101
|
Chen Y, Zeng X, Lee J, Sehrawat A, Lafferty TK, Boslett JJ, Klunk WE, Pascoal TA, Villemagne VL, Cohen AD, Lopez O, Yates NA, Karikari TK. Effect of blood collection tube containing protease inhibitors on the pre-analytical stability of Alzheimer's disease plasma biomarkers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.05.24303504. [PMID: 38496591 PMCID: PMC10942510 DOI: 10.1101/2024.03.05.24303504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
INTRODUCTION The reliability of plasma Alzheimer's disease (AD) biomarkers can be compromised by protease-induced degradation. This limits the feasibility of conducting plasma biomarker studies in environments that lack the capacity for immediate processing and appropriate storage of blood samples. We hypothesized that blood collection tube supplementation with protease inhibitors can improve the stability of plasma biomarkers at room temperatures (RT). This study conducted a comparative analysis of blood biomarker stability in traditional ethylenediaminetetraacetic acid (EDTA) tubes versus BD™ P100 collection tubes, the latter being coated with a protease inhibitor cocktail. The stability of six plasma AD biomarkers was evaluated over time under RT conditions. METHODS We evaluated three experimental approaches. In Approach 1, pooled plasma samples underwent storage at RT for up to 96 hours. In Approach 2, plasma samples isolated upfront from whole blood collected into EDTA or P100 tubes were stored at RT for 0h or 24h before biomarker measurements. In Approach 3, whole blood samples were collected into paired EDTA or P100 tubes, followed by storage at RT for 0h or 24h before isolating the plasma for analyses. Biomarkers were measured with Single Molecule Array (Simoa) and immunoprecipitation-mass spectrometry (IP-MS) assays. RESULTS Both the IP-MS and Simoa methods revealed that the use of P100 tubes significantly improved the stability of Aβ42 and Aβ40 across all approaches. Additionally, the Aβ42/Aβ40 ratio levels were significantly stabilized only in the IP-MS assay in Approach 3. No significant differences were observed in the levels of plasma p-tau181, GFAP, and NfL for samples collected using either tube type in any of the approaches. CONCLUSION Supplementation of blood collection tubes with protease inhibitors could reduce the protease-induced degradation of plasma Aβ42 and Aβ40, and the Aβ ratio for IP-MS assay. This has crucial implications for preanalytical procedures, particularly in resource-limited settings.
Collapse
|
102
|
Nihashi T, Sakurai K, Kato T, Kimura Y, Ito K, Nakamura A, Terasawa T. Blood levels of glial fibrillary acidic protein for predicting clinical progression to Alzheimer's disease in adults without dementia: a systematic review and meta-analysis protocol. Diagn Progn Res 2024; 8:4. [PMID: 38439065 PMCID: PMC10913586 DOI: 10.1186/s41512-024-00167-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/02/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND There is urgent clinical need to identify reliable prognostic biomarkers that predict the progression of dementia symptoms in individuals with early-phase Alzheimer's disease (AD) especially given the research on and predicted applications of amyloid-beta (Aβ)-directed immunotherapies to remove Aβ from the brain. Cross-sectional studies have reported higher levels of cerebrospinal fluid and blood glial fibrillary acidic protein (GFAP) in individuals with AD-associated dementia than in cognitively unimpaired individuals. Further, recent longitudinal studies have assessed the prognostic potential of baseline blood GFAP levels as a predictor of future cognitive decline in cognitively unimpaired individuals and in those with mild cognitive impairment (MCI) due to AD. In this systematic review and meta-analysis, we propose analyzing longitudinal studies on blood GFAP levels to predict future cognitive decline. METHODS This study will include prospective and retrospective cohort studies that assessed blood GFAP levels as a prognostic factor and any prediction models that incorporated blood GFAP levels in cognitively unimpaired individuals or those with MCI. The primary outcome will be conversion to MCI or AD in cognitively unimpaired individuals or conversion to AD in individuals with MCI. Articles from PubMed and Embase will be extracted up to December 31, 2023, without language restrictions. An independent dual screening of abstracts and potentially eligible full-text reports will be conducted. Data will be dual-extracted using the CHeck list for critical appraisal, data extraction for systematic Reviews of prediction Modeling Studies (CHARMS)-prognostic factor, and CHARMS checklists, and we will dual-rate the risk of bias and applicability using the Quality In Prognosis Studies and Prediction Study Risk-of-Bias Assessment tools. We will qualitatively synthesize the study data, participants, index biomarkers, predictive model characteristics, and clinical outcomes. If appropriate, random-effects meta-analyses will be performed to obtain summary estimates. Finally, we will assess the body of evidence using the Grading of Recommendation, Assessment, Development, and Evaluation Approach. DISCUSSION This systematic review and meta-analysis will comprehensively evaluate and synthesize existing evidence on blood GFAP levels for prognosticating presymptomatic individuals and those with MCI to help advance risk-stratified treatment strategies for early-phase AD. TRIAL REGISTRATION PROSPERO CRD42023481200.
Collapse
Affiliation(s)
- Takashi Nihashi
- Department of Radiology, National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
- Department of Biomarker Research, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi, 474-8511, Japan
| | - Keita Sakurai
- Department of Radiology, National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Takashi Kato
- Department of Radiology, National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Yasuyuki Kimura
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Kengo Ito
- National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Akinori Nakamura
- Department of Biomarker Research, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi, 474-8511, Japan
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Teruhiko Terasawa
- Section of General Internal Medicine, Department of Emergency Medicine and General Internal Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan.
| |
Collapse
|
103
|
Komai M, Noda Y, Ikeda A, Kaneshiro N, Kamikubo Y, Sakurai T, Uehara T, Takasugi N. Nuclear SphK2/S1P signaling is a key regulator of ApoE production and Aβ uptake in astrocytes. J Lipid Res 2024; 65:100510. [PMID: 38280459 PMCID: PMC10907773 DOI: 10.1016/j.jlr.2024.100510] [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: 10/24/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/29/2024] Open
Abstract
The link between changes in astrocyte function and the pathological progression of Alzheimer's disease (AD) has attracted considerable attention. Interestingly, activated astrocytes in AD show abnormalities in their lipid content and metabolism. In particular, the expression of apolipoprotein E (ApoE), a lipid transporter, is decreased. Because ApoE has anti-inflammatory and amyloid β (Aβ)-metabolizing effects, the nuclear receptors, retinoid X receptor (RXR) and LXR, which are involved in ApoE expression, are considered promising therapeutic targets for AD. However, the therapeutic effects of agents targeting these receptors are limited or vary considerably among groups, indicating the involvement of an unknown pathological factor that modifies astrocyte and ApoE function. Here, we focused on the signaling lipid, sphingosine-1-phosphate (S1P), which is mainly produced by sphingosine kinase 2 (SphK2) in the brain. Using astrocyte models, we found that upregulation of SphK2/S1P signaling suppressed ApoE induction by both RXR and LXR agonists. We also found that SphK2 activation reduced RXR binding to the APOE promoter region in the nucleus, suggesting the nuclear function of SphK2/S1P. Intriguingly, suppression of SphK2 activity by RNA knockdown or specific inhibitors upregulated lipidated ApoE induction. Furthermore, the induced ApoE facilitates Aβ uptake in astrocytes. Together with our previous findings that SphK2 activity is upregulated in AD brain and promotes Aβ production in neurons, these results indicate that SphK2/S1P signaling is a promising multifunctional therapeutic target for AD that can modulate astrocyte function by stabilizing the effects of RXR and LXR agonists, and simultaneously regulate neuronal pathogenesis.
