathology in population studies.
Collapse
Affiliation(s)
- Daniel Panikkar
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Sithara Vivek
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Eileen Crimmins
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Jessica Faul
- Institute for Social Research, Survey Research Center, University of Michigan, Ann Arbor, MI, USA
| | - Kenneth M. Langa
- Institute for Social Research, Survey Research Center, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Veterans Affairs Ann Arbor Center for Clinical Management Research, Ann Arbor, MI, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
27
|
Petrozziello T, Huntress SS, Castillo-Torres AL, Quinn JP, Connors TR, Auger CA, Mills AN, Kim SE, Liu S, Mahmood F, Boudi A, Wu M, Sapp E, Kivisäkk P, Sunderesh SR, Pouladi MA, Arnold SE, Hyman BT, Rosas HD, DiFiglia M, Mouro Pinto R, Kegel-Gleason K, Sadri-Vakili G. Age-Dependent Increase in Tau Phosphorylation at Serine 396 in Huntington's Disease Prefrontal Cortex. J Huntingtons Dis 2023; 12:267-281. [PMID: 37694372 DOI: 10.3233/jhd-230588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
BACKGROUND To date, it is still controversial whether tau phosphorylation plays a role in Huntington's disease (HD), as previous studies demonstrated either no alterations or increases in phosphorylated tau (pTau) in HD postmortem brain and mouse models. OBJECTIVE The goal of this study was to determine whether total tau and pTau levels are altered in HD. METHODS Immunohistochemistry, cellular fractionations, and western blots were used to measure total tau and pTau levels in a large cohort of HD and control postmortem prefrontal cortex (PFC). Furthermore, western blots were performed to assess tau, and pTau levels in HD and control isogenic embryonic stem cell (ESC)-derived cortical neurons and neuronal stem cells (NSCs). Similarly, western blots were used to assess tau and pTau levels in HttQ111 and transgenic R6/2 mice. Lastly, total tau levels were assessed in HD and healthy control plasma using Quanterix Simoa assay. RESULTS Our results revealed that, while there was no difference in total tau or pTau levels in HD PFC compared to controls, the levels of tau phosphorylated at S396 were increased in PFC samples from HD patients 60 years or older at time of death. Additionally, tau and pTau levels were not changed in HD ESC-derived cortical neurons and NSCs. Similarly, total tau or pTau levels were not altered in HttQ111 and transgenic R6/2 mice compared to wild-type littermates. Lastly, tau levels were not changed in plasma from a small cohort of HD patients compared to controls. CONCLUSIONS Together these findings demonstrate that pTau-S396 levels increase significantly with age in HD PFC.
Collapse
Affiliation(s)
- Tiziana Petrozziello
- Sean M. Healey & AMG Center for ALS at Mass General, MassGeneral Brigham, Boston, MA, USA
| | - Sommer S Huntress
- Sean M. Healey & AMG Center for ALS at Mass General, MassGeneral Brigham, Boston, MA, USA
| | | | - James P Quinn
- Department of Neurology, MassGeneral Brigham, Boston, MA, USA
| | | | - Corinne A Auger
- Department of Neurology, MassGeneral Brigham, Boston, MA, USA
| | - Alexandra N Mills
- Sean M. Healey & AMG Center for ALS at Mass General, MassGeneral Brigham, Boston, MA, USA
| | - Spencer E Kim
- Sean M. Healey & AMG Center for ALS at Mass General, MassGeneral Brigham, Boston, MA, USA
| | - Sophia Liu
- Department of Neurology, MassGeneral Brigham, Boston, MA, USA
| | - Farah Mahmood
- Department of Neurology, MassGeneral Brigham, Boston, MA, USA
| | - Adel Boudi
- Department of Neurology, MassGeneral Brigham, Boston, MA, USA
| | - Muzhou Wu
- Center for Genomic Medicine, MassGeneral Brigham, Boston, MA, USA
| | - Ellen Sapp
- Department of Neurology, MassGeneral Brigham, Boston, MA, USA
| | - Pia Kivisäkk
- Department of Neurology, MassGeneral Brigham, Boston, MA, USA
| | | | - Mahmoud A Pouladi
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Steven E Arnold
- Department of Neurology, MassGeneral Brigham, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, MassGeneral Brigham, Boston, MA, USA
| | - H Diana Rosas
- Department of Neurology, MassGeneral Brigham, Boston, MA, USA
| | - Marian DiFiglia
- Department of Neurology, MassGeneral Brigham, Boston, MA, USA
| | - Ricardo Mouro Pinto
- Department of Neurology, MassGeneral Brigham, Boston, MA, USA
- Center for Genomic Medicine, MassGeneral Brigham, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | | | - Ghazaleh Sadri-Vakili
- Sean M. Healey & AMG Center for ALS at Mass General, MassGeneral Brigham, Boston, MA, USA
| |
Collapse
|
28
|
Mahali S, Martinez R, King M, Verbeck A, Harari O, Benitez BA, Horie K, Sato C, Temple S, Karch CM. Defective proteostasis in induced pluripotent stem cell models of frontotemporal lobar degeneration. Transl Psychiatry 2022; 12:508. [PMID: 36494352 PMCID: PMC9734180 DOI: 10.1038/s41398-022-02274-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
Impaired proteostasis is associated with normal aging and is accelerated in neurodegeneration. This impairment may lead to the accumulation of protein, which can be toxic to cells and tissue. In a subset of frontotemporal lobar degeneration with tau pathology (FTLD-tau) cases, pathogenic mutations in the microtubule-associated protein tau (MAPT) gene are sufficient to cause tau accumulation and neurodegeneration. However, the pathogenic events triggered by the expression of the mutant tau protein remain poorly understood. Here, we show that molecular networks associated with lysosomal biogenesis and autophagic function are disrupted in brains from FTLD-tau patients carrying a MAPT p.R406W mutation. We then used human induced pluripotent stem cell (iPSC)-derived neurons and 3D cerebral organoids from patients carrying the MAPT p.R406W mutation and CRISPR/Cas9, corrected controls to evaluate proteostasis. MAPT p.R406W was sufficient to induce morphological and functional deficits in the lysosomal pathway in iPSC-neurons. These phenotypes were reversed upon correction of the mutant allele with CRISPR/Cas9. Treatment with mTOR inhibitors led to tau degradation specifically in MAPT p.R406W neurons. Together, our findings suggest that MAPT p.R406W is sufficient to cause impaired lysosomal function, which may contribute to disease pathogenesis and serve as a cellular phenotype for drug screening.
Collapse
Affiliation(s)
- Sidhartha Mahali
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Rita Martinez
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Melvin King
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Anthony Verbeck
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Oscar Harari
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, USA
| | - Bruno A Benitez
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, USA
| | - Kanta Horie
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Chihiro Sato
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | | | - Celeste M Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA.
- Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, USA.
| |
Collapse
|
29
|
Serum glial fibrillary acidic protein is a body fluid biomarker: A valuable prognostic for neurological disease – A systematic review. Int Immunopharmacol 2022; 107:108624. [DOI: 10.1016/j.intimp.2022.108624] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/28/2022] [Accepted: 02/10/2022] [Indexed: 12/14/2022]
|
30
|
Kraus D, Abdelrahim H, Waisbourd-Zinman O, Domin E, Zeharia A, Staretz-Chacham O. Elevated Alpha-Fetoprotein in Infantile-Onset Niemann-Pick Type C Disease with Liver Involvement. CHILDREN 2022; 9:children9040545. [PMID: 35455589 PMCID: PMC9032157 DOI: 10.3390/children9040545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 11/29/2022]
Abstract
Niemann-Pick disease type C (NPC) is a rare autosomal recessive neuro-visceral lipid storage disease. We describe nine cases of infantile-onset NPC with various genetic mutations in the NPC1 gene, which presented with neonatal cholestasis. Serum alpha-fetoprotein (AFP) levels were obtained as part of their workup during the first four months of life. In eight of nine (89%) patients, serum AFP demonstrated elevated levels. Seven infants displayed marked elevations, ranging from 4 to 300 times the upper limit for age-adjusted norms. In most patients, AFP levels peaked during the initial test and declined over time as cholestasis resolved. We conclude that elevated AFP levels are a common, although non-specific, marker for NPC-associated liver disease. These findings demonstrate the benefit of including AFP levels in the workup of neonatal liver disease, especially if there is accompanied cholestasis and if NPC is suspected.
