1
|
Beltrami S, Rizzo S, Schiuma G, Cianci G, Narducci M, Baroni M, Di Luca D, Rizzo R, Bortolotti D. West Nile virus non-structural protein 1 promotes amyloid Beta deposition and neurodegeneration. Int J Biol Macromol 2025; 305:141032. [PMID: 39954900 DOI: 10.1016/j.ijbiomac.2025.141032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Recent observations highlight a notable surge in West Nile Virus (WNV) infections in Europe that can lead to neuroinvasive consequences associated with neurodegeneration, mainly triggered by WNV Non-Structural protein 1 (NS1). During viral replication, various protein-protein interactions take place, allowing viral proteins to interact with host factors. NS1 is actively secreted in the bloodstream by infected cells and is known to affect endothelial permeability and host immune response. Focusing on the recently discovered antimicrobial roles of Amyloid-Beta (Aβ) in the context Central Nervous System (CNS), we connected WNV late pathology to overlapping features encountered in neurodegenerative diseases. In fact, CNS viral infections, or presence of specific viral components, activate glial cells, which in turn increase Aβ expression as an antiviral mechanism, leading to Aβ accumulation and neuronal damage. Considering West Nile neuroinvasive disease (WNND) as a possible complication of WNV infection, we investigated the impact of soluble WNV (s)NS1 on glial and neuronal cells, in 2D and 3D in vitro models. We reported an increased Aβ deposition after WNV sNS1 treatment, particularly of Aβ-142 isoform, and increased glial activation with a subsequent neurotoxicity. These findings underscore the crucial role of sNS1 in CNS-related effects during WNV infection, suggesting a novel pathogenetic role.
Collapse
Affiliation(s)
- Silvia Beltrami
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara 44121, Italy.
| | - Sabrina Rizzo
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara 44121, Italy.
| | - Giovanna Schiuma
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara 44121, Italy.
| | - Giorgia Cianci
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara 44121, Italy.
| | - Marco Narducci
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara 44121, Italy; Temple University, Japan Campus, 1 Chome-14-29 Taishido, Setagaya City, Tokyo 154-0004, Japan.
| | - Marcello Baroni
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara 44121, Italy.
| | - Dario Di Luca
- Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy.
| | - Roberta Rizzo
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara 44121, Italy.
| | - Daria Bortolotti
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara 44121, Italy.
| |
Collapse
|
2
|
Metzler LAP, Metzger JM, Gerred KJ, Emborg ME, Kapoor A. Expression patterns of blood-based biomarkers of neurodegeneration and inflammation across adulthood in rhesus macaques. Exp Gerontol 2025; 203:112736. [PMID: 40122475 PMCID: PMC11998483 DOI: 10.1016/j.exger.2025.112736] [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: 12/20/2024] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
As the global human population rapidly ages and diseases of aging become more prevalent, preclinical models of age-related neurodegenerative disorders are increasingly important for identifying early diagnostic biomarkers, monitoring disease progression, and evaluating treatment responsiveness. Rhesus macaques are an ideal species for studies on neurodegeneration due to their phylogenetic relatedness to humans and their complex brain anatomy and physiology. Technological advances in assay sensitivity have facilitated the identification of blood-based biomarkers of neurodegeneration and inflammation in human populations. The aim of this study was to translate these methods for use in male and female rhesus macaques across adulthood. We collected plasma samples from 47 rhesus macaques representing pre-adult (1-5 years, n = 6 female, n = 5 male), young (5-7 years, n = 5 female, n = 7 male), middle (8-16 years, n = 7 female, n = 7 male), and older adult (17-22 years, n = 6 female, n = 4 male) subjects. Quantified biomarkers included neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), amyloid beta (Aβ42, Aβ40, and their ratio), total tau, phosphorylated tau (pTau181), interleukin (IL) 2, IL-6, IL-8, and IL-10. Plasma NfL and IL-6 levels were significantly increased with age in both sexes, with a marked rise during middle adulthood. The ratio of Aβ42/Aβ40 was significantly lower in the middle and older aged females compared to the youngest group. There was no effect of age or sex on total tau or pTau181 levels. Overall, these results demonstrate the feasibility of evaluating blood biomarkers of neurodegeneration and inflammation in rhesus macaques during adulthood.