Collapse
Affiliation(s)
- Masato Komai
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita, Okayama, Japan; Research Fellow of Japan Society for the Promotion of Science, Chiyoda, Tokyo, Japan
| | - Yuka Noda
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita, Okayama, Japan
| | - Atsuya Ikeda
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita, Okayama, Japan
| | - Nanaka Kaneshiro
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita, Okayama, Japan
| | - Yuji Kamikubo
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, Japan
| | - Takashi Sakurai
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, Japan
| | - Takashi Uehara
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita, Okayama, Japan
| | - Nobumasa Takasugi
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita, Okayama, Japan; Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, Japan.
| |
Collapse
|
104
|
Che N, Ou R, Li C, Zhang L, Wei Q, Wang S, Jiang Q, Yang T, Xiao Y, Lin J, Zhao B, Chen X, Shang H. Plasma GFAP as a prognostic biomarker of motor subtype in early Parkinson's disease. NPJ Parkinsons Dis 2024; 10:48. [PMID: 38429295 PMCID: PMC10907600 DOI: 10.1038/s41531-024-00664-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 02/21/2024] [Indexed: 03/03/2024] Open
Abstract
Parkinson's disease (PD) is a heterogeneous movement disorder with different motor subtypes including tremor dominant (TD), indeterminate and postural instability, and gait disturbance (PIGD) motor subtypes. Plasma glial fibrillary acidic protein (GFAP) was elevated in PD patients and may be regarded as a biomarker for motor and cognitive progression. Here we explore if there was an association between plasma GFAP and different motor subtypes and whether baseline plasma GFAP level can predict motor subtype conversion. Patients with PD classified as TD, PIGD or indeterminate subtypes underwent neurological evaluation at baseline and 2 years follow-up. Plasma GFAP in PD patients and controls were measured using an ultrasensitive single molecule array. The study enrolled 184 PD patients and 95 control subjects. Plasma GFAP levels were significantly higher in the PIGD group compared to the TD group at 2-year follow-up. Finally, 45% of TD patients at baseline had a subtype shift and 85% of PIGD patients at baseline remained as PIGD subtypes at 2 years follow-up. Baseline plasma GFAP levels were significantly higher in TD patients converted to PIGD than non-converters in the baseline TD group. Higher baseline plasma GFAP levels were significantly associated with the TD motor subtype conversion (OR = 1.283, P = 0.033) and lower baseline plasma GFAP levels in PIGD patients were likely to shift to TD and indeterminate subtype (OR = 0.551, P = 0.021) after adjusting for confounders. Plasma GFAP may serve as a clinical utility biomarker in differentiating motor subtypes and predicting baseline motor subtypes conversion in PD patients.
Collapse
Affiliation(s)
- Ningning Che
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruwei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chunyu Li
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingyu Zhang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qianqian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shichan Wang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qirui Jiang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tianmi Yang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Xiao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junyu Lin
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bi Zhao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xueping Chen
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
105
|
Varma VR, An Y, Kac PR, Bilgel M, Moghekar A, Loeffler T, Amschl D, Troncoso J, Blennow K, Zetterberg H, Ashton NJ, Resnick SM, Thambisetty M. Longitudinal progression of blood biomarkers reveals a key role of astrocyte reactivity in preclinical Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.25.24301779. [PMID: 38343809 PMCID: PMC10854357 DOI: 10.1101/2024.01.25.24301779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/26/2024]
Abstract
Defining the progression of blood biomarkers of Alzheimer's disease (AD) is essential for targeting treatments in patients most likely to benefit from early intervention. We delineated the temporal ordering of blood biomarkers a decade prior to the onset of AD symptoms in participants in the Baltimore Longitudinal Study of Aging. We show that increased astrocyte reactivity, assessed by elevated glial fibrillary acidic protein (GFAP) levels is an early event in the progression of blood biomarker changes in preclinical AD. In AD-converters who are initially cognitively unimpaired (N=158, 377 serial plasma samples), higher plasma GFAP levels are observed as early as 10-years prior to the onset of cognitive impairment due to incident AD compared to individuals who remain cognitively unimpaired (CU, N=160, 379 serial plasma samples). Plasma GFAP levels in AD-converters remain elevated 5-years prior to and coincident with the onset of cognitive impairment due to AD. In participants with neuropathologically confirmed AD, plasma GFAP levels are elevated relative to cognitively normal individuals and intermediate in those who remain cognitively unimpaired despite significant AD pathology (asymptomatic AD). Higher plasma GFAP levels at death are associated with greater severity of both neuritic plaques and neurofibrillary tangles. In the 5XFAD transgenic model of AD, we observed greater GFAP levels in the cortex and hippocampus of transgenic mice relative to wild-type prior to the development of cognitive impairment. Reactive astrocytosis, an established biological response to neuronal injury, may be an early initiator of AD pathogenesis and a promising therapeutic target.
Collapse
Affiliation(s)
- V R Varma
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, United States of America
| | - Y An
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - P R Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - M Bilgel
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - A Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - T Loeffler
- Scantox Neuro GmbH, Parkring 12, 8074, Grambach, Austria
| | - D Amschl
- Scantox Neuro GmbH, Parkring 12, 8074, Grambach, Austria
| | - J Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - K Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - H Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - N J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- King's College London, Institute of Psychiatry, Psychology and Neuroscience Maurice Wohl Institute Clinical Neuroscience Institute London UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation London UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - S M Resnick
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - M Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, United States of America
| |
Collapse
|
106
|
Batista AF, Khan KA, Papavergi MT, Lemere CA. The Importance of Complement-Mediated Immune Signaling in Alzheimer's Disease Pathogenesis. Int J Mol Sci 2024; 25:817. [PMID: 38255891 PMCID: PMC10815224 DOI: 10.3390/ijms25020817] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/05/2024] [Accepted: 01/07/2024] [Indexed: 01/24/2024] Open
Abstract
As an essential component of our innate immune system, the complement system is responsible for our defense against pathogens. The complement cascade has complex roles in the central nervous system (CNS), most of what we know about it stems from its role in brain development. However, in recent years, numerous reports have implicated the classical complement cascade in both brain development and decline. More specifically, complement dysfunction has been implicated in neurodegenerative disorders, such as Alzheimer's disease (AD), which is the most common form of dementia. Synapse loss is one of the main pathological hallmarks of AD and correlates with memory impairment. Throughout the course of AD progression, synapses are tagged with complement proteins and are consequently removed by microglia that express complement receptors. Notably, astrocytes are also capable of secreting signals that induce the expression of complement proteins in the CNS. Both astrocytes and microglia are implicated in neuroinflammation, another hallmark of AD pathogenesis. In this review, we provide an overview of previously known and newly established roles for the complement cascade in the CNS and we explore how complement interactions with microglia, astrocytes, and other risk factors such as TREM2 and ApoE4 modulate the processes of neurodegeneration in both amyloid and tau models of AD.
Collapse
Affiliation(s)
- André F. Batista
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.F.B.); (K.A.K.); (M.-T.P.)
| | - Khyrul A. Khan
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.F.B.); (K.A.K.); (M.-T.P.)
| | - Maria-Tzousi Papavergi
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.F.B.); (K.A.K.); (M.-T.P.)