Collapse
Affiliation(s)
- Dror Kraus
- Institute of Neurology, Schneider Children’s Medical Center of Israel, Petach-Tikva 4920235, Israel; (D.K.); (H.A.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; (O.W.-Z.); (A.Z.)
| | - Huda Abdelrahim
- Institute of Neurology, Schneider Children’s Medical Center of Israel, Petach-Tikva 4920235, Israel; (D.K.); (H.A.)
| | - Orith Waisbourd-Zinman
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; (O.W.-Z.); (A.Z.)
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children’s Medical Center of Israel, Petach-Tikva 4920235, Israel
| | - Elena Domin
- Clinical Biochemistry (Metabolic) Laboratory, Sheba Medical Center, Ramat Gan 52621, Israel;
| | - Avraham Zeharia
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; (O.W.-Z.); (A.Z.)
- Day Hospitalization Department, Schneider Children’s Medical Center of Israel, Petach-Tikva 4920235, Israel
| | - Orna Staretz-Chacham
- Pediatric Metabolic Clinic, Pediatric Division, Soroka Medical Center, Ben-Gurion University, Beer Sheva 8480101, Israel
- Correspondence: ; Tel.: +972-8-6400508; Fax: +972-8-6400545
| |
Collapse
|
31
|
Sarnowski C, Ghanbari M, Bis JC, Logue M, Fornage M, Mishra A, Ahmad S, Beiser AS, Boerwinkle E, Bouteloup V, Chouraki V, Cupples LA, Damotte V, DeCarli CS, DeStefano AL, Djoussé L, Fohner AE, Franz CE, Kautz TF, Lambert JC, Lyons MJ, Mosley TH, Mukamal KJ, Pase MP, Portilla Fernandez EC, Rissman RA, Satizabal CL, Vasan RS, Yaqub A, Debette S, Dufouil C, Launer LJ, Kremen WS, Longstreth WT, Ikram MA, Seshadri S. Meta-analysis of genome-wide association studies identifies ancestry-specific associations underlying circulating total tau levels. Commun Biol 2022; 5:336. [PMID: 35396452 PMCID: PMC8993877 DOI: 10.1038/s42003-022-03287-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 03/17/2022] [Indexed: 01/27/2023] Open
Abstract
Circulating total-tau levels can be used as an endophenotype to identify genetic risk factors for tauopathies and related neurological disorders. Here, we confirmed and better characterized the association of the 17q21 MAPT locus with circulating total-tau in 14,721 European participants and identified three novel loci in 953 African American participants (4q31, 5p13, and 6q25) at P < 5 × 10-8. We additionally detected 14 novel loci at P < 5 × 10-7, specific to either Europeans or African Americans. Using whole-exome sequence data in 2,279 European participants, we identified ten genes associated with circulating total-tau when aggregating rare variants. Our genetic study sheds light on genes reported to be associated with neurological diseases including stroke, Alzheimer's, and Parkinson's (F5, MAP1B, and BCAS3), with Alzheimer's pathological hallmarks (ADAMTS12, IL15, and FHIT), or with an important function in the brain (PARD3, ELFN2, UBASH3B, SLIT3, and NSD3), and suggests that the genetic architecture of circulating total-tau may differ according to ancestry.
Collapse
Affiliation(s)
- Chloé Sarnowski
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mark Logue
- National Center for PTSD, Behavioral Sciences Division, VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry and Biomedical Genetics, Boston University School of Medicine, Boston, MA, USA
| | - Myriam Fornage
- University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Aniket Mishra
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000, Bordeaux, France
| | - Shahzad Ahmad
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Alexa S Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Eric Boerwinkle
- University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Vincent Bouteloup
- Centre Inserm U1219 Bordeaux Population Health, CIC1401-EC, Institut de Santé Publique, d'Epidémiologie et de Développement, Université de Bordeaux, CHU de Bordeaux, Pôle Santé Publique, Bordeaux, France
| | - Vincent Chouraki
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE- LabEx DISTALZ - Risk factors and molecular determinants of aging diseases, F-59000, Lille, France
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
| | - Vincent Damotte
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE- LabEx DISTALZ - Risk factors and molecular determinants of aging diseases, F-59000, Lille, France
| | - Charles S DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Davis, CA, USA
| | - Anita L DeStefano
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Luc Djoussé
- Department of Medicine, Division of Aging, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alison E Fohner
- Institute of Public Health Genetics and Department of Epidemiology and Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - Carol E Franz
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, USA
| | - Tiffany F Kautz
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Jean-Charles Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE- LabEx DISTALZ - Risk factors and molecular determinants of aging diseases, F-59000, Lille, France
| | - Michael J Lyons
- Department of Psychology and Brain Sciences, Boston University, Boston, MA, USA
| | | | - Kenneth J Mukamal
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Matthew P Pase
- Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | | | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Claudia L Satizabal
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Ramachandran S Vasan
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Preventive Medicine & Epidemiology, Boston University School of Medicine, Boston, MA, USA
| | - Amber Yaqub
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Stephanie Debette
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000, Bordeaux, France
- Bordeaux University Hospital, Department of Neurology, Institute for Neurodegenerative Diseases, Bordeaux, France
| | - Carole Dufouil
- Bordeaux University Hospital, Department of Neurology, Institute for Neurodegenerative Diseases, Bordeaux, France
| | | | - William S Kremen
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, USA
| | - William T Longstreth
- Departments of Neurology and Epidemiology, University of Washington, Seattle, WA, USA
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sudha Seshadri
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| |
Collapse
|
32
|
Papapetropoulos S, Pontius A, Finger E, Karrenbauer V, Lynch DS, Brennan M, Zappia S, Koehler W, Schoels L, Hayer SN, Konno T, Ikeuchi T, Lund T, Orthmann-Murphy J, Eichler F, Wszolek ZK. Adult-Onset Leukoencephalopathy With Axonal Spheroids and Pigmented Glia: Review of Clinical Manifestations as Foundations for Therapeutic Development. Front Neurol 2022; 12:788168. [PMID: 35185751 PMCID: PMC8850408 DOI: 10.3389/fneur.2021.788168] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/14/2021] [Indexed: 12/11/2022] Open
Abstract
A comprehensive review of published literature was conducted to elucidate the genetics, neuropathology, imaging findings, prevalence, clinical course, diagnosis/clinical evaluation, potential biomarkers, and current and proposed treatments for adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP), a rare, debilitating, and life-threatening neurodegenerative disorder for which disease-modifying therapies are not currently available. Details on potential efficacy endpoints for future interventional clinical trials in patients with ALSP and data related to the burden of the disease on patients and caregivers were also reviewed. The information in this position paper lays a foundation to establish an effective clinical rationale and address the clinical gaps for creation of a robust strategy to develop therapeutic agents for ALSP, as well as design future clinical trials, that have clinically meaningful and convergent endpoints.