Collapse
Affiliation(s)
- Ludwig A P Metzler
- Wisconsin National Primate Research Center, University of Wisconsin Madison, Madison, WI 53715, USA.
| | - Jeanette M Metzger
- Wisconsin National Primate Research Center, University of Wisconsin Madison, Madison, WI 53715, USA.
| | - Keenan J Gerred
- Wisconsin National Primate Research Center, University of Wisconsin Madison, Madison, WI 53715, USA.
| | - Marina E Emborg
- Wisconsin National Primate Research Center, University of Wisconsin Madison, Madison, WI 53715, USA; Department of Medical Physics, University of Wisconsin Madison, Madison, WI 53715, USA.
| | - Amita Kapoor
- Wisconsin National Primate Research Center, University of Wisconsin Madison, Madison, WI 53715, USA.
| |
Collapse
|
3
|
Singh-Reilly N, Graff-Radford J, Li D, Mielke MM, Machulda MM, Schwarz CG, Senjem ML, Jack CR, Lowe VJ, Josephs KA, Whitwell JL. Aβ 42/40 and p-tau 181 as disease biomarkers in atypical Alzheimer's disease. J Alzheimers Dis 2025:13872877251333450. [PMID: 40267266 DOI: 10.1177/13872877251333450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
BackgroundStudies suggest that plasma Alzheimer's disease (AD) biomarkers may aid in the overall diagnosis of AD, but their utility among patients with atypical clinical presentations of AD are unknown.ObjectiveThe main objective of this study was to determine the relationship between amyloid-β (Aβ) and tau plasma biomarkers and PET measures of both Aβ and tau in atypical AD. The secondary objective was to determine if plasma biomarkers could differentiate patients with different atypical AD phenotypes and whether they were related to measures of disease severity.MethodsWe assessed whether plasma p-tau 181 and Aβ42/40 were associated with Aβ and tau PET uptake, clinical phenotype and severity in 77 patients with PET biomarker-confirmed atypical AD.ResultsPlasma Aβ42/40 ratio showed positive associations with tau PET uptake, with higher (more normal) Aβ42/40 ratio associated with higher tau uptake; the ratio was not associated with Aβ PET. No associations were noted with plasma p-tau 181. Plasma Aβ42/40 ratio and p-tau 181 concentrations were not associated with AD phenotype or cognitive severity.ConclusionPlasma Aβ42/40 ratio and p-tau 181 concentrations are not associated with amyloid or tau PET or with clinical severity among individuals presenting with atypical AD.
Collapse
Affiliation(s)
| | | | - Danni Li
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Wake Forest University, Winston-Salem, NC, USA
| | - Mary M Machulda
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
4
|
Kim H, Kulshreshtha A, Alonso A, Goldstein FC, Johnson EC, Gold ME, Quyyumi AA, Lah JJ. The association between pulse wave velocity and cerebrospinal fluid biomarkers for Alzheimer's disease. J Alzheimers Dis 2025:13872877251331235. [PMID: 40255037 DOI: 10.1177/13872877251331235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
We examined the association between arterial stiffness using non-invasive pulse wave velocity (PWV) and cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarkers. We conducted a cross-sectional multivariate logistic regression analysis using established cut-off values for PWV and CSF biomarkers. Of the 739 participants, 69% were female, 84% were White, 12% were Black, and the mean age was 62. After adjustment for potential confounders, participants with high PWV had 94% (OR = 1.94, 95% CI 1.20-3.20) greater odds of AD biomarker positivity for tTau/Aβ42 and 108% (OR = 2.08, 95% Cl, 1.27-3.46) for pTau181/Aβ42. Our results suggest that higher arterial stiffness is associated with AD CSF biomarker positivity.
Collapse
Affiliation(s)
- Hyena Kim
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Ambar Kulshreshtha
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Family and Preventive Medicine, Emory School of Medicine, Atlanta, GA, USA
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Felicia C Goldstein
- Department of Neurology, Emory School of Medicine, Atlanta, GA, USA
- Emory Goizueta Alzheimer's Disease Research Center, Emory University, Atlanta, GA, USA
| | - Erik Cb Johnson
- Department of Neurology, Emory School of Medicine, Atlanta, GA, USA
- Emory Goizueta Alzheimer's Disease Research Center, Emory University, Atlanta, GA, USA
| | - Matthew E Gold
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory School of Medicine, Atlanta, GA, USA
| | - Arshed A Quyyumi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory School of Medicine, Atlanta, GA, USA
| | - James J Lah
- Department of Neurology, Emory School of Medicine, Atlanta, GA, USA
- Emory Goizueta Alzheimer's Disease Research Center, Emory University, Atlanta, GA, USA
| |
Collapse
|
5
|
Butler HM, Keller E, McCrorey M, Keceli G, Combs CK, Kayed R, Namakkal-S R, Paolocci N, Jacobs Wolf B, Wold LE, Del Monte F. Particulate matter and co-occurring genetic risk induce oxidative stress and cardiac and brain Alzheimer's pathology. Commun Biol 2025; 8:603. [PMID: 40221628 PMCID: PMC11993720 DOI: 10.1038/s42003-025-07701-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 02/07/2025] [Indexed: 04/14/2025] Open
Abstract
Amyloid-beta (Aβ) aggregates, an Alzheimer's disease (AD) pathological hallmark, extend beyond the brain to the heart of heart failure (HF) and AD patients. Being diseases of the elderly, increased prevalence is expected as the population ages. However, changes in the incidence and prevalence of dementia over the past decades, and the independent association of exposure to air particulate matter (PM) with poor cognitive function, adverse cardiovascular effects, and oxidative stress hint to the contribution of other factors beyond senescence. Therefore we evaluate whether, and by which mechanism(s), PM exposure affects heart and brain proteinopathy with/without genetic predisposition.AD-prone and control mice are exposed for three months to filtered air (FA) or concentrated ambient PM < 2.5μm in diameter (PM2.5), and evaluated for Aβ pathology, cognitive and cardiac function, and markers of oxidative stress. Aβ pathology become noticeable in AD hearts and worsens with PM2.5 in AD brains. Functionally, PM2.5 lead to anxiety and memory deficits and worsens diastolic function. Redox homeostasis is negatively impacted by genotype and PM2.5. This study identifies environmental pollution as a potential key contributor to early progression of heart and brain proteinopathy, delineating a crucial timepoint for early interventions to limit multiorgan damage in vulnerable patients.