- School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Cynthia A. Lemere
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.F.B.); (K.A.K.); (M.-T.P.)
| |
Collapse
|
107
|
Bhattarai P, Gunasekaran TI, Reyes-Dumeyer D, Jülich D, Tayran H, Yilmaz E, Flaherty D, Lantigua R, Medrano M, Rivera D, Recio P, Ertekin-Taner N, Teich AF, Dickson DW, Holley S, Mayeux R, Kizil C, Vardarajan BN. Rare genetic variation in Fibronectin 1 ( FN1 ) protects against APOEe4 in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.02.573895. [PMID: 38260431 PMCID: PMC10802344 DOI: 10.1101/2024.01.02.573895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The risk of developing Alzheimer's disease (AD) significantly increases in individuals carrying the APOEε4 allele. Elderly cognitively healthy individuals with APOEε4 also exist, suggesting the presence of cellular mechanisms that counteract the pathological effects of APOEε4 ; however, these mechanisms are unknown. We hypothesized that APOEε4 carriers without dementia might carry genetic variations that could protect them from developing APOEε4- mediated AD pathology. To test this, we leveraged whole genome sequencing (WGS) data in National Institute on Aging Alzheimer's Disease Family Based Study (NIA-AD FBS), Washington Heights/Inwood Columbia Aging Project (WHICAP), and Estudio Familiar de Influencia Genetica en Alzheimer (EFIGA) cohorts and identified potentially protective variants segregating exclusively among unaffected APOEε4 carriers. In homozygous unaffected carriers above 70 years old, we identified 510 rare coding variants. Pathway analysis of the genes harboring these variants showed significant enrichment in extracellular matrix (ECM)-related processes, suggesting protective effects of functional modifications in ECM proteins. We prioritized two genes that were highly represented in the ECM-related gene ontology terms, (FN1) and collagen type VI alpha 2 chain ( COL6A2 ) and are known to be expressed at the blood-brain barrier (BBB), for postmortem validation and in vivo functional studies. The FN1 and COL6A2 protein levels were increased at the BBB in APOEε4 carriers with AD. Brain expression of cognitively unaffected homozygous APOEε4 carriers had significantly lower FN1 deposition and less reactive gliosis compared to homozygous APOEε4 carriers with AD, suggesting that FN1 might be a downstream driver of APOEε4 -mediated AD-related pathology and cognitive decline. To validate our findings, we used zebrafish models with loss-of-function (LOF) mutations in fn1b - the ortholog for human FN1 . We found that fibronectin LOF reduced gliosis, enhanced gliovascular remodeling and potentiated the microglial response, suggesting that pathological accumulation of FN1 could impair toxic protein clearance, which is ameliorated with FN1 LOF. Our study suggests vascular deposition of FN1 is related to the pathogenicity of APOEε4 , LOF variants in FN1 may reduce APOEε4 -related AD risk, providing novel clues to potential therapeutic interventions targeting the ECM to mitigate AD risk.
Collapse
|
108
|
Rathee S, Sen D, Pandey V, Jain SK. Advances in Understanding and Managing Alzheimer's Disease: From Pathophysiology to Innovative Therapeutic Strategies. Curr Drug Targets 2024; 25:752-774. [PMID: 39039673 DOI: 10.2174/0113894501320096240627071400] [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: 04/19/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 07/24/2024]
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disorder characterized by the presence of amyloid-β (Aβ) plaques and tau-containing neurofibrillary tangles, leading to cognitive and physical decline. Representing the majority of dementia cases, AD poses a significant burden on healthcare systems globally, with onset typically occurring after the age of 65. While most cases are sporadic, about 10% exhibit autosomal forms associated with specific gene mutations. Neurofibrillary tangles and Aβ plaques formed by misfolded tau proteins and Aβ peptides contribute to neuronal damage and cognitive impairment. Currently, approved drugs, such as acetylcholinesterase inhibitors and N-methyl D-aspartate receptor agonists, offer only partial symptomatic relief without altering disease progression. A promising development is using lecanemab, a humanized IgG1 monoclonal antibody, as an immune therapeutic approach. Lecanemab demonstrates selectivity for polymorphic Aβ variants and binds to large soluble Aβ aggregates, providing a potential avenue for targeted treatment. This shift in understanding the role of the adaptive immune response in AD pathogenesis opens new possibilities for therapeutic interventions aiming to address the disease's intricate mechanisms. This review aims to summarize recent advancements in understanding Alzheimer's disease pathophysiology and innovative therapeutic approaches, providing valuable insights for both researchers and clinicians.
Collapse
Affiliation(s)
- Sunny Rathee
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Debasis Sen
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Vishal Pandey
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Sanjay K Jain
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| |
Collapse
|
109
|
Zhang M, Chen H, Huang W, Guo T, Ma G, Han Y, Shu N. Relationship between topological efficiency of white matter structural connectome and plasma biomarkers across the Alzheimer's disease continuum. Hum Brain Mapp 2024; 45:e26566. [PMID: 38224535 PMCID: PMC10785192 DOI: 10.1002/hbm.26566] [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: 08/23/2023] [Revised: 11/11/2023] [Accepted: 11/30/2023] [Indexed: 01/17/2024] Open
Abstract
Both plasma biomarkers and brain network topology have shown great potential in the early diagnosis of Alzheimer's disease (AD). However, the specific associations between plasma AD biomarkers, structural network topology, and cognition across the AD continuum have yet to be fully elucidated. This retrospective study evaluated participants from the Sino Longitudinal Study of Cognitive Decline cohort between September 2009 and October 2022 with available blood samples or 3.0-T MRI brain scans. Plasma biomarker levels were measured using the Single Molecule Array platform, including β-amyloid (Aβ), phosphorylated tau181 (p-tau181), glial fibrillary acidic protein (GFAP), and neurofilament light chain (NfL). The topological structure of brain white matter was assessed using network efficiency. Trend analyses were carried out to evaluate the alterations of the plasma markers and network efficiency with AD progression. Correlation and mediation analyses were conducted to further explore the relationships among plasma markers, network efficiency, and cognitive performance across the AD continuum. Among the plasma markers, GFAP emerged as the most sensitive marker (linear trend: t = 11.164, p = 3.59 × 10-24 ; quadratic trend: t = 7.708, p = 2.25 × 10-13 ; adjusted R2 = 0.475), followed by NfL (linear trend: t = 6.542, p = 2.9 × 10-10 ; quadratic trend: t = 3.896, p = 1.22 × 10-4 ; adjusted R2 = 0.330), p-tau181 (linear trend: t = 8.452, p = 1.61 × 10-15 ; quadratic trend: t = 6.316, p = 1.05 × 10-9 ; adjusted R2 = 0.346) and Aβ42/Aβ40 (linear trend: t = -3.257, p = 1.27 × 10-3 ; quadratic trend: t = -1.662, p = 9.76 × 10-2 ; adjusted R2 = 0.101). Local efficiency decreased in brain regions across the frontal and temporal cortex and striatum. The principal component of local efficiency within these regions was correlated with GFAP (Pearson's R = -0.61, p = 6.3 × 10-7 ), NfL (R = -0.57, p = 6.4 × 10-6 ), and p-tau181 (R = -0.48, p = 2.0 × 10-4 ). Moreover, network efficiency mediated the relationship between general cognition and GFAP (ab = -0.224, 95% confidence interval [CI] = [-0.417 to -0.029], p = .0196 for MMSE; ab = -0.198, 95% CI = [-0.42 to -0.003], p = .0438 for MOCA) or NfL (ab = -0.224, 95% CI = [-0.417 to -0.029], p = .0196 for MMSE; ab = -0.198, 95% CI = [-0.42 to -0.003], p = .0438 for MOCA). Our findings suggest that network efficiency mediates the association between plasma biomarkers, specifically GFAP and NfL, and cognitive performance in the context of AD progression, thus highlighting the potential utility of network-plasma approaches for early detection, monitoring, and intervention strategies in the management of AD.