Collapse
Affiliation(s)
- Spyros Papapetropoulos
- Vigil Neuroscience, Inc, Cambridge, MA, United States
- Massachusetts General Hospital, Boston, MA, United States
| | | | - Elizabeth Finger
- Clinical Neurological Sciences, Western University, London, ON, Canada
| | - Virginija Karrenbauer
- Neurology Medical Unit, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - David S. Lynch
- National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | | | | | | | - Ludger Schoels
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University Hospital Tuebingen, Tuebingen, Germany
- German Research Center for Neurodegenerative Diseases, Tuebingen, Germany
| | - Stefanie N. Hayer
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University Hospital Tuebingen, Tuebingen, Germany
- German Research Center for Neurodegenerative Diseases, Tuebingen, Germany
| | - Takuya Konno
- Brain Research Institute, Niigata University, Niigata, Japan
| | - Takeshi Ikeuchi
- Brain Research Institute, Niigata University, Niigata, Japan
| | - Troy Lund
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | | | | | | |
Collapse
|
33
|
Amyotrophic lateral sclerosis: Correlations between fluid biomarkers of NfL, TDP-43, and tau, and clinical characteristics. PLoS One 2021; 16:e0260323. [PMID: 34843548 PMCID: PMC8629269 DOI: 10.1371/journal.pone.0260323] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/07/2021] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES We previously reported the diagnostic and prognostic performance of neurofilament light chain (NfL), TAR DNA-binding protein 43 (TDP-43), and total tau (t-tau) in cerebrospinal fluid (CSF) and plasma as amyotrophic lateral sclerosis (ALS) biomarkers. The present study aimed to elucidate associations between clinical characteristics and the markers as well as mutual associations of the markers in ALS patients using the same dataset. METHODS NfL, TDP-43, and t-tau levels in CSF and plasma in 75 ALS patients were analyzed. The associations between those markers and clinical details were investigated by uni- and multivariate analyses. Correlations between the markers were analyzed univariately. RESULTS In multivariate analysis of CSF proteins, the disease progression rate (DPR) was positively correlated with NfL (β: 0.51, p = 0.007) and t-tau (β: 0.37, p = 0.03). Plasma NfL was correlated with age (β: 0.53, p = 0.005) and diagnostic grade (β: -0.42, p = 0.02) in multivariate analysis. Plasma TDP-43 was correlated negatively with split hand index (β: -0.48, p = 0.04) and positively with % vital capacity (β: 0.64, p = 0.03) in multivariate analysis. Regarding mutual biomarker analysis, a negative correlation between CSF-NfL and TDP-43 was identified (r: -0.36, p = 0.002). CONCLUSIONS Elevated NfL and t-tau levels in CSF may be biomarkers to predict rapid DPR from onset to sample collection. The negative relationship between CSF NfL and TDP-43 suggests that elevation of CSF TDP-43 in ALS is not a simple consequence of its release into CSF during neurodegeneration. The negative correlation between plasma TDP-43 and split hand index may support the pathophysiological association between plasma TDP-43 and ALS.
Collapse
|
34
|
Jiao B, Liu H, Guo L, Liao X, Zhou Y, Weng L, Xiao X, Zhou L, Wang X, Jiang Y, Yang Q, Zhu Y, Zhou L, Zhang W, Wang J, Yan X, Tang B, Shen L. Performance of Plasma Amyloid β, Total Tau, and Neurofilament Light Chain in the Identification of Probable Alzheimer's Disease in South China. Front Aging Neurosci 2021; 13:749649. [PMID: 34776933 PMCID: PMC8579066 DOI: 10.3389/fnagi.2021.749649] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/24/2021] [Indexed: 12/31/2022] Open
Abstract
Background: Alzheimer's disease (AD) is the most common type of dementia and has no effective treatment to date. It is essential to develop a minimally invasive blood-based biomarker as a tool for screening the general population, but the efficacy remains controversial. This cross-sectional study aimed to evaluate the ability of plasma biomarkers, including amyloid β (Aβ), total tau (t-tau), and neurofilament light chain (NfL), to detect probable AD in the South Chinese population. Methods: A total of 277 patients with a clinical diagnosis of probable AD and 153 healthy controls with normal cognitive function (CN) were enrolled in this study. The levels of plasma Aβ42, Aβ40, t-tau, and NfL were detected using ultra-sensitive immune-based assays (SIMOA). Lumbar puncture was conducted in 89 patients with AD to detect Aβ42, Aβ40, t-tau, and phosphorylated (p)-tau levels in the cerebrospinal fluid (CSF) and to evaluate the consistency between plasma and CSF biomarkers through correlation analysis. Finally, the diagnostic value of plasma biomarkers was further assessed by constructing a receiver operating characteristic (ROC) curve. Results: After adjusting for age, sex, and the apolipoprotein E (APOE) alleles, compared to the CN group, the plasma t-tau, and NfL were significantly increased in the AD group (p < 0.01, Bonferroni correction). Correlation analysis showed that only the plasma t-tau level was positively correlated with the CSF t-tau levels (r = 0.319, p = 0.003). The diagnostic model combining plasma t-tau and NfL levels, and age, sex, and APOE alleles, showed the best performance for the identification of probable AD [area under the curve (AUC) = 0.89, sensitivity = 82.31%, specificity = 83.66%]. Conclusion: Blood biomarkers can effectively distinguish patients with probable AD from controls and may be a non-invasive and efficient method for AD pre-screening.
Collapse
Affiliation(s)
- Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Hui Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lina Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xinxin Liao
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yafang Zhou
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Ling Weng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Xuewen Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yaling Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qijie Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Zhou
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Weiwei Zhang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Xinxiang Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| |
Collapse
|
35
|
Petrozziello T, Amaral AC, Dujardin S, Farhan SMK, Chan J, Trombetta BA, Kivisäkk P, Mills AN, Bordt EA, Kim SE, Dooley PM, Commins C, Connors TR, Oakley DH, Ghosal A, Gomez-Isla T, Hyman BT, Arnold SE, Spires-Jones T, Cudkowicz ME, Berry JD, Sadri-Vakili G. Novel genetic variants in MAPT and alterations in tau phosphorylation in amyotrophic lateral sclerosis post-mortem motor cortex and cerebrospinal fluid. Brain Pathol 2021; 32:e13035. [PMID: 34779076 PMCID: PMC8877756 DOI: 10.1111/bpa.13035] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/22/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Although the molecular mechanisms underlying amyotrophic lateral sclerosis (ALS) are not yet fully understood, several studies report alterations in tau phosphorylation in both sporadic and familial ALS. Recently, we have demonstrated that phosphorylated tau at S396 (pTau‐S396) is mislocalized to synapses in ALS motor cortex (mCTX) and contributes to mitochondrial dysfunction. Here, we demonstrate that while there was no overall increase in total tau, pTau‐S396, and pTau‐S404 in ALS post‐mortem mCTX, total tau and pTau‐S396 were increased in C9ORF72‐ALS. Additionally, there was a significant decrease in pTau‐T181 in ALS mCTX compared controls. Furthermore, we leveraged the ALS Knowledge Portal and Project MinE data sets and identified ALS‐specific genetic variants across MAPT, the gene encoding tau. Lastly, assessment of cerebrospinal fluid (CSF) samples revealed a significant increase in total tau levels in bulbar‐onset ALS together with a decrease in CSF pTau‐T181:tau ratio in all ALS samples, as reported previously. While increases in CSF tau levels correlated with a faster disease progression as measured by the revised ALS functional rating scale (ALSFRS‐R), decreases in CSF pTau‐T181:tau ratio correlated with a slower disease progression, suggesting that CSF total tau and pTau‐T181 ratio may serve as biomarkers of disease in ALS. Our findings highlight the potential role of pTau‐T181 in ALS, as decreases in CSF pTau‐T181:tau ratio may reflect the significant decrease in pTau‐T181 in post‐mortem mCTX. Taken together, these results indicate that tau phosphorylation is altered in ALS post‐mortem mCTX as well as in CSF and, importantly, the newly described pathogenic or likely pathogenic variants identified in MAPT in this study are adjacent to T181 and S396 phosphorylation sites further highlighting the potential role of these tau functional domains in ALS. Although the molecular mechanisms underlying amyotrophic lateral sclerosis (ALS) are not yet fully understood, recent studies report alterations in tau phosphorylation in ALS. Our study builds on these findings and demonstrates that tau phosphorylation is altered in post‐mortem ALS motor cortex and highlights new and ALS‐specific variants in MAPT, the gene encoding tau. Lastly, we report alterations in phosphorylated tau in ALS cerebrospinal fluid that may function as a predictive biomarker for ALS.![]()