Collapse
Affiliation(s)
- Helen M Butler
- College of Graduate Studies, Medical University of South Carolina, Charleston, SC, USA
| | - Everette Keller
- College of Medicine, Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Marice McCrorey
- College of Graduate Studies, Medical University of South Carolina, Charleston, SC, USA
| | - Gizem Keceli
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - Colin K Combs
- School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Rakez Kayed
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rajasekaran Namakkal-S
- Department of Pathology/Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nazareno Paolocci
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - Bethany Jacobs Wolf
- College of Medicine, Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Loren E Wold
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Federica Del Monte
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC, USA.
- Department of Medicine and Surgery, University of Bologna Alma Mater, Bologna, Italy.
| |
Collapse
|
6
|
Farhan R, Hashmi SA, Kapur J, D'Abreu A, Punia V, Manning C, Smith VL, Zawar I. Exploring biomarkers of neurodegeneration in epilepsy: Critical insights. Epileptic Disord 2025. [PMID: 40197800 DOI: 10.1002/epd2.70023] [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: 12/16/2024] [Revised: 02/26/2025] [Accepted: 03/25/2025] [Indexed: 04/10/2025]
Abstract
The advent of biofluid biomarkers for neurodegenerative disorders has precipitated a surge in recent evidence regarding their role in epilepsy. In this literature review, we examine the diagnostic, prognostic, and therapeutic potential of several biomarkers, including amyloid-beta (Aβ) protein, total (t-tau), phosphorylated tau (p-tau) protein, alpha-synuclein, neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), and others in epilepsy. Recent studies highlight mid-life Aβ levels as a risk factor for late-onset epilepsy. Several studies also show that amyloid pathology correlates with cognitive impairment in people with epilepsy (PWE). T-tau and p-tau levels in CSF and serum show diagnostic potential, particularly for temporal lobe epilepsy (TLE). Tau may also have significant prognostic utility in cognition of PWE and status epilepticus. Despite promising findings, larger prospective studies are needed to validate these biomarkers for routine clinical use in older PWE. Mouse models demonstrate tau's association with increased seizure susceptibility and mortality and the association of tau reduction with reduced seizure severity. This further highlights the need to investigate tau-targeting therapies in future studies in older PWE. Recent small-scale retrospective studies link NfL's role in cognitive impairment and status epilepticus, suggest a prognostic role of alpha-synuclein in certain epilepsies, and propose emerging diagnostic and prognostic roles of other biomarkers in epilepsy, including GFAP, cytoskeletal proteins, and S100B. However, larger longitudinal studies are needed to confirm these findings. We propose integrating some of these biomarkers into clinical practice for selected older adults with epilepsy. This integration could improve diagnostic accuracy, prognosticate outcomes, and identify therapeutic targets that may improve seizure control and mitigate the progression of cognitive decline in PWE.