Collapse
Affiliation(s)
- Mingkai Zhang
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Haojie Chen
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain ResearchBeijing Normal UniversityBeijingChina
- BABRI CentreBeijing Normal UniversityBeijingChina
- Beijing Key Laboratory of Brain Imaging and ConnectomicsBeijing Normal UniversityBeijingChina
| | - Weijie Huang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain ResearchBeijing Normal UniversityBeijingChina
- BABRI CentreBeijing Normal UniversityBeijingChina
- Beijing Key Laboratory of Brain Imaging and ConnectomicsBeijing Normal UniversityBeijingChina
| | - Tengfei Guo
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Guolin Ma
- Department of RadiologyChina‐Japan Friendship HospitalBeijingChina
| | - Ying Han
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
- School of Biomedical EngineeringHainan UniversityHaikouChina
- National Clinical Research Center for Geriatric DiseasesBeijingChina
- Center of Alzheimer's DiseaseBeijing Institute for Brain DisordersBeijingChina
| | - Ni Shu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain ResearchBeijing Normal UniversityBeijingChina
- BABRI CentreBeijing Normal UniversityBeijingChina
- Beijing Key Laboratory of Brain Imaging and ConnectomicsBeijing Normal UniversityBeijingChina
| |
Collapse
|
110
|
Chen X, Chen A, Wei J, Huang Y, Deng J, Chen P, Yan Y, Lin M, Chen L, Zhang J, Huang Z, Zeng X, Gong C, Zheng X. Dexmedetomidine alleviates cognitive impairment by promoting hippocampal neurogenesis via BDNF/TrkB/CREB signaling pathway in hypoxic-ischemic neonatal rats. CNS Neurosci Ther 2024; 30:e14486. [PMID: 37830170 PMCID: PMC10805444 DOI: 10.1111/cns.14486] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023] Open
Abstract
AIMS Dexmedetomidine (DEX) has been reported to alleviate hypoxic-ischemic brain damage (HIBD) in neonates. This study aimed to investigate whether DEX improves cognitive impairment by promoting hippocampal neurogenesis via the BDNF/TrkB/CREB signaling pathway in neonatal rats with HIBD. METHODS HIBD was induced in postnatal day 7 rats using the Rice-Vannucci method, and DEX (25 μg/kg) was administered intraperitoneally immediately after the HIBD induction. The BDNF/TrkB/CREB pathway was regulated by administering the TrkB receptor antagonist ANA-12 through intraperitoneal injection or by delivering adeno-associated virus (AAV)-shRNA-BDNF via intrahippocampal injection. Western blot was performed to measure the levels of BDNF, TrkB, and CREB. Immunofluorescence staining was utilized to identify the polarization of astrocytes and evaluate the levels of neurogenesis in the dentate gyrus of the hippocampus. Nissl and TTC staining were performed to evaluate the extent of neuronal damage. The MWM test was conducted to evaluate spatial learning and memory ability. RESULTS The levels of BDNF and neurogenesis exhibited a notable decrease in the hippocampus of neonatal rats after HIBD, as determined by RNA-sequencing technology. Our results demonstrated that treatment with DEX effectively increased the protein expression of BDNF and the phosphorylation of TrkB and CREB, promoting neurogenesis in the dentate gyrus of the hippocampus in neonatal rats with HIBD. Specifically, DEX treatment significantly augmented the expression of BDNF in hippocampal astrocytes, while decreasing the proportion of detrimental A1 astrocytes and increasing the proportion of beneficial A2 astrocytes in neonatal rats with HIBD. Furthermore, inhibiting the BDNF/TrkB/CREB pathway using either ANA-12 or AAV-shRNA-BDNF significantly counteracted the advantageous outcomes of DEX on hippocampal neurogenesis, neuronal survival, and cognitive improvement. CONCLUSIONS DEX promoted neurogenesis in the hippocampus by activating the BDNF/TrkB/CREB pathway through the induction of polarization of A1 astrocytes toward A2 astrocytes, subsequently mitigating neuronal damage and cognitive impairment in neonates with HIBD.
Collapse
Affiliation(s)
- Xiaohui Chen
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Andi Chen
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Jianjie Wei
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Yongxin Huang
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Jianhui Deng
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Pinzhong Chen
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Yanlin Yan
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Mingxue Lin
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Lifei Chen
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Jiuyun Zhang
- Fujian Provincial Key Laboratory of Emergency MedicineFuzhouChina
| | - Zhibin Huang
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Xiaoqian Zeng
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Cansheng Gong
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Xiaochun Zheng
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
- Fujian Provincial Key Laboratory of Emergency MedicineFuzhouChina
- Fujian Emergency Medical Center, Fujian Provincial Key Laboratory of Critical Care MedicineFujian Provincial Co‐Constructed Laboratory of “Belt and Road”FuzhouChina
| |
Collapse
|
111
|
Ohno M. A Strategy for Allowing Earlier Diagnosis and Rigorous Evaluation of BACE1 Inhibitors in Preclinical Alzheimer's Disease. J Alzheimers Dis 2024; 99:431-445. [PMID: 38701146 DOI: 10.3233/jad-231451] [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: 05/05/2024]
Abstract
Given continued failure of BACE1 inhibitor programs at symptomatic and prodromal stages of Alzheimer's disease (AD), clinical trials need to target the earlier preclinical stage. However, trial design is complex in this population with negative diagnosis of classical hippocampal amnesia on standard memory tests. Besides recent advances in brain imaging, electroencephalogram, and fluid-based biomarkers, new cognitive markers should be established for earlier diagnosis that can optimize recruitment to BACE1 inhibitor trials in presymptomatic AD. Notably, accelerated long-term forgetting (ALF) is emerging as a sensitive cognitive measure that can discriminate between asymptomatic individuals with high risks for developing AD and healthy controls. ALF is a form of declarative memory impairment characterized by increased forgetting rates over longer delays (days to months) despite normal storage within the standard delays of testing (20-60 min). Therefore, ALF may represent a harbinger of preclinical dementia and the impairment of systems memory consolidation, during which memory traces temporarily stored in the hippocampus become gradually integrated into cortical networks. This review provides an overview of the utility of ALF in a rational design of next-generation BACE1 inhibitor trials in preclinical AD. I explore potential mechanisms underlying ALF and relevant early-stage biomarkers useful for BACE1 inhibitor evaluation, including synaptic protein alterations, astrocytic dysregulation and neuron hyperactivity in the hippocampal-cortical network. Furthermore, given the physiological role of the isoform BACE2 as an AD-suppressor gene, I also discuss the possible association between the poor selectivity of BACE1 inhibitors and their side effects (e.g., cognitive worsening) in prior clinical trials.
Collapse
Affiliation(s)
- Masuo Ohno
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
| |
Collapse
|
112
|
Park H, Kwon HS, Lee KY, Kim YE, Son JW, Choi NY, Lee EJ, Han MH, Park DW, Kim S, Koh SH. GV1001 modulates neuroinflammation and improves memory and behavior through the activation of gonadotropin-releasing hormone receptors in a triple transgenic Alzheimer's disease mouse model. Brain Behav Immun 2024; 115:295-307. [PMID: 37884161 DOI: 10.1016/j.bbi.2023.10.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/22/2023] [Accepted: 10/22/2023] [Indexed: 10/28/2023] Open
Abstract
GV1001 protects neural cells from amyloid-β (Aβ) toxicity and other stressors in in vitro studies and demonstrates clinically beneficial effects in patients with moderate to severe Alzheimer's disease (AD). Here, we investigated the protective effects and mechanism of action of GV1001 in triple transgenic AD (3xTg-AD) mice. We found that GV1001 improved memory and cognition in middle- and old-aged 3xTg-AD mice. Additionally, it reduced Aβ oligomer and phospho-tau (Ser202 and Thr205) levels in the brain, and mitigated neuroinflammation by promoting a neuroprotective microglial and astrocyte phenotype while diminishing the neurotoxic ones. In vitro, GV1001 bound to gonadotropin releasing hormone receptors (GnRHRs) with high affinity. Levels of cyclic adenosine monophosphate, a direct downstream effector of activated GnRHRs, increased after GV1001 treatment. Furthermore, inhibition of GnRHRs blocked GV1001-induced effects. Thus, GV1001 might improve cognitive and memory functions of 3xTg-AD mice by suppressing neuroinflammation and reducing Aβ oligomers levels and phospho-tau by activating GnRHRs and their downstream signaling pathways.
Collapse
Affiliation(s)
- Hyunhee Park
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Hyuk Sung Kwon
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Kyu-Yong Lee
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Ye Eun Kim
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Jeong-Woo Son
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Na-Young Choi
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Eun Ji Lee
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Myung-Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Dong Woo Park
- Department of Radiology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Sangjae Kim
- Teloid Inc., 3580 Wilshire Boulevard, Suite 900-31, Los Angeles, CA 90010, USA.