Collapse
Affiliation(s)
- Tiziana Petrozziello
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ana C Amaral
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Simon Dujardin
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sali M K Farhan
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - James Chan
- Biostatistics Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bianca A Trombetta
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Pia Kivisäkk
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexandra N Mills
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Evan A Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Spencer E Kim
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Patrick M Dooley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Caitlin Commins
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Theresa R Connors
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Derek H Oakley
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anubrata Ghosal
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Teresa Gomez-Isla
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Steven E Arnold
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tara Spires-Jones
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, University of Edinburgh, UK
| | - Merit E Cudkowicz
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - James D Berry
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ghazaleh Sadri-Vakili
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
36
|
Tsai HH, Chen YF, Yen RF, Lo YL, Yang KC, Jeng JS, Tsai LK, Chang CF. Plasma soluble TREM2 is associated with white matter lesions independent of amyloid and tau. Brain 2021; 144:3371-3380. [PMID: 34515756 DOI: 10.1093/brain/awab332] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/21/2021] [Accepted: 08/08/2021] [Indexed: 12/20/2022] Open
Abstract
Cerebral small vessel disease is one of the most common causes of cognitive decline and stroke. While several lines of evidence have established a relationship between inflammation and cerebrovascular pathology, the mechanistic link has not yet been elucidated. Recent studies suggest activation of immune mediators, including the soluble form of triggering receptor expressed on myeloid cells 2 (TREM2), may be critical regulators. In this study, we compared the plasma levels of soluble TREM2 and its correlations with neuroimaging markers and cerebral amyloid load in ten patients with Alzheimer's disease and 66 survivors of spontaneous intracerebral haemorrhage with cerebral amyloid angiopathy or hypertensive small vessel disease, two of the most common types of sporadic small vessel disease. We performed brain MRI and 11C-Pittsburgh compound B PET for all participants to evaluate radiological small vessel disease markers and cerebral amyloid burden, and 18F-T807 PET in a subgroup of patients to evaluate cortical tau pathology. Plasma soluble TREM2 levels were comparable between patients with Alzheimer's disease and small vessel disease (P=0.690). In patients with small vessel disease, plasma soluble TREM2 was significantly associated with white matter hyperintensity volume (P<0.001), but not with cerebral amyloid load. Among patients with Alzheimer's disease and cerebral amyloid angiopathy, plasma soluble TREM2 was independently associated with a tau-positive scan (P=0.001) and white matter hyperintensity volume (P=0.013), but not amyloid load (P=0.221). Our results indicate plasma soluble TREM2 is associated with white matter hyperintensity independent of amyloid and tau pathology. These findings highlight the potential utility of plasma soluble TREM2 as a strong predictive marker for small vessel disease-related white matter injury and hold clinical implications for targeting the innate immune response when treating this disease.
Collapse
Affiliation(s)
- Hsin-Hsi Tsai
- Department of Neurology, National Taiwan University Hospital Beihu Branch, Taipei, Taiwan.,Department of Neurology, 3Medical Imaging, and 4Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ya-Fang Chen
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ruoh-Fang Yen
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yen-Ling Lo
- Department of Neurology, National Taiwan University Hospital Beihu Branch, Taipei, Taiwan
| | - Kai-Chien Yang
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jiann-Shing Jeng
- Department of Neurology, 3Medical Imaging, and 4Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Kai Tsai
- Department of Neurology, 3Medical Imaging, and 4Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Che-Feng Chang
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
37
|
Robbins M, Clayton E, Kaminski Schierle GS. Synaptic tau: A pathological or physiological phenomenon? Acta Neuropathol Commun 2021; 9:149. [PMID: 34503576 PMCID: PMC8428049 DOI: 10.1186/s40478-021-01246-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/17/2022] Open
Abstract
In this review, we discuss the synaptic aspects of Tau pathology occurring during Alzheimer's disease (AD) and how this may relate to memory impairment, a major hallmark of AD. Whilst the clinical diagnosis of AD patients is a loss of working memory and long-term declarative memory, the histological diagnosis is the presence of neurofibrillary tangles of hyperphosphorylated Tau and Amyloid-beta plaques. Tau pathology spreads through synaptically connected neurons to impair synaptic function preceding the formation of neurofibrillary tangles, synaptic loss, axonal retraction and cell death. Alongside synaptic pathology, recent data suggest that Tau has physiological roles in the pre- or post- synaptic compartments. Thus, we have seen a shift in the research focus from Tau as a microtubule-stabilising protein in axons, to Tau as a synaptic protein with roles in accelerating spine formation, dendritic elongation, and in synaptic plasticity coordinating memory pathways. We collate here the myriad of emerging interactions and physiological roles of synaptic Tau, and discuss the current evidence that synaptic Tau contributes to pathology in AD.
Collapse
|
38
|
Castillo-Mendieta T, Arana-Lechuga Y, Campos-Peña V, Sosa AL, Orozco-Suarez S, Pinto-Almazán R, Segura-Uribe J, Javier Rodríguez-Sánchez de Tagle A, Ruiz-Sánchez E, Guerra-Araiza C. Plasma Levels of Amyloid-β Peptides and Tau Protein in Mexican Patients with Alzheimer's Disease. J Alzheimers Dis 2021; 82:S271-S281. [PMID: 34151786 DOI: 10.3233/jad-200912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) causes memory deficit and alterations in other cognitive functions, mainly in adults over 60 years of age. As the diagnosis confirmation is performed by a postmortem neuropathological examination of the brain, this disease can be confused with other types of dementia at early stages. About 860,000 Mexicans are affected by dementia, most of them with insufficient access to adequate comprehensive health care services. Plasma biomarkers could be a rapid option for early diagnosis of the disease. OBJECTIVE This study aimed to analyze some plasma biomarkers (amyloid-β, tau, and lipids) in Mexican AD patients and control subjects with no associated neurodegenerative diseases. METHODS Plasma amyloid-β peptides (Aβ40 and Aβ42), total and phosphorylated tau protein (T-tau and P-tau), and cholesterol and triglyceride levels were quantified by enzyme-linked immunosorbent assay in AD patients and control subjects. RESULTS In Mexican AD patients, we found significantly lower levels of Aβ42 (p < 0.05) compared to the control group. In contrast, significantly higher levels of P-tau (p < 0.05) and triglycerides (p < 0.05) were observed in AD patients compared to controls. Furthermore, a significant correlation was found between the severity of dementia and plasma P-tau levels, Aβ42/Aβ40 and P-tau/T-tau ratios, and triglycerides concentrations. This correlation increased gradually with cognitive decline. CONCLUSION The detection of these plasma biomarkers is an initial step in searching for a timely, less invasive, and cost-efficient diagnosis in Mexicans.