Collapse
Affiliation(s)
| | - Syeda Amrah Hashmi
- Department of Neurology, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Jaideep Kapur
- Department of Neurology, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
- Brain Institute, University of Virginia, Charlottesville, Virginia, USA
| | - Anelyssa D'Abreu
- Department of Neurology, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Vineet Punia
- Epilepsy Division, Department of Neurology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Carol Manning
- Department of Neurology, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Vanessa L Smith
- Neuropathology Division, Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Ifrah Zawar
- Department of Neurology, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
7
|
Fries GR, De La Garza S, Zhao NO, Bass AW, Lima CNC, Kobori N, Barichello T, Turecki G, Schulz PE, Diniz BS, Soares JC. Association between epigenetic aging acceleration and amyloid biomarkers in bipolar disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.06.25325186. [PMID: 40297439 PMCID: PMC12036414 DOI: 10.1101/2025.04.06.25325186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Objectives Bipolar disorder (BD) has been associated with an elevated risk of Alzheimer's Disease (AD). We assessed AD biomarkers in BD and tested whether epigenetic aging (EA) acceleration is a potential mechanism driving variability in these markers. Design Setting Participants Cross-sectional study of n=59 living individuals with BD and n=20 age- and sex-equated control participants, as well as analyses of postmortem brain samples (Brodmann area 9/46) from n=46 individuals with BD. Measurements Amyloid beta (Aβ) 40 , Aβ 42 , and total Tau levels were measured in plasma from individuals with BD and controls, and Aβ 42 levels were measured in brains. EA and its acceleration (blood: GrimAge and DunedinPACE; brains: DNAmClock Cortical ) were estimated for all samples. Individuals with BD were split into quartiles with accelerated or slower EA if they were in the first or fourth quartiles for GrimAge acceleration (AgeAccelGrim), DunedinPACE, or DNAmClock Cortical acceleration (DNAmClock Cortical Accel). Results Individuals with BD showed an increase in Aβ 40 (p=.049) and a decrease in the Aβ 42/40 ratio (p=.035) compared to controls. A decrease in the Aβ 42/40 ratio was also found in individuals with BD with high versus low AgeAccelGrim (p=.028). Brain Aβ 42 levels significantly correlated with DNAmClock Cortical Accel (r 2 =.270, p=.007), with those with high EA acceleration showing higher brain Aβ 42 after controlling for confounders (p=.008). Conclusions Our results provide preliminary evidence that EA may explain the variability in AD risk in individuals with BD and could act as a target for preventing dementia and AD in BD.
Collapse
|
8
|
Schöll M, Vrillon A, Ikeuchi T, Quevenco FC, Iaccarino L, Vasileva-Metodiev SZ, Burnham SC, Hendrix J, Epelbaum S, Zetterberg H, Palmqvist S. Cutting through the noise: A narrative review of Alzheimer's disease plasma biomarkers for routine clinical use. J Prev Alzheimers Dis 2025; 12:100056. [PMID: 39814656 DOI: 10.1016/j.tjpad.2024.100056] [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: 10/31/2024] [Revised: 12/16/2024] [Accepted: 12/30/2024] [Indexed: 01/18/2025]
Abstract
As novel, anti-amyloid therapies have become more widely available, access to timely and accurate diagnosis has become integral to ensuring optimal treatment of patients with early-stage Alzheimer's disease (AD). Plasma biomarkers are a promising tool for identifying AD pathology; however, several technical and clinical factors need to be considered prior to their implementation in routine clinical use. Given the rapid pace of advancements in the field and the wide array of available biomarkers and tests, this review aims to summarize these considerations, evaluate available platforms, and discuss the steps needed to bring plasma biomarker testing to the clinic. We focus on plasma phosphorylated(p)-tau, specifically plasma p-tau217, as a robust candidate across both primary and secondary care settings. Despite the high performance and robustness demonstrated in research, plasma p-tau217, like all plasma biomarkers, can be affected by analytical and pre-analytical variability as well as patient comorbidities, sex, ethnicity, and race. This review also discusses the advantages of the two-point cut-off approach to mitigating these factors, and the challenges raised by the resulting intermediate range measurements, where clinical guidance is still unclear. Further validation of plasma p-tau217 in heterogeneous, real-world cohorts will help to increase confidence in testing and support establishing a standardized approach. Plasma biomarkers are poised to become a more affordable and less invasive alternative to PET and CSF testing. However, understanding the factors that impact plasma biomarker measurement and interpretation is critical prior to their implementation in routine clinical use.
Collapse
Affiliation(s)
- M Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK; Department of Neuropsychiatry, Sahlgrenska University Hospital, Mölndal, Sweden
| | - A Vrillon
- French Institute of Health and Medical Research (Inserm), Paris, France
| | - T Ikeuchi
- Niigata University Brain Research Institute, Niigata, Japan
| | - F C Quevenco
- Eli Lilly and Company, Indianapolis, IN, United States
| | - L Iaccarino
- Eli Lilly and Company, Indianapolis, IN, United States
| | | | - S C Burnham
- Eli Lilly and Company, Indianapolis, IN, United States
| | - J Hendrix
- Eli Lilly and Company, Indianapolis, IN, United States
| | - S Epelbaum
- Eli Lilly and Company, Indianapolis, IN, United States
| | - H Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, 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
| | - S Palmqvist
- Clinical Memory Research Unit, Clinical Sciences in Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Sweden.