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea; Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, South Korea.
| |
Collapse
|
113
|
Zhao Z, Liu Y, Ruan S, Hu Y. Current Anti-Amyloid-β Therapy for Alzheimer's Disease Treatment: From Clinical Research to Nanomedicine. Int J Nanomedicine 2023; 18:7825-7845. [PMID: 38144511 PMCID: PMC10749171 DOI: 10.2147/ijn.s444115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/12/2023] [Indexed: 12/26/2023] Open
Abstract
Recent successive approval of anti-amyloid-β (Aβ) monoclonal antibodies as disease-modifying therapies against Alzheimer's disease (AD) has raised great confidence in the development of anti-AD therapies; however, the current therapies still face the dilemma of significant adverse reactions and limited effects. In this review, we summarized the therapeutic characteristics of the approved anti-Aβ immunotherapies and dialectically analyzed the gains and losses from clinical trials. The review further proposed the reasonable selection of animal models in preclinical studies from the perspective of different animal models of Aβ deposition and deals in-depth with the recent advances of exploring preclinical nanomedical application in Aβ targeted therapy, aiming to provide a reliable systematic summary for the development of novel anti-Aβ therapies. Collectively, this review comprehensively dissects the pioneering work of Aβ-targeted therapies and proposed perspective insight into AD-modified therapies.
Collapse
Affiliation(s)
- Zixuan Zhao
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, People’s Republic of China
- The Institute of Brain Science, Wannan Medical College, Wuhu, 241000, People’s Republic of China
| | - Yun Liu
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, People’s Republic of China
- The Institute of Brain Science, Wannan Medical College, Wuhu, 241000, People’s Republic of China
| | - Shirong Ruan
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, People’s Republic of China
- The Institute of Brain Science, Wannan Medical College, Wuhu, 241000, People’s Republic of China
| | - Yixuan Hu
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, People’s Republic of China
- The Institute of Brain Science, Wannan Medical College, Wuhu, 241000, People’s Republic of China
| |
Collapse
|
114
|
Weber DM, Taylor SW, Lagier RJ, Kim JC, Goldman SM, Clarke NJ, Vaillancourt DE, Duara R, McFarland KN, Wang WE, Golde TE, Racke MK. Clinical utility of plasma Aβ42/40 ratio by LC-MS/MS in Alzheimer's disease assessment. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.12.23299878. [PMID: 38168329 PMCID: PMC10760303 DOI: 10.1101/2023.12.12.23299878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
INTRODUCTION Plasma Aβ42/40 ratio can be used to help predict amyloid PET status, but its clinical utility in Alzheimer's disease (AD) assessment is unclear. METHODS Aβ42/40 ratio was measured by LC-MS/MS in 250 specimens with associated amyloid PET imaging, diagnosis, and demographic data, and 6,192 consecutive clinical specimens submitted for Aβ42/40 testing. RESULTS High diagnostic sensitivity and negative predictive value (NPV) for Aβ-PET positivity were observed, consistent with the clinical performance of other plasma LC-MS/MS assays, but with greater separation between Aβ42/40 values for individuals with positive vs negative Aβ-PET results. Assuming a moderate prevalence of Aβ-PET positivity, a cutpoint was identified with 99% NPV, which could help predict that AD is likely not the cause of patients' cognitive impairment and help reduce PET evaluation by about 40%. DISCUSSION Using high-throughput plasma Aβ42/40 LC-MS/MS assays can help reduce PET evaluations in patients with low likelihood of AD pathology, allowing for cost savings.
Collapse
Affiliation(s)
- Darren M Weber
- Quest Diagnostics Nichols Institute, San Juan Capistrano, CA USA
| | - Steven W Taylor
- Quest Diagnostics Nichols Institute, San Juan Capistrano, CA USA
| | - Robert J Lagier
- Quest Diagnostics Nichols Institute, San Juan Capistrano, CA USA
| | - Jueun C Kim
- Quest Diagnostics Nichols Institute, San Juan Capistrano, CA USA
| | - Scott M Goldman
- Quest Diagnostics Nichols Institute, San Juan Capistrano, CA USA
| | - Nigel J Clarke
- Quest Diagnostics Nichols Institute, San Juan Capistrano, CA USA
| | - David E Vaillancourt
- Department of Applied Physiology and Kinesiology, Fixel Institute for Neurological Disorders, and 1Florida ADRC, University of Florida, Gainesville, FL USA
| | - Ranjan Duara
- Department of Applied Physiology and Kinesiology, Fixel Institute for Neurological Disorders, and 1Florida ADRC, University of Florida, Gainesville, FL USA
- Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL USA
| | - Karen N McFarland
- Department of Neurology, Center for Translational Research in Neurodegenerative Disease, 1Florida Alzheimer's Disease Research Center (ADRC), University of Florida, Gainesville, FL USA
| | - Wei-En Wang
- Department of Applied Physiology and Kinesiology, Fixel Institute for Neurological Disorders, and 1Florida ADRC, University of Florida, Gainesville, FL USA
| | - Todd E Golde
- Department of Neurology, Center for Translational Research in Neurodegenerative Disease, 1Florida Alzheimer's Disease Research Center (ADRC), University of Florida, Gainesville, FL USA
- Department of Pharmacology and Chemical Biology and Department of Neurology Center for Neurodegenerative Disease, Goizueta Institute Emory Brain Health, Emory University, School of Medicine. Atlanta, GA USA
| | - Michael K Racke
- Quest Diagnostics Nichols Institute, San Juan Capistrano, CA USA
| |
Collapse
|
115
|
Steward A, Biel D, Dewenter A, Roemer S, Wagner F, Dehsarvi A, Rathore S, Otero Svaldi D, Higgins I, Brendel M, Dichgans M, Shcherbinin S, Ewers M, Franzmeier N. ApoE4 and Connectivity-Mediated Spreading of Tau Pathology at Lower Amyloid Levels. JAMA Neurol 2023; 80:1295-1306. [PMID: 37930695 PMCID: PMC10628846 DOI: 10.1001/jamaneurol.2023.4038] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/07/2023] [Indexed: 11/07/2023]
Abstract
Importance For the Alzheimer disease (AD) therapies to effectively attenuate clinical progression, it may be critical to intervene before the onset of amyloid-associated tau spreading, which drives neurodegeneration and cognitive decline. Time points at which amyloid-associated tau spreading accelerates may depend on individual risk factors, such as apolipoprotein E ε4 (ApoE4) carriership, which is linked to faster disease progression; however, the association of ApoE4 with amyloid-related tau spreading is unclear. Objective To assess if ApoE4 carriers show accelerated amyloid-related tau spreading and propose amyloid positron emission tomography (PET) thresholds at which tau spreading accelerates in ApoE4 carriers vs noncarriers. Design, Setting, and Participants This cohort study including combined ApoE genotyping, amyloid PET, and longitudinal tau PET from 2 independent samples: the Alzheimer's Disease Neuroimaging Initiative (ADNI; n = 237; collected from April 2015 to August 2022) and Avid-A05 (n = 130; collected from December 2013 to July 2017) with a mean (SD) tau PET follow-up time of 1.9 (0.96) years in ADNI and 1.4 (0.23) years in Avid-A05. ADNI is an observational multicenter Alzheimer disease neuroimaging initiative and Avid-A05 an observational clinical trial. Participants classified as cognitively normal (152 in ADNI and 77 in Avid-A05) or mildly cognitively impaired (107 in ADNI and 53 in Avid-A05) were selected based on ApoE genotyping, amyloid-PET, and longitudinal tau PET data availability. Participants with ApoE ε2/ε4 genotype or classified as having dementia were excluded. Resting-state functional magnetic resonance imaging connectivity templates were based on 42 healthy participants in ADNI. Main Outcomes and Measures Mediation of amyloid PET on the association between ApoE4 status and subsequent tau PET increase through Braak stage regions and interaction between ApoE4 status and amyloid PET with annual tau PET increase through Braak stage regions and connectivity-based spreading stages (tau epicenter connectivity ranked regions). Results The mean (SD) age was 73.9 (7.35) years among the 237 ADNI participants and 70.2 (9.7) years among the 130 Avid-A05 participants. A total of 107 individuals in ADNI (45.1%) and 45 in Avid-A05 (34.6%) were ApoE4 carriers. Across both samples, we found that higher amyloid PET-mediated ApoE4-related tau PET increased globally (ADNI b, 0.15; 95% CI, 0.05-0.28; P = .001 and Avid-A05 b, 0.33; 95% CI, 0.14-0.54; P < .001) and in earlier Braak regions. Further, we found a significant association between ApoE4 status by amyloid PET interaction and annual tau PET increases consistently through early Braak- and connectivity-based stages where amyloid-related tau accumulation was accelerated in ApoE4carriers vs noncarriers at lower centiloid thresholds, corrected for age and sex. Conclusions and Relevance The findings in this study indicate that amyloid-related tau accumulation was accelerated in ApoE4 carriers at lower amyloid levels, suggesting that ApoE4 may facilitate earlier amyloid-driven tau spreading across connected brain regions. Possible therapeutic implications might be further investigated to determine when best to prevent tau spreading and thus cognitive decline depending on ApoE4 status.