Collapse
Affiliation(s)
- Tzayaka Castillo-Mendieta
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico.,Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Yoaly Arana-Lechuga
- Sleep Disorders Clinic, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Victoria Campos-Peña
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| | - Ana Luisa Sosa
- Clínica de Demencia, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| | - Sandra Orozco-Suarez
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Rodolfo Pinto-Almazán
- Laboratorio de Biología Molecular en Enfermedades Metabólicas y Neurodegenerativas, Unidad de Investigación, Hospital Regional de Alta Especialidad de Ixtapaluca, Ixtapaluca, State of Mexico, Mexico
| | - Julia Segura-Uribe
- Subdirección de Gestión de la Investigación, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City, Mexico
| | - Aldo Javier Rodríguez-Sánchez de Tagle
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico.,Coordinación de QFBT, Universidad del Valle de México-Chapultepec, México City, México
| | - Elizabeth Ruiz-Sánchez
- Laboratorio de Neurotoxicología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| | - Christian Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| |
Collapse
|
39
|
Fong TG, Vasunilashorn SM, Ngo L, Libermann TA, Dillon ST, Schmitt EM, Pascual-Leone A, Arnold SE, Jones RN, Marcantonio ER, Inouye SK. Association of Plasma Neurofilament Light with Postoperative Delirium. Ann Neurol 2020; 88:984-994. [PMID: 32881052 DOI: 10.1002/ana.25889] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To examine the association of the plasma neuroaxonal injury markers neurofilament light (NfL), total tau, glial fibrillary acid protein, and ubiquitin carboxyl-terminal hydrolase L1 with delirium, delirium severity, and cognitive performance. METHODS Delirium case-no delirium control (n = 108) pairs were matched by age, sex, surgery type, cognition, and vascular comorbidities. Biomarkers were measured in plasma collected preoperatively (PREOP), and 2 days (POD2) and 30 days postoperatively (PO1MO) using Simoa technology (Quanterix, Lexington, MA). The Confusion Assessment Method (CAM) and CAM-S (Severity) were used to measure delirium and delirium severity, respectively. Cognitive function was measured with General Cognitive Performance (GCP) scores. RESULTS Delirium cases had higher NfL on POD2 and PO1MO (median matched pair difference = 16.2pg/ml and 13.6pg/ml, respectively; p < 0.05). Patients with PREOP and POD2 NfL in the highest quartile (Q4) had increased risk for incident delirium (adjusted odds ratio [OR] = 3.7 [95% confidence interval (CI) = 1.1-12.6] and 4.6 [95% CI = 1.2-18.2], respectively) and experienced more severe delirium, with sum CAM-S scores 7.8 points (95% CI = 1.6-14.0) and 9.3 points higher (95% CI = 3.2-15.5). At PO1MO, delirium cases had continued high NfL (adjusted OR = 9.7, 95% CI = 2.3-41.4), and those with Q4 NfL values showed a -2.3 point decline in GCP score (-2.3 points, 95% CI = -4.7 to -0.9). INTERPRETATION Patients with the highest PREOP or POD2 NfL levels were more likely to develop delirium. Elevated NfL at PO1MO was associated with delirium and greater cognitive decline. These findings suggest NfL may be useful as a predictive biomarker for delirium risk and long-term cognitive decline, and once confirmed would provide pathophysiological evidence for neuroaxonal injury following delirium. ANN NEUROL 2020;88:984-994.
Collapse
Affiliation(s)
- Tamara G Fong
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.,Aging Brain Center, Hebrew SeniorLife, Boston, Massachusetts, USA.,Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
| | - Sarinnapha M Vasunilashorn
- Division of General Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.,Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Long Ngo
- Division of General Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.,Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Towia A Libermann
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.,Beth Israel Deaconess Medical Center Genomics, Proteomics, Bioinformatics, and Systems Biology Center, Boston, Massachusetts, USA
| | - Simon T Dillon
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.,Beth Israel Deaconess Medical Center Genomics, Proteomics, Bioinformatics, and Systems Biology Center, Boston, Massachusetts, USA
| | - Eva M Schmitt
- Aging Brain Center, Hebrew SeniorLife, Boston, Massachusetts, USA.,Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
| | - Alvaro Pascual-Leone
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Guttmann Brain Health Institute, Guttmann Institute, Autonomous University of Barcelona, Barcelona, Spain
| | - Steven E Arnold
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Richard N Jones
- Departments of Psychiatry and Human Behavior and Neurology, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Edward R Marcantonio
- Division of General Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.,Division of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Sharon K Inouye
- Aging Brain Center, Hebrew SeniorLife, Boston, Massachusetts, USA.,Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA.,Division of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
40
|
Eren E, Hunt JFV, Shardell M, Chawla S, Tran J, Gu J, Vogt NM, Johnson SC, Bendlin BB, Kapogiannis D. Extracellular vesicle biomarkers of Alzheimer's disease associated with sub-clinical cognitive decline in late middle age. Alzheimers Dement 2020; 16:1293-1304. [PMID: 32588967 PMCID: PMC7984100 DOI: 10.1002/alz.12130] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/08/2020] [Accepted: 05/17/2020] [Indexed: 12/12/2022]
Abstract
Introduction Neuronal extracellular vesicle (nEV) tau and insulin signaling biomarkers may detect preclinical Alzheimer's disease and age‐associated cognitive decline. Methods This case‐control study used repeated serum samples from 73 cognitively declining and 73 stable Wisconsin Registry for Alzheimer's Prevention participants (62.4 ± 6.3 years old). We immunocaptured nEVs; measured tau and insulin signaling biomarkers; and examined biomarker differences by group, their performance in group classification in training and test datasets (97, 49 individuals, respectively), and whether they predict cognitive performance change. Results Declining compared to stable individuals showed higher baseline total, p231‐, and p181‐tau with older age and higher annualized change for p‐IR and p‐IGF‐1R. Combining biomarkers classified decliners with 94% area under the curve (AUC), 86.0% sensitivity and 86.7% specificity, in training data, and 75% AUC, 71.4% sensitivity, and 77.3% specificity, in test data. Insulin biomarkers predicted cognitive performance change prospectively. Discussion Combining nEV biomarkers can identify individuals with age‐associated cognitive decline.