| |
Collapse
|
9
|
Sullivan AC, Zuniga G, Ramirez P, Fernandez R, Wang CP, Li J, Davila L, Pelton K, Gomez S, Sohn C, Gonzalez E, Lopez-Cruzan M, Gonzalez DA, Parker A, Zilli E, de Erausquin GA, Seshadri S, Espinoza S, Musi N, Frost B. A Phase IIa clinical trial to evaluate the effects of anti-retroviral therapy in Alzheimer's disease (ART-AD). NPJ DEMENTIA 2025; 1:2. [PMID: 40104524 PMCID: PMC11917871 DOI: 10.1038/s44400-024-00001-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/09/2024] [Indexed: 03/20/2025]
Abstract
Retrotransposons constitute over 40% of the human genome. Studies in Drosophila, mice, cultured cells, and human brain show that retrotransposons are activated in tauopathies, including Alzheimer's disease, and causally drive neurodegeneration. The reverse transcriptase inhibitor 3TC (lamivudine) reduces retrotransposon activation and suppresses tau neurotoxicity among model systems. This phase 2a open-label trial (Pilot Study to Investigate the Safety and Feasibility of Anti-Retroviral Therapy for Alzheimer's Disease, NCT04552795, registered 09/10/2020) followed 12 participants with early Alzheimer's disease (MMSE > 24, CDR = 0.5) over 24 weeks to assess safety, tolerability, and feasibility of daily 300 mg 3TC treatment. The sample was well-educated (12-20 years) and culturally diverse (25% from underrepresented groups). In addition to a favorable safety profile and stable cognitive measures, notable significant changes in fluid-based biomarkers include reduction of glial fibrillary acidic protein (GFAP) (P = 0.03) in CSF, suggestive of reduced neuroinflammation, and elevation of Aβ42/40 (P = 0.009) in plasma, suggestive of reduced plaque load in the brain. These results warrant further exploration in a larger, placebo-controlled trial.
Collapse
Affiliation(s)
- A. Campbell Sullivan
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Neurology, University of Texas Health San Antonio, San Antonio, TX USA
| | - Gabrielle Zuniga
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX USA
| | - Paulino Ramirez
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX USA
- Brown University Center for Alzheimer’s Disease Research, Providence, RI USA
| | - Roman Fernandez
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, TX USA
| | - Chen-Pin Wang
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, TX USA
| | - Ji Li
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX USA
| | - Lisa Davila
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX USA
- Brown University Center for Alzheimer’s Disease Research, Providence, RI USA
| | - Kristine Pelton
- Brown University Center for Alzheimer’s Disease Research, Providence, RI USA
| | - Sandra Gomez
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Claira Sohn
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX USA
| | - Elias Gonzalez
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX USA
| | - Marisa Lopez-Cruzan
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Psychiatry and Behavioral Sciences, University of Texas Health San Antonio, Department of Neurological Sciences, San Antonio, TX USA
| | - David A. Gonzalez
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Neurology, University of Texas Health San Antonio, San Antonio, TX USA
- Rush University Medical Center, Chicago, IL USA
| | - Alicia Parker
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Neurology, University of Texas Health San Antonio, San Antonio, TX USA
| | - Eduardo Zilli
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Neurology, University of Texas Health San Antonio, San Antonio, TX USA
| | - Gabriel A. de Erausquin
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Neurology, University of Texas Health San Antonio, San Antonio, TX USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Neurology, University of Texas Health San Antonio, San Antonio, TX USA
| | - Sara Espinoza
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Nicolas Musi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Bess Frost
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX USA
- Brown University Center for Alzheimer’s Disease Research, Providence, RI USA
| |
Collapse
|
10
|
Safi A, Giunti E, Melikechi O, Xia W, Melikechi N. Identification of blood plasma protein ratios for distinguishing Alzheimer's disease from healthy controls using machine learning. Heliyon 2025; 11:e42349. [PMID: 39981365 PMCID: PMC11840181 DOI: 10.1016/j.heliyon.2025.e42349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/22/2025] Open
Abstract
Early detection of Alzheimer's disease is essential for effective treatment and the development of therapies that modify disease progression. Developing sensitive and specific noninvasive diagnostic tools is crucial for improving clinical outcomes and advancing our understanding of this condition. Liquid biopsy techniques, especially those involving plasma biomarkers, provide a promising noninvasive method for early diagnosis and disease monitoring. In this study, we analyzed the plasma proteomic profiles of 38 healthy individuals, with an average age of 66.5 years, and 22 patients with Alzheimer's disease, with an average age of 79.7 years. Proteins in the plasma were quantified using specialized panels designed for proteomic extension assays. Through computational analysis using a linear support vector machine algorithm, we identified 82 differentially expressed proteins between the two groups. From these, we calculated 6642 possible protein ratios and identified specific combinations of these ratios as significant features for distinguishing between individuals with Alzheimer's disease and healthy individuals. Notably, the protein ratios kynureninase to macrophage scavenger receptor type 1, Neurocan to protogenin, and interleukin-5 receptor alpha to glial cell line-derived neurotrophic factor receptor alpha 1 achieving accuracy up to 98 % in differentiating between the two groups. This study underscores the potential of leveraging protein relationships, expressed as ratios, in advancing Alzheimer's disease diagnostics. Furthermore, our findings highlight the promise of liquid biopsy techniques as a noninvasive and accurate approach for early detection and monitoring of Alzheimer's disease using blood plasma.