Collapse
Affiliation(s)
- Anna Steward
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Davina Biel
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Anna Dewenter
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Sebastian Roemer
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Department of Neurology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Fabian Wagner
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Amir Dehsarvi
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | | | | | | | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| | | | - Michael Ewers
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
116
|
Edwards NC, Lao PJ, Alshikho MJ, Ericsson OM, Rizvi B, Petersen ME, O’Bryant S, Flores-Aguilar L, Simoes S, Mapstone M, Tudorascu DL, Janelidze S, Hansson O, Handen BL, Christian BT, Lee JH, Lai F, Rosas HD, Zaman S, Lott IT, Yassa MA, Gutierrez J, Wilcock DM, Head E, Brickman AM. Cerebrovascular disease drives Alzheimer plasma biomarker concentrations in adults with Down syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.28.23298693. [PMID: 38076904 PMCID: PMC10705616 DOI: 10.1101/2023.11.28.23298693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Importance By age 40 years over 90% of adults with Down syndrome (DS) have Alzheimer's disease (AD) pathology and most progress to dementia. Despite having few systemic vascular risk factors, individuals with DS have elevated cerebrovascular disease (CVD) markers that track with the clinical progression of AD, suggesting a role for CVD that is hypothesized to be mediated by inflammatory factors. Objective To examine the pathways through which small vessel CVD contributes to AD-related pathophysiology and neurodegeneration in adults with DS. Design Cross sectional analysis of neuroimaging, plasma, and clinical data. Setting Participants were enrolled in Alzheimer's Biomarker Consortium - Down Syndrome (ABC-DS), a multisite study of AD in adults with DS. Participants One hundred eighty-five participants (mean [SD] age=45.2 [9.3] years) with available MRI and plasma biomarker data were included. White matter hyperintensity (WMH) volumes were derived from T2-weighted FLAIR MRI scans and plasma biomarker concentrations of amyloid beta (Aβ42/Aβ40), phosphorylated tau (p-tau217), astrocytosis (glial fibrillary acidic protein, GFAP), and neurodegeneration (neurofilament light chain, NfL) were measured with ultrasensitive immunoassays. Main Outcomes and Measures We examined the bivariate relationships of WMH, Aβ42/Aβ40, p-tau217, and GFAP with age-residualized NfL across AD diagnostic groups. A series of mediation and path analyses examined causal pathways linking WMH and AD pathophysiology to promote neurodegeneration in the total sample and groups stratified by clinical diagnosis. Results There was a direct and indirect bidirectional effect through GFAP of WMH on p-tau217 concentration, which was associated with NfL concentration in the entire sample. Among cognitively stable participants, WMH was directly and indirectly, through GFAP, associated with p-tau217 concentration, and in those with MCI, there was a direct effect of WMH on p-tau217 and NfL concentrations. There were no associations of WMH with biomarker concentrations among those diagnosed with dementia. Conclusions and Relevance The findings suggest that among individuals with DS, CVD promotes neurodegeneration by increasing astrocytosis and tau pathophysiology in the presymptomatic phases of AD. This work joins an emerging literature that implicates CVD and its interface with neuroinflammation as a core pathological feature of AD in adults with DS.
Collapse
Affiliation(s)
- Natalie C. Edwards
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
- Department of Neuroscience, Columbia University, New York City, NY, USA
| | - Patrick J. Lao
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Mohamad J. Alshikho
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Olivia M. Ericsson
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Batool Rizvi
- Department of Neurobiology & Behavior, University of California, Irvine, CA, USA
| | | | - Sid O’Bryant
- University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Lisi Flores-Aguilar
- Department of Pathology and Laboratory Medicine, University of California Irvine School of Medicine, University of California, Irvine, CA, USA
| | - Sabrina Simoes
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Mark Mapstone
- Department of Neurology, University of California, Irvine, CA, USA
| | - Dana L. Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | | | - Joseph H. Lee
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Florence Lai
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - H Diana Rosas
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Center for Neuroimaging of Aging and neurodegenerative Diseases, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Shahid Zaman
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Ira T. Lott
- Department of Pediatrics and Neurology, School of Medicine, University of California, Irvine, CA, USA
| | - Michael A. Yassa
- Department of Neurobiology & Behavior, University of California, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA
| | - José Gutierrez
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Donna M. Wilcock
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
- Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California Irvine School of Medicine, University of California, Irvine, CA, USA
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| |
Collapse
|
117
|
Lv X, Cheng Z, Wang Q, Gao F, Dai L, Du C, Liu C, Xie Q, Shen Y, Shi J. High burdens of phosphorylated tau protein and distinct precuneus atrophy in sporadic early-onset Alzheimer's disease. Sci Bull (Beijing) 2023; 68:2817-2826. [PMID: 37919158 DOI: 10.1016/j.scib.2023.10.019] [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: 07/15/2023] [Revised: 09/16/2023] [Accepted: 10/17/2023] [Indexed: 11/04/2023]
Abstract
Early-onset Alzheimer's disease (EOAD) is a rare devastating subclassification of Alzheimer's disease (AD). EOAD affects individuals <65 years old, and accounts for 5%-10% of all AD cases. Previous studies on EOAD primarily focused on familial forms, whereas research on sporadic EOAD (sEOAD), which represents 85%-90% of EOAD cases, is limited. In this prospective cohort study, participants were recruited between 2018 and 2023 and included patients with sEOAD (n = 110), late-onset AD (LOAD, n = 89), young controls (YC, n = 50), and older controls (OC, n = 25). All AD patients fulfilled the diagnostic criteria based on biomarker evidence. Familial EOAD patients or non-AD dementia patients were excluded. Single molecule array technology was used to measure fluid biomarkers, including cerebrospinal fluid (CSF) and plasma amyloid beta (Aβ) 40, Aβ42, phosphorylated tau (P-tau) 181, total tau (T-tau), serum neurofilament light chain and glial fibrillary acidic protein (GFAP). Patients with sEOAD exhibited more severe executive function impairment and bilateral precuneus atrophy (P < 0.05, family-wise error corrected) than patients with LOAD. Patients with sEOAD showed elevated CSF and plasma P-tau181 levels (154.0 ± 81.2 pg/mL, P = 0.002; and 6.1 ± 2.3 pg/mL, P = 0.046). Moreover, precuneus atrophy was significantly correlated with serum GFAP levels in sEOAD (P < 0.001). Serum GFAP levels (area under the curve (AUC) = 96.0%, cutoff value = 154.3 pg/mL) displayed excellent diagnostic value in distinguishing sEOAD patients from the control group. These preliminary findings highlight the crucial role of tau protein phosphorylation in the pathogenesis and progression of sEOAD.