Collapse
Affiliation(s)
- Erden Eren
- Laboratory of Clinical InvestigationIntramural Research ProgramNational Institute on AgingNIHBaltimoreMarylandUSA
| | - Jack F. V. Hunt
- Wisconsin Alzheimer's Disease Research CenterUniversity of WisconsinMadisonWisconsinUSA
| | | | - Sahil Chawla
- Laboratory of Clinical InvestigationIntramural Research ProgramNational Institute on AgingNIHBaltimoreMarylandUSA
| | - Joyce Tran
- Laboratory of Clinical InvestigationIntramural Research ProgramNational Institute on AgingNIHBaltimoreMarylandUSA
| | - Jeffrey Gu
- Laboratory of Clinical InvestigationIntramural Research ProgramNational Institute on AgingNIHBaltimoreMarylandUSA
| | - Nick M. Vogt
- Wisconsin Alzheimer's Disease Research CenterUniversity of WisconsinMadisonWisconsinUSA
| | - Sterling C. Johnson
- Wisconsin Alzheimer's Disease Research CenterUniversity of WisconsinMadisonWisconsinUSA
- Geriatric Research Education and Clinical Center of the Wm. S. MiddletonMemorial Veterans HospitalMadisonWisconsinUSA
| | - Barbara B. Bendlin
- Wisconsin Alzheimer's Disease Research CenterUniversity of WisconsinMadisonWisconsinUSA
| | - Dimitrios Kapogiannis
- Laboratory of Clinical InvestigationIntramural Research ProgramNational Institute on AgingNIHBaltimoreMarylandUSA
| |
Collapse
|
41
|
Pase MP, Beiser AS, Himali JJ, Satizabal CL, Aparicio HJ, DeCarli C, Chêne G, Dufouil C, Seshadri S. Assessment of Plasma Total Tau Level as a Predictive Biomarker for Dementia and Related Endophenotypes. JAMA Neurol 2020; 76:598-606. [PMID: 30830207 DOI: 10.1001/jamaneurol.2018.4666] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Importance Blood-based biomarkers have the potential to improve the identification of persons with the greatest dementia risk for inclusion in dementia prevention trials through low-cost and minimally invasive screening. Objective To investigate the use of plasma total tau as a blood biomarker for dementia and related endophenotypes. Design, Setting, and Participants This prospective cohort study used data from the US community-based Framingham Heart Study with replication in the Memento study, a multicenter cohort of persons with mild cognitive impairment or subjective cognitive complaints recruited from memory clinics across France. Total tau levels were measured from stored plasma samples in Framingham Heart Study participants during 2004 to 2011. Dementia follow-up occurred across a median of 6 years (interquartile range, 5-8 years) for persons 65 years and older who were dementia free at baseline. Plasma and/or cerebrospinal fluid samples were obtained from Memento study participants from April 19, 2011, to June 22, 2016. Dementia follow-up took place over a median of 4 years (interquartile range, 3-5 years). Data analysis was performed from January to November 2018. Exposures Plasma total tau level measured using single-molecule array technology. Main Outcomes and Measures Incidence of dementia of any cause (all dementia) and dementia due to clinical Alzheimer disease (AD dementia). Results Among the 1453 participants in the Framingham dementia study sample, the mean (SD) age was 75 (7) years; 792 (54.5%) were female. Among the 367 individuals in the replication cohort, the mean (SD) age was 69 (9) years; 217 (59.1%) were female. Of 134 cases of incident all dementia in the Framingham sample, 105 were AD dementia. After adjustment for age and sex, each SD unit increase in the log of plasma total tau level was associated with a 35% increase in AD dementia risk (hazard ratio [HR], 1.35; 95% CI, 1.10-1.67). The addition of plasma total tau to a model including age and sex improved the stratification of participants for risk of AD dementia (net reclassification improvement, 0.382; 95% CI, 0.030-0.716). Higher plasma total tau level was associated with poorer cognition across 7 cognitive tasks (P < .05) and smaller hippocampi (hippocampal volume: β [SE] = 0.002 [0.001]; P = .003) as well as neurofibrillary tangles (β [SE] = 0.95 [0.45]; P = .04) and microinfarcts (odds ratio, 3.04; 95% CI, 1.26-7.37) at autopsy. In the replication cohort, plasma total tau level weakly correlated with cerebrospinal fluid total tau level (Spearman correlation coefficient, 0.16; P = .07), but plasma total tau was at least as strongly associated with incident AD dementia as cerebrospinal fluid total tau (log plasma total tau: HR, 2.33; 95% CI, 1.00-5.48; log cerebrospinal fluid total tau: HR, 2.14; 95% CI, 1.33-3.44) after adjustment for age and sex. Conclusions and Relevance The findings suggest that plasma total tau levels may improve the prediction of future dementia, are associated with dementia endophenotypes, and may be used as a biomarker for risk stratification in dementia prevention trials.
Collapse
Affiliation(s)
- Matthew P Pase
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts.,Framingham Heart Study, Framingham, Massachusetts.,Centre for Human Psychopharmacology, Swinburne University of Technology, Hawthorn, Australia.,Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health and The University of Melbourne, Melbourne, Australia
| | - Alexa S Beiser
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts.,Framingham Heart Study, Framingham, Massachusetts.,Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Jayandra J Himali
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts.,Framingham Heart Study, Framingham, Massachusetts.,Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Claudia L Satizabal
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts.,Framingham Heart Study, Framingham, Massachusetts.,Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, Texas
| | - Hugo J Aparicio
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts.,Framingham Heart Study, Framingham, Massachusetts
| | - Charles DeCarli
- Framingham Heart Study, Framingham, Massachusetts.,Department of Neurology, School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California Davis, Sacramento, California
| | - Geneviève Chêne
- Inserm Unit 1219 Bordeaux Population Health, CIC 1401-EC (Clinical Epidemiology), University of Bordeaux, ISPED (Bordeaux School of Public Health), Bordeaux University Hospital, Bordeaux, France
| | - Carole Dufouil
- Inserm Unit 1219 Bordeaux Population Health, CIC 1401-EC (Clinical Epidemiology), University of Bordeaux, ISPED (Bordeaux School of Public Health), Bordeaux University Hospital, Bordeaux, France
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts.,Framingham Heart Study, Framingham, Massachusetts.,Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, Texas
| |
Collapse
|
42
|
Tvarijonaviciute A, Zamora C, Ceron JJ, Bravo-Cantero AF, Pardo-Marin L, Valverde S, Lopez-Jornet P. Salivary biomarkers in Alzheimer's disease. Clin Oral Investig 2020; 24:3437-3444. [PMID: 31989369 DOI: 10.1007/s00784-020-03214-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/15/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVES The objective of this study was to evaluate the changes that can occur in saliva components in patients with Alzheimer's disease (AD) of different severity and determine if any of these components could be a biomarker of this disease. Therefore, a panel of selected analytes related to the amyloid cascade, the immune and adrenergic systems, among others, were analyzed in the saliva of patients with Alzheimer's disease. METHODS A total of 152 patients with AD and controls were included. The severity of the disease was established according to the Global Deterioration Scale. Unstimulated whole saliva was collected. RESULTS Salivary amyloid-β42 was significantly lower, and complement C4 was significantly higher in the patients with AD than in the controls (p < 0.05 in both cases). Only complement C4 maintained its significant effect in the multivariate regression analysis. However, the area under the receiver operating characteristic curve of C4 was 0.613. No changes were found in any analyte regarding the severity of the disease. CONCLUSIONS A decrease in amyloid-β42 and an increase in complement C4 were detected in the saliva of patients with AD, but the changes did not show a high diagnostic performance for the detection of AD and were not associated with its severity. CLINICAL RELEVANCE Although some analytes showed significant differences in saliva in patients with AD, in our study conditions the salivary biomarkers analyzed were not of enough diagnostic utility for being used in routine.
Collapse
Affiliation(s)
- Asta Tvarijonaviciute
- Interdisciplinary Laboratory of Clinical Analysis, Interlab-UMU, Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Espinardo, 30100, Murcia, Spain.
| | - Carmen Zamora
- Department of Oral Medicine, Faculty of Medicine, Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Espinardo, 30100, Murcia, Spain
| | - Jose J Ceron
- Interdisciplinary Laboratory of Clinical Analysis, Interlab-UMU, Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Espinardo, 30100, Murcia, Spain
| | | | - Luis Pardo-Marin
- Interdisciplinary Laboratory of Clinical Analysis, Interlab-UMU, Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Espinardo, 30100, Murcia, Spain
| | - Sandra Valverde
- Interdisciplinary Laboratory of Clinical Analysis, Interlab-UMU, Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Espinardo, 30100, Murcia, Spain
| | - Pia Lopez-Jornet
- Department of Oral Medicine, Faculty of Medicine, Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Espinardo, 30100, Murcia, Spain
| |
Collapse
|
43
|
Mayaki AM, Abdul Razak IS, Noraniza MA, Mazlina M, Rasedee A. Biofluid Markers of Equine Neurological Disorders Reviewed From Human Perspectives. J Equine Vet Sci 2019; 86:102907. [PMID: 32067661 DOI: 10.1016/j.jevs.2019.102907] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 12/26/2019] [Accepted: 12/26/2019] [Indexed: 02/07/2023]
Abstract
Neurological disorders (NDs) are often fatal to horses. Thus, symptoms of equine NDs commonly indicate euthanasia. Current diagnostic approaches for equine NDs is based on clinical signs, differential diagnoses, analysis of cerebrospinal fluid (CSF), assessment of histopathological lesions, and imaging. However, advances in biofluid biomarkers in the diagnosis of human neurological diseases can potentially be applied to equine NDs. In this review, we described the established human blood and CSF neurobiomarkers that could potentially be used to diagnose equine NDs.