Collapse
Affiliation(s)
- Ali Safi
- Kennedy College of Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA
| | - Elisa Giunti
- Bedford VA Healthcare System, Bedford, MA, 01730, USA
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Omar Melikechi
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Weiming Xia
- Kennedy College of Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA
- Bedford VA Healthcare System, Bedford, MA, 01730, USA
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Noureddine Melikechi
- Kennedy College of Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA
| |
Collapse
|
11
|
Yang S, Zhang X, Du X, Yan P, Zhang J, Wang W, Wang J, Zhang L, Sun H, Liu Y, Xu X, Di Y, Zhong J, Wu C, Reinhardt JD, Zheng Y, Wu T. Prediction of cognitive conversion within the Alzheimer's disease continuum using deep learning. Alzheimers Res Ther 2025; 17:41. [PMID: 39948600 PMCID: PMC11823042 DOI: 10.1186/s13195-025-01686-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/28/2025] [Indexed: 02/16/2025]
Abstract
BACKGROUND Early diagnosis and accurate prognosis of cognitive decline in Alzheimer's disease (AD) is important to timely assignment to optimal treatment modes. We aimed to develop a deep learning model to predict cognitive conversion to guide re-assignment decisions to more intensive therapies where needed. METHODS Longitudinal data including five variable sets, i.e. demographics, medical history, neuropsychological outcomes, laboratory and neuroimaging results, from the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort were analyzed. We first developed a deep learning model to predicted cognitive conversion using all five variable sets. We then gradually removed variable sets to obtained parsimonious models for four different years of forecasting after baseline within acceptable frames of reduction in overall model fit (AUC remaining > 0.8). RESULTS A total of 607 individuals were included at baseline, of whom 538 participants were followed up at 12 months, 482 at 24 months, 268 at 36 months and 280 at 48 months. Predictive performance was excellent with AUCs ranging from 0.87 to 0.92 when all variable sets were considered. Parsimonious prediction models that still had a good performance with AUC 0.80-0.84 were established, each only including two variable sets. Neuropsychological outcomes were included in all parsimonious models. In addition, biomarker was included at year 1 and year 2, imaging data at year 3 and demographics at year 4. Under our pre-set threshold, the rate of upgrade to more intensive therapies according to predicted cognitive conversion was always higher than according to actual cognitive conversion so as to decrease the false positive rate, indicating the proportion of patients who would have missed upgraded treatment based on prognostic models although they actually needed it. CONCLUSIONS Neurophysiological tests combined with other indicator sets that vary along the AD continuum can improve can provide aid for clinical treatment decisions leading to improved management of the disease. TRAIL REGISTRATION INFORMATION ClinicalTrials.gov Identifier: NCT00106899 (Registration Date: 31 March 2005).
Collapse
Affiliation(s)
- Siyu Yang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of Neurology, Nanjing Second Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210003, China
| | - Xintong Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xinyu Du
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Peng Yan
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jing Zhang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Wei Wang
- Department of Neurology, Nanjing Second Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210003, China
| | - Jing Wang
- China-Australia Joint Research Centre for Infectious Diseases, School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shanxi, 710061, China
| | - Lei Zhang
- China-Australia Joint Research Centre for Infectious Diseases, School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shanxi, 710061, China
- Artificial Intelligence and Modelling in Epidemiology Program, Melbourne Sexual Health Centre, Alfred Health, Melbourne, Australia
- Central Clinical School, Faculty of Medicine, Monash University, Melbourne, Australia
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Huaiqing Sun
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Yin Liu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Xinran Xu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Yaxuan Di
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jin Zhong
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Caiyun Wu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jan D Reinhardt
- Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, Sichuan, 610207, China.
- Swiss Paraplegic Research, Nottwil, Switzerland.