Collapse
Affiliation(s)
- Xinyi Lv
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zhaozhao Cheng
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Qiong Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Feng Gao
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Linbin Dai
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Chen Du
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Chang Liu
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Qiang Xie
- Department of Nuclear Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Yong Shen
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei 230001, China.
| | - Jiong Shi
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
118
|
Denman CR, Park SM, Jo J. Gut-brain axis: gut dysbiosis and psychiatric disorders in Alzheimer's and Parkinson's disease. Front Neurosci 2023; 17:1268419. [PMID: 38075261 PMCID: PMC10704039 DOI: 10.3389/fnins.2023.1268419] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/30/2023] [Indexed: 02/17/2025] Open
Abstract
Gut dysbiosis and psychiatric symptoms are common early manifestations of Alzheimer's disease (AD) and Parkinson's disease (PD). These diseases, characterised by progressive neuron loss and pathological protein accumulation, impose debilitating effects on patients. Recently, these pathological proteins have been linked with gut dysbiosis and psychiatric disorders. The gut-brain axis links the enteric and central nervous systems, acting as a bidirectional communication pathway to influence brain function and behavior. The relationship triad between gut dysbiosis, psychiatric disorders, and neurodegeneration has been investigated in pairs; however, evidence suggests that they are all interrelated and a deeper understanding is required to unravel the nuances of neurodegenerative diseases. Therefore, this review aims to summarise the current literature on the roles of gut dysbiosis and psychiatric disorders in pathological protein-related neurodegenerative diseases. We discussed how changes in the gut environment can influence the development of psychiatric symptoms and the progression of neurodegeneration and how these features overlap in AD and PD. Moreover, research on the interplay between gut dysbiosis, psychiatric disorders, and neurodegeneration remains in its early phase. In this review, we highlighted potential therapeutic approaches aimed at mitigating gastrointestinal problems and psychiatric disorders to alter the rate of neurodegeneration. Further research to assess the molecular mechanisms underlying AD and PD pathogenesis remains crucial for developing more effective treatments and achieving earlier diagnoses. Moreover, exploring non-invasive, early preventive measures and interventions is a relatively unexplored but important avenue of research in neurodegenerative diseases.
Collapse
Affiliation(s)
- Charlotte R. Denman
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Republic of Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Junghyun Jo
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Republic of Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
119
|
Goetzl EJ. An emerging spectrum of therapeutic targets for Alzheimer's disease. FASEB J 2023; 37:e23238. [PMID: 37795882 DOI: 10.1096/fj.202301461r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/16/2023] [Accepted: 09/21/2023] [Indexed: 10/06/2023]
Abstract
Development of effective preventative and therapeutic measures for Alzheimer's disease has been unsuccessful because of the long and subtly symptomatic preclinical period, difficulties obtaining tissue and biochemical data from living patients, and the many complex underlying pathogenic processes. Recent applications of sensitive specific bioimaging techniques, analyses of RNAs and proteins of neural cell-derived extracellular vesicles in blood, and sophisticated genetic procedures in cellular and rodent models have yielded hopeful new therapeutic targets. These newer targets are described here in relation to their neural cellular location, potential genetic modifications and possible pharmacological approaches.
Collapse
Affiliation(s)
- Edward J Goetzl
- Department of Medicine, University of California Medical Center, San Francisco, California, USA
- Research Department, Campus for Jewish Living, San Francisco, California, USA
| |
Collapse
|
120
|
Wang YTT, Rosa-Neto P, Gauthier S. Advanced brain imaging for the diagnosis of Alzheimer disease. Curr Opin Neurol 2023; 36:481-490. [PMID: 37639461 DOI: 10.1097/wco.0000000000001198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
PURPOSE OF REVIEW The purpose is to review the latest advances of brain imaging for the diagnosis of Alzheimer disease (AD). RECENT FINDINGS Brain imaging techniques provide valuable and complementary information to support the diagnosis of Alzheimer disease in clinical and research settings. The recent FDA accelerated approvals of aducanumab, lecanemab and donanemab made amyloid-PET critical in helping determine the optimal window for anti-amyloid therapeutic interventions. Tau-PET, on the other hand, is considered of key importance for the tracking of disease progression and for monitoring therapeutic interventions in clinical trials. PET imaging for microglial activation, astrocyte reactivity and synaptic degeneration are still new techniques only used in the research field, and more studies are needed to validate their use in the clinical diagnosis of AD. Finally, artificial intelligence has opened new prospective in the early detection of AD using MRI modalities. SUMMARY Brain imaging techniques using PET improve our understanding of the different AD-related pathologies and their relationship with each other along the course of disease. With more robust validation, machine learning and deep learning algorithms could be integrated with neuroimaging modalities to serve as valuable tools for clinicians to make early diagnosis and prognosis of AD.
Collapse
|
121
|
The Neuroscientist Comments. Neuroscientist 2023; 29:515-516. [PMID: 37615103 DOI: 10.1177/10738584231191753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
|
122
|
Ferreira PCL, Therriault J, Tissot C, Ferrari-Souza JP, Benedet AL, Povala G, Bellaver B, Leffa DT, Brum WS, Lussier FZ, Bezgin G, Servaes S, Vermeiren M, Macedo AC, Cabrera A, Stevenson J, Triana-Baltzer G, Kolb H, Rahmouni N, Klunk WE, Lopez O, Villemagne VL, Cohen A, Tudorascu DL, Zimmer ER, Karikari TK, Ashton NJ, Zetterberg H, Blennow K, Gauthier S, Rosa-Neto P, Pascoal TA. Plasma p-tau231 and p-tau217 inform on tau tangles aggregation in cognitively impaired individuals. Alzheimers Dement 2023; 19:4463-4474. [PMID: 37534889 PMCID: PMC10592380 DOI: 10.1002/alz.13393] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 08/04/2023]
Abstract
INTRODUCTION Phosphorylated tau (p-tau) biomarkers have been recently proposed to represent brain amyloid-β (Aβ) pathology. Here, we evaluated the plasma biomarkers' contribution beyond the information provided by demographics (age and sex) to identify Aβ and tau pathologies in individuals segregated as cognitively unimpaired (CU) and impaired (CI). METHODS We assessed 138 CU and 87 CI with available plasma p-tau231, 217+ , and 181, Aβ42/40, GFAP and Aβ- and tau-PET. RESULTS In CU, only plasma p-tau231 and p-tau217+ significantly improved the performance of the demographics in detecting Aβ-PET positivity, while no plasma biomarker provided additional information to identify tau-PET positivity. In CI, p-tau217+ and GFAP significantly contributed to demographics to identify both Aβ-PET and tau-PET positivity, while p-tau231 only provided additional information to identify tau-PET positivity. DISCUSSION Our results support plasma p-tau231 and p-tau217+ as state markers of early Aβ deposition, but in later disease stages they inform on tau tangle accumulation. HIGHLIGHTS It is still unclear how much plasma biomarkers contribute to identification of AD pathology across the AD spectrum beyond the information already provided by demographics (age + sex). Plasma p-tau231 and p-tau217+ contribute to demographic information to identify brain Aβ pathology in preclinical AD. In CI individuals, plasma p-tau231 contributes to age and sex to inform on the accumulation of tau tangles, while p-tau217+ and GFAP inform on both Aβ deposition and tau pathology.