Collapse
Affiliation(s)
- Abubakar Musa Mayaki
- Department of Veterinary Pre-Clinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Intan Shameha Abdul Razak
- Department of Veterinary Pre-Clinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
| | - Mohd Adzahan Noraniza
- Department of Farm and Exotic Animal Medicine and Surgery, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Mazlan Mazlina
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Abdullah Rasedee
- Department of Veterinary Laboratory Diagnosis, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
44
|
Milà-Alomà M, Suárez-Calvet M, Molinuevo JL. Latest advances in cerebrospinal fluid and blood biomarkers of Alzheimer's disease. Ther Adv Neurol Disord 2019; 12:1756286419888819. [PMID: 31897088 PMCID: PMC6920596 DOI: 10.1177/1756286419888819] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and its diagnosis has classically been based on clinical symptoms. Recently, a biological rather than a syndromic definition of the disease has been proposed that is based on biomarkers that reflect neuropathological changes. In AD, there are two main biomarker categories, namely neuroimaging and fluid biomarkers [cerebrospinal fluid (CSF) and blood]. As a complex and multifactorial disease, AD biomarkers are important for an accurate diagnosis and to stage the disease, assess the prognosis, test target engagement, and measure the response to treatment. In addition, biomarkers provide us with information that, even if it does not have a current clinical use, helps us to understand the mechanisms of the disease. In addition to the pathological hallmarks of AD, which include amyloid-β and tau deposition, there are multiple concomitant pathological events that play a key role in the disease. These include, but are not limited to, neurodegeneration, inflammation, vascular dysregulation or synaptic dysfunction. In addition, AD patients often have an accumulation of other proteins including α-synuclein and TDP-43, which may have a pathogenic effect on AD. In combination, there is a need to have biomarkers that reflect different aspects of AD pathogenesis and this will be important in the future to establish what are the most suitable applications for each of these AD-related biomarkers. It is unclear whether sex, gender, or both have an effect on the causes of AD. There may be differences in fluid biomarkers due to sex but this issue has often been neglected and warrants further research. In this review, we summarize the current state of the principal AD fluid biomarkers and discuss the effect of sex on these biomarkers.
Collapse
Affiliation(s)
- Marta Milà-Alomà
- Barcelonaβeta Brain Research Center (BBRC),
Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research
Institute), Barcelona
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC),
Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research
Institute), Barcelona
- Department of Neurology, Hospital del Mar,
Barcelona
| | - José Luís Molinuevo
- Scientific Director, Alzheimer’s Prevention
Program, Barcelonaβeta Brain Research Center, Wellington 30, Barcelona,
08005, Spain
- IMIM (Hospital del Mar Medical Research
Institute), Barcelona
- CIBER Fragilidad y Envejecimiento Saludable,
Madrid, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
45
|
Bittar A, Bhatt N, Kayed R. Advances and considerations in AD tau-targeted immunotherapy. Neurobiol Dis 2019; 134:104707. [PMID: 31841678 PMCID: PMC6980703 DOI: 10.1016/j.nbd.2019.104707] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/27/2019] [Accepted: 12/08/2019] [Indexed: 12/18/2022] Open
Abstract
The multifactorial and complex nature of Alzheimer’s disease (AD) has made it difficult to identify therapeutic targets that are causally involved in the disease process. However, accumulating evidence from experimental and clinical studies that investigate the early disease process point towards the required role of tau in AD etiology. Importantly, a large number of studies investigate and characterize the plethora of pathological forms of tau protein involved in disease onset and propagation. Immunotherapy is one of the most clinical approaches anticipated to make a difference in the field of AD therapeutics. Tau –targeted immunotherapy is the new direction after the failure of amyloid beta (Aß)-targeted immunotherapy and the growing number of studies that highlight the Aß-independent disease process. It is now well established that immunotherapy alone will most likely be insufficient as a monotherapy. Therefore, this review discusses updates on tau-targeted immunotherapy studies, AD-relevant tau species, updates on promising biomarkers and a prospect on combination therapies to surround the disease propagation in an efficient and timely manner.
Collapse
Affiliation(s)
- Alice Bittar
- Department of Neurology, The Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States of America.
| | - Nemil Bhatt
- Department of Neuroscience, Cell Biology and Anatomy, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States of America.
| | - Rakez Kayed
- Department of Neurology, The Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States of America.
| |
Collapse
|
46
|
Mohammed RS, Ibrahim W, Sabry D, El-Jaafary SI. Occupational metals exposure and cognitive performance among foundry workers using tau protein as a biomarker. Neurotoxicology 2019; 76:10-16. [PMID: 31593711 DOI: 10.1016/j.neuro.2019.09.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/14/2019] [Accepted: 09/30/2019] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Human exposure to heavy metals is a potential risk for developing cognitive impairment. Aluminum (Al) foundry is one of industries that involve occupational exposure to different metals. AIM OF THE WORK to evaluate the cognitive performance of Aluminum foundry workers in relation to different metals exposure. MATERIALS AND METHODS a cross sectional study conducted on 75 Al foundry workers and 75 non-occupationally exposed subjects as controls. Personal interview with specially designed questionnaire, Assessment of cognitive functions done using Montreal cognitive assessment (MocA), Stress, depression and sleep were also assessed. Serum levels of Aluminum (AL), Lead (Pb), manganese (Mn), Zinc (Zn) and tau protein were measured. RESULTS Exposed group showed significant increase in serum levels of Aluminum, lead, Manganese and tau protein, p value < 0.005 (mean ± SD 0.56 ± 0.18, 22.3 ± 5.01, 42.04 ± 7.4, 1.53 ± 0.58 Vs 0.36 ± 0.11, 13.4 ± 1.29, 39.4 ± 4.4, 1.03 ± 0.44 respectively) with significant decrease of zinc level compared to control (mean ± SD 46.4 ± 5.2 Vs 88.8 ± 6.04, p value 0.005). There was a significant decrease MocA scores among exposed population, (mean ± SD 24.4 ± 3.4 compared to 28.4 ± 1.3 in non exposed, p value < 0.005). which was affected by serum levels of lead, aluminum, manganese and tau protein (β -0.165, -8.958, -.286, -2.341 respectively and p < 0.005).Stress scores was higher in exposed workers than control but not affecting cognitive performance. CONCLUSION occupational exposure to metals can cause cognitive dysfunction which may be subtle, so there is a need for formal cognitive testing at baseline, and on regular intervals during working period. Serum tau protein could be used as a prognostic biomarker for the hazardous effect of occupational exposure to these metals on the neuronal cells.
Collapse
Affiliation(s)
- Rateba S Mohammed
- Occupational and Environmental Medicine Department, Faculty of Medicine, Cairo University Hospitals, Kasralainy street, Cairo, Egypt.
| | - Walaa Ibrahim
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University Hospitals, Kasralainy street, Cairo, Egypt.
| | - Dina Sabry
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University Hospitals, Kasralainy street, Cairo, Egypt.
| | | |
Collapse
|
47
|
Pase MP, Himali JJ, Aparicio HJ, Romero JR, Satizabal CL, Maillard P, DeCarli C, Beiser AS, Seshadri S. Plasma total-tau as a biomarker of stroke risk in the community. Ann Neurol 2019; 86:463-467. [PMID: 31271449 PMCID: PMC7297542 DOI: 10.1002/ana.25542] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/23/2019] [Accepted: 06/26/2019] [Indexed: 11/10/2022]
Abstract
Higher plasma total-tau level is associated with incident dementia, but its relationship with stroke risk is unknown. In this prospective community-based study, we evaluated plasma total-tau level as a biomarker of stroke risk in 2,794 Framingham Heart Study participants. Persons with plasma total-tau levels in the top quintile, versus the bottom 4, had an increased risk of incident stroke over a mean follow-up of 8.3 years (hazard ratio = 2.01; 95% confidence interval = 1.32-3.08) following adjustments for age, sex, and stroke risk factors. Our findings demonstrate that plasma total-tau relates to the risk of stroke in a community sample. ANN NEUROL 2019;86:463-467.