- Department of Health Sciences and Medicine, University of Lucerne, Lucerne, Switzerland.
| | - Yu Zheng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Ting Wu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
12
|
Angelopoulou E, Androni X, Villa C, Hatzimanolis A, Scarmeas N, Papageorgiou S. Blood-based biomarkers in mild behavioral impairment: an updated overview. Front Neurol 2025; 16:1534193. [PMID: 39980634 PMCID: PMC11839432 DOI: 10.3389/fneur.2025.1534193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/24/2025] [Indexed: 02/22/2025] Open
Abstract
Identifying individuals at-risk for dementia is one of the critical objectives of current research efforts, highlighting the need for simple, cost-effective, and minimally invasive biomarkers. Mild behavioral impairment (MBI), characterized by the emergence of persistent neuropsychiatric manifestations in older adults, has attracted increasing attention as a potential early indicator of cognitive decline and dementia. A growing number of studies have recently begun to explore the relationship between MBI and several blood-based biomarkers associated with Alzheimer's disease (AD) pathology, neurodegeneration, as well as systemic metabolic and inflammatory dysregulation. In this context, MBI has been associated with lower plasma Aβ42/Αβ40 ratio, higher plasma phosphorylated tau at threonine 181 (p-tau181), increased neurofilament light chain (NfL) levels, as well as disturbances in metabolic markers, including homocysteine, insulin and ferritin, suggesting a multifaceted neurobiological basis for this syndrome. These findings offer insights into the underlying pathophysiology of MBI, and connection between neuropsychiatric symptoms and progression of AD. In this narrative review, we aim to summarize and critically discuss the emerging literature evidence linking MBI to blood-based biomarkers, hoping to shed more light on MBI's pathophysiology, its connection to AD-related neurobiology, as well as its potential practical utility for predicting cognitive impairment, guiding early interventions and managing the risk for dementia.
Collapse
Affiliation(s)
- Efthalia Angelopoulou
- 1st Department of Neurology, Aiginiteio University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Xenia Androni
- 1st Department of Neurology, Aiginiteio University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Alexandros Hatzimanolis
- 1st Department of Psychiatry, Aiginiteio University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginiteio University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Sokratis Papageorgiou
- 1st Department of Neurology, Aiginiteio University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
13
|
Rudolph MD, Sutphen CL, Register TC, Lockhart SN, Rundle MM, Hughes TM, Bateman JR, Solingapuram Sai KK, Whitlow CT, Craft S, Mielke MM. Evaluation of plasma p-tau217 for detecting amyloid pathology in a diverse and heterogeneous community-based cohort. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.20.25320851. [PMID: 39974029 PMCID: PMC11838994 DOI: 10.1101/2025.01.20.25320851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
INTRODUCTION Studies suggest excellent performance of plasma p-tau217 for detecting amyloid pathology, though studies in more diverse populations are needed to validate previously determined cutpoints. METHODS Plasma p-tau217 utility for detecting amyloid pathology (Aβ) via amyloid PET (n=598) and/or cerebrospinal fluid (CSF; n=154) was assessed in a heterogeneous, community-based cohort in the Wake Forest Alzheimer's Disease Research Center (WFADRC). Participants (n=598) were 21% Black; 313 cognitive unimpaired (CU), 214 mild cognitive impairment (MCI), and 64 dementia (DEM); 49% prediabetic, 44% hypertensive; 29% overweight/obese; and 64% with mild-to-moderate kidney disease. Gaussian-mixture models, logistic regression, and receiver operating curve analyses were performed. RESULTS Plasma p-tau217 was associated with elevated Aβ deposition and accurately classified Aβ-positive participants (PET: AUC: 94%-97%, cutpoint≥.338 pg/mL; CSF: AUC = .84, cutpoint≥.307 pg/mL). DISCUSSION Plasma p-tau217 is an accurate indicator of amyloid pathology in a heterogeneous cohort, and superior to other plasma biomarkers assessed. Longitudinal analyses assessing impact of comorbidities on p-tau217 utility for disease progression are underway.
Collapse
Affiliation(s)
- Marc D. Rudolph
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Courtney L. Sutphen
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Thomas C. Register
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Samuel N. Lockhart
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Melissa M. Rundle
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Timothy M. Hughes
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - James R. Bateman
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Kiran K. Solingapuram Sai
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Christopher T. Whitlow
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Suzanne Craft
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Michelle M. Mielke
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| |
Collapse
|
14
|
Jungwon J, Lee JH, Choi CH, Lee J. DTI-ALPS index as a predictor of cognitive decline over 1 year. Neuroradiology 2025; 67:163-170. [PMID: 39680094 DOI: 10.1007/s00234-024-03521-w] [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: 07/08/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline and impaired daily functioning. The glymphatic system removes neurotoxic waste, including amyloid-beta (Aβ), an important factor in AD pathogenesis. This study used the Diffusion Tensor Imaging Analysis Along the Perivascular Space (DTI-ALPS) index, which reflects glymphatic function, to explore its relationship with cognitive decline in patients with probable AD. METHODS We conducted a longitudinal study of 16 participants aged 60-79 years with probable AD who were evaluated using the Clinical Dementia Rating (CDR) and Mini-Mental State Examination (MMSE). Glymphatic function was assessed using the DTI-ALPS index; plasma Aβ 42/40 ratios were measured to account for amyloid pathology. The relationship between the DTI-ALPS index and baseline cognitive function was analyzed using multiple regression models adjusted for age, sex, and plasma Aβ 42/40 ratios. Associations between the DTI-ALPS index and cognitive decline over 1 year were assessed by a model using the percentage change in the MMSE z-score as the outcome variable. RESULTS Higher DTI-ALPS index was significantly associated with better baseline cognitive function as assessed by MMSE (standardized beta = 1.17, p < 0.001) and lower clinical severity as assessed by CDR (standardized beta = - 1.00, p = 0.006). Over the 1-year follow-up, greater baseline DTI-ALPS index values were associated with less cognitive decline (standardized beta = - 0.85, p = 0.018). CONCLUSION Our findings suggest that DTI-ALPS index is associated with cognitive performance and is a biomarker for predicting cognitive decline in AD. Future studies should consider larger sample sizes and longer follow-up periods to validate these findings.
Collapse
Affiliation(s)
- Joo Jungwon
- Department of Psychiatry, Chungbuk National University Hospital, 1 Sunhwan-ro, Cheongju, 28644, Republic of Korea
| | - Ji Hyung Lee
- Department of Psychiatry, Chungbuk National University Hospital, 1 Sunhwan-ro, Cheongju, 28644, Republic of Korea
| | - Chi-Hoon Choi
- Department of Radiology, Chungbuk National University College of Medicine, 1Chungdae-ro, Cheongju, 28644, South Korea
| | - Jeonghwan Lee
- Department of Psychiatry, Chungbuk National University Hospital, 1 Sunhwan-ro, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
15
|
Salunkhe J, Ugale R. Recent updates on immunotherapy in neurodegenerative diseases. Brain Res 2024; 1845:149205. [PMID: 39197568 DOI: 10.1016/j.brainres.2024.149205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/01/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
Neurodegeneration is a progressive event leading to specific neuronal loss due to the accumulation of aberrant proteins. These pathologic forms of proteins further worsen and interfere with normal physiologic mechanisms, which can lead to abnormal proliferation of immune cells and subsequent inflammatory cascades and ultimately neuronal loss. Recently, immunotherapies targeting abnormal, pathologic forms of protein have shown a promising approach to modify the progression of neurodegeneration. Recent advances in immunotherapy have led to the development of novel antibodies against the proteinopathies which can eradicate aggregations of protein as evident from preclinical and clinical studies. Nonetheless, only a few of them have successfully received clinical approval, while others have been discontinued due to a lack of clinical efficacy endpoints. The current review discusses the status of investigational antibodies under clinical trials, their targets for therapeutic action, and evidence for failure or success.
Collapse
Affiliation(s)
- Jotiram Salunkhe
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, India
| | - Rajesh Ugale
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, India.
| |
Collapse
|
16
|
Li L, Wei N, Guo Y, Zhu X, Wang L, Zhu Y, Fang K, Ma S, Zhang Y, Zhang Y, Zhou X, Zhao G, Bu Y, Zhou L. Detection of Aβ40 in cerebrospinal fluid and plasma of Alzheimer's disease patients using photoelectrochemical biosensors. Mikrochim Acta 2024; 192:5. [PMID: 39630274 DOI: 10.1007/s00604-024-06816-0] [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: 08/06/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
An ultra-sensitive photoelectrochemical (PEC) biosensor for amyloid-beta 40 (Aβ40), a biomarker for Alzheimer's disease (AD), was developed using g-C₃N₄ modified with gold nanoparticles (Au NPs) to form Au-C₃N₄. This was further combined with TiO₂ to create a tightly bonded TiO₂/Au-C₃N₄ heterojunction, leading to a highly responsive photocatalytic process. Furthermore, the incorporation of noble metal Au NPs not only enhances photocurrent generation but also securely immobilizes the aptamer through Au-S bonds, providing additional surface binding sites. This significantly increases the sensor's capture efficiency. The sensor exhibited excellent performance, featuring a linear detection range from 10-15 to 10-11 g/mL and a remarkably low detection limit (LOD) of 0.33 fg/mL. Moreover, the validation in clinical settings demonstrated the successful detection in real cerebrospinal fluid (CSF) and plasma, from AD patients and non-AD controls. These results strongly suggest that PEC biosensors possess significant potential as cost-effective and highly sensitive tools for detecting ultra