Collapse
Affiliation(s)
- Pamela C. L Ferreira
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Cécile Tissot
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - João Pedro Ferrari-Souza
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Andréa L. Benedet
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
| | - Guilherme Povala
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Bruna Bellaver
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Douglas T. Leffa
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Wagner S. Brum
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
| | - Firoza Z. Lussier
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Marie Vermeiren
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Arthur C. Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Arlec Cabrera
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Gallen Triana-Baltzer
- Neuroscience Biomarkers, Janssen Research and Development, La Jolla, CA, 92121-1126, USA
| | - Hartmuth Kolb
- Neuroscience Biomarkers, Janssen Research and Development, La Jolla, CA, 92121-1126, USA
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - William E. Klunk
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Oscar Lopez
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Victor L. Villemagne
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Ann Cohen
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Dana L. Tudorascu
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Eduardo R. Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeuctis, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
- Brain Insitute of Rio Grande do Sul, PUCRS, Porto Alegre, 90619-900, Brazil
| | - Thomas K. Karikari
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, 431 41, Sweden
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, WC1N 3BG, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1N 3BG, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, HKG, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
| | - Serge Gauthier
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
| | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, QC H4H 1R3, Canada
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Tharick A. Pascoal
- Department of Psychiatry, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
123
|
Hickman LB, Stern JM, Silverman DHS, Salamon N, Vossel K. Clinical, imaging, and biomarker evidence of amyloid- and tau-related neurodegeneration in late-onset epilepsy of unknown etiology. Front Neurol 2023; 14:1241638. [PMID: 37830092 PMCID: PMC10565489 DOI: 10.3389/fneur.2023.1241638] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/05/2023] [Indexed: 10/14/2023] Open
Abstract
Accumulating evidence suggests amyloid and tau-related neurodegeneration may play a role in development of late-onset epilepsy of unknown etiology (LOEU). In this article, we review recent evidence that epilepsy may be an initial manifestation of an amyloidopathy or tauopathy that precedes development of Alzheimer's disease (AD). Patients with LOEU demonstrate an increased risk of cognitive decline, and patients with AD have increased prevalence of preceding epilepsy. Moreover, investigations of LOEU that use CSF biomarkers and imaging techniques have identified preclinical neurodegeneration with evidence of amyloid and tau deposition. Overall, findings to date suggest a relationship between acquired, non-lesional late-onset epilepsy and amyloid and tau-related neurodegeneration, which supports that preclinical or prodromal AD is a distinct etiology of late-onset epilepsy. We propose criteria for assessing elevated risk of developing dementia in patients with late-onset epilepsy utilizing clinical features, available imaging techniques, and biomarker measurements. Further research is needed to validate these criteria and assess optimal treatment strategies for patients with probable epileptic preclinical AD and epileptic prodromal AD.
Collapse
Affiliation(s)
- L. Brian Hickman
- Mary S. Easton Center for Alzheimer’s Research and Care, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Neurology, UCLA Seizure Disorder Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - John M. Stern
- Department of Neurology, UCLA Seizure Disorder Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Daniel H. S. Silverman
- Mary S. Easton Center for Alzheimer’s Research and Care, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Noriko Salamon
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Keith Vossel
- Mary S. Easton Center for Alzheimer’s Research and Care, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
124
|
Mastrangelo A, Vacchiano V, Zenesini C, Ruggeri E, Baiardi S, Cherici A, Avoni P, Polischi B, Santoro F, Capellari S, Liguori R, Parchi P. Amyloid-Beta Co-Pathology Is a Major Determinant of the Elevated Plasma GFAP Values in Amyotrophic Lateral Sclerosis. Int J Mol Sci 2023; 24:13976. [PMID: 37762278 PMCID: PMC10531493 DOI: 10.3390/ijms241813976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/29/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Recent studies reported increased plasma glial acidic fibrillary protein (GFAP) levels in amyotrophic lateral sclerosis (ALS) patients compared to controls. We expanded these findings in a larger cohort, including 156 ALS patients and 48 controls, and investigated the associations of plasma GFAP with clinical variables and other biofluid biomarkers. Plasma GFAP and Alzheimer's disease (AD) cerebrospinal fluid (CSF) biomarkers were assessed by the single molecule array and the Lumipulse platforms, respectively. In ALS patients, plasma GFAP was higher than in controls (p < 0.001) and associated with measures of cognitive decline. Twenty ALS patients (12.8%) showed a positive amyloid status (A+), of which nine also exhibited tau pathology (A+T+, namely ALS-AD). ALS-AD patients showed higher plasma GFAP than A- ALS participants (p < 0.001) and controls (p < 0.001), whereas the comparison between A- ALS and controls missed statistical significance (p = 0.07). Plasma GFAP distinguished ALS-AD subjects more accurately (area under the curve (AUC) 0.932 ± 0.027) than plasma p-tau181 (AUC 0.692 ± 0.058, p < 0.0001) and plasma neurofilament light chain protein (AUC, 0.548 ± 0.088, p < 0.0001). Cognitive measures differed between ALS-AD and other ALS patients. AD co-pathology deeply affects plasma GFAP values in ALS patients. Plasma GFAP is an accurate biomarker for identifying AD co-pathology in ALS, which can influence the cognitive phenotype.
Collapse
Affiliation(s)
- Andrea Mastrangelo
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, 40139 Bologna, Italy; (A.M.); (S.B.); (P.A.); (S.C.); (R.L.)
| | - Veria Vacchiano
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Corrado Zenesini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Edoardo Ruggeri
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Simone Baiardi
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, 40139 Bologna, Italy; (A.M.); (S.B.); (P.A.); (S.C.); (R.L.)
| | - Arianna Cherici
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Patrizia Avoni
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, 40139 Bologna, Italy; (A.M.); (S.B.); (P.A.); (S.C.); (R.L.)
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Barbara Polischi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Francesca Santoro
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Sabina Capellari
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, 40139 Bologna, Italy; (A.M.); (S.B.); (P.A.); (S.C.); (R.L.)
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Rocco Liguori
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, 40139 Bologna, Italy; (A.M.); (S.B.); (P.A.); (S.C.); (R.L.)
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Piero Parchi
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, 40139 Bologna, Italy; (A.M.); (S.B.); (P.A.); (S.C.); (R.L.)
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| |
Collapse
|
125
|
Liu T, Zuo H, Ma D, Song D, Zhao Y, Cheng O. Cerebrospinal fluid GFAP is a predictive biomarker for conversion to dementia and Alzheimer's disease-associated biomarkers alterations among de novo Parkinson's disease patients: a prospective cohort study. J Neuroinflammation 2023; 20:167. [PMID: 37475029 PMCID: PMC10357612 DOI: 10.1186/s12974-023-02843-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/27/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Dementia is a prevalent non-motor manifestation among individuals with advanced Parkinson's disease (PD). Glial fibrillary acidic protein (GFAP) is an inflammatory marker derived from astrocytes. Research has demonstrated the potential of plasma GFAP to forecast the progression to dementia in PD patients with mild cognitive impairment (PD-MCI). However, the predictive role of cerebrospinal fluid (CSF) GFAP on future cognitive transformation and alterations in Alzheimer's disease (AD)-associated CSF biomarkers in newly diagnosed PD patients has not been investigated. METHODS 210 de novo PD patients from the Parkinson's Progression Markers Initiative were recruited. Cognitive progression in PD participants was evaluated using Cox regression. Cross-sectional and longitudinal associations between baseline CSF GFAP and cognitive function and AD-related CSF biomarkers were evaluated using multiple linear regression and generalized linear mixed model. RESULTS At baseline, the mean age of PD participants was 60.85 ± 9.78 years, including 142 patients with normal cognition (PD-NC) and 68 PD-MCI patients. The average follow-up time was 6.42 ± 1.69 years. A positive correlation was observed between baseline CSF GFAP and age (β = 0.918, p < 0.001). There was no statistically significant difference in baseline CSF GFAP levels between PD-NC and PD-MCI groups. Higher baseline CSF GFAP predicted greater global cognitive decline over time in early PD patients (Montreal Cognitive Assessment, β = - 0.013, p = 0.014). Furthermore, Cox regression showed that high baseline CSF GFAP levels were associated with a high risk of developing dementia over an 8-year period in the PD-NC group (adjusted HR = 3.070, 95% CI 1.119-8.418, p = 0.029). In addition, the baseline CSF GFAP was positively correlated with the longitudinal changes of not only CSF α-synuclein (β = 0.313, p < 0.001), but also CSF biomarkers associated with AD, namely, amyloid-β 42 (β = 0.147, p = 0.034), total tau (β = 0.337, p < 0.001) and phosphorylated tau (β = 0.408, p < 0.001). CONCLUSIONS CSF GFAP may be a valuable prognostic tool that can predict the severity and progression of cognitive deterioration, accompanied with longitudinal changes in AD-associated pathological markers in early PD.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Hongzhou Zuo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Di Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Dan Song
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Yuying Zhao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Oumei Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| |
Collapse
|