Collapse
Affiliation(s)
- Matthew P Pase
- Melbourne Dementia Research Centre, Florey Institute for Neuroscience and Mental Health and University of Melbourne, Melbourne, Australia
- Framingham Heart Study, Framingham, MA
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, Australia
| | - Jayandra J Himali
- Framingham Heart Study, Framingham, MA
- Department of Neurology, Boston University School of Medicine, Boston, MA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Hugo J Aparicio
- Framingham Heart Study, Framingham, MA
- Department of Neurology, Boston University School of Medicine, Boston, MA
| | | | - Claudia L Satizabal
- Framingham Heart Study, Framingham, MA
- Department of Neurology, Boston University School of Medicine, Boston, MA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX
| | - Pauline Maillard
- Department of Neurology, School of Medicine and Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California, Davis, Sacramento, CA
| | - Charles DeCarli
- Department of Neurology, School of Medicine and Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California, Davis, Sacramento, CA
| | - Alexa S Beiser
- Framingham Heart Study, Framingham, MA
- Department of Neurology, Boston University School of Medicine, Boston, MA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Sudha Seshadri
- Framingham Heart Study, Framingham, MA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX
| |
Collapse
|
48
|
|
49
|
Reciprocal Predictive Relationships between Amyloid and Tau Biomarkers in Alzheimer's Disease Progression: An Empirical Model. J Neurosci 2019; 39:7428-7437. [PMID: 31350262 DOI: 10.1523/jneurosci.1056-19.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/01/2019] [Accepted: 07/19/2019] [Indexed: 01/09/2023] Open
Abstract
There is an urgent need to understand the relationships between amyloid-β (Aβ) and tau in the progression of Alzheimer's disease to identify treatment targets. Here we examine reciprocal predictions of brain Aβ burden quantified by positron emission tomography and CSF concentrations of Aβ42 and phosphorylated tau (p-tau). Each biomarker was examined over 48 months in two separate cross-lagged models; one in asymptomatic healthy elderly people (men and women), and one in patients with Alzheimer's disease (AD) dementia or mild cognitive impairment (MCI). The models examine predictions of each biomarker on the progression of the others, considering each previous and concurrent measure. In healthy elderly, lower CSF Aβ42 predicted Aβ deposition and reciprocally, Aβ burden predicted a decrease in CSF Aβ42. Lower CSF Aβ42 predicted an increase in CSF p-tau, and CSF p-tau predicted Aβ deposition. In AD/MCI, lower CSF Aβ42 predicted Aβ deposition and Aβ burden reciprocally predicted CSF Aβ42 changes; however, in contrast to healthy elderly, CSF p-tau concentrations did not predict Aβ biomarkers, or vice versa. In post hoc models examining cognitive status, CSF Aβ42 predicted Mini Mental State Examination (MMSE) scores in healthy elderly, whereas Aβ burden and CSF p-tau predicted MMSE scores in AD/MCI. The findings describe reciprocal predictions between Aβ and tau biomarkers in healthy elderly and they implicate mechanisms underlying low CSF Aβ42 in Alzheimer's disease pathogenesis and progression. In symptomatic Alzheimer's disease, CSF Aβ42 and Aβ deposition predicted each other; however, Aβ and CSF p-tau progressed independently and they independently predicted cognitive decline.SIGNIFICANCE STATEMENT This study offers empirical evidence concerning the hypothesized "amyloid cascade", as it progressed over 4 years in healthy elderly people and in Alzheimer's disease patients. In healthy elderly, CSF amyloid changes predicted amyloid deposition, CSF phosphorylated tau concentrations, and a decline in cognitive status. Phosphorylated tau concentrations specifically predicted amyloid deposition. In Alzheimer's disease patients, although amyloid deposition and CSF amyloid changes continued to "cascade", there was no evidence to suggest that amyloid and tau biomarkers predicted each other, although both amyloid deposition and CSF tau progression predicted cognitive decline independently. Taking advantage of repeated amyloid PET and CSF measures, this dynamic view offers new insight into the progression of Alzheimer's disease biomarkers and their relationships with cognitive decline.
Collapse
|
50
|
Teng IT, Li X, Yadikar HA, Yang Z, Li L, Lyu Y, Pan X, Wang KK, Tan W. Identification and Characterization of DNA Aptamers Specific for Phosphorylation Epitopes of Tau Protein. J Am Chem Soc 2018; 140:14314-14323. [PMID: 30277395 DOI: 10.1021/jacs.8b08645] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tau proteins are proteins that stabilize microtubules, but their hyperphosphorylation can result in the formation of protein aggregates and, over time, neurodegeneration. This phenomenon, termed tauopathy, is pathologically involved in several neurodegenerative disorders. DNA aptamers are single-stranded oligonucleotides capable of specific binding to target molecules. Using tau epitopes predisposed for phosphorylation, we identified six distinct aptamers that bind to tau at two phosphorylatable epitopes (Thr-231 and Ser-202) and to full-length Tau441 proteins with nanomolar affinity. In addition, several of these aptamers also inhibit tau phosphorylation (IT4, IT5, IT6) and tau oligomerization (IT3, IT4, IT5, IT6). This is the first report to identify tau epitope-specific aptamers. Such tau aptamers can be used to detect tau in biofluids and uncover the mechanism of tauopathy. They can be further developed into novel therapeutic agents in mitigating tauopathy-associated neurodegenerative disorders.
Collapse
Affiliation(s)
- I-Ting Teng
- Department of Chemistry, Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, UF Health Cancer Center , UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville , Florida 32611-7200 , United States
| | - Xiaowei Li
- Department of Chemistry, Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, UF Health Cancer Center , UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville , Florida 32611-7200 , United States
| | - Hamad Ahmad Yadikar
- Department of Emergency Medicine, Department of Chemistry, Department of Neuroscience, and Department of Psychiatry , McKnight Brain Institute, University of Florida , Gainesville , Florida 32611 , United States
| | - Zhihui Yang
- Department of Emergency Medicine, Department of Chemistry, Department of Neuroscience, and Department of Psychiatry , McKnight Brain Institute, University of Florida , Gainesville , Florida 32611 , United States
| | - Long Li
- Department of Chemistry, Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, UF Health Cancer Center , UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville , Florida 32611-7200 , United States
| | - Yifan Lyu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha 410082 , China
| | - Xiaoshu Pan
- Department of Chemistry, Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, UF Health Cancer Center , UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville , Florida 32611-7200 , United States
| | - Kevin K Wang
- Department of Emergency Medicine, Department of Chemistry, Department of Neuroscience, and Department of Psychiatry , McKnight Brain Institute, University of Florida , Gainesville , Florida 32611 , United States.,Brain Rehabilitation Research Center (BRRC) , Malcom Randall Veterans Affairs Medical Center , 1601 SW Archer Road , Gainesville Florida 32608 , United States
| | - Weihong Tan
- Department of Chemistry, Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, UF Health Cancer Center , UF Genetics Institute and McKnight Brain Institute, University of Florida , Gainesville , Florida 32611-7200 , United States.,Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province , Hunan University , Changsha 410082 , China
| |
Collapse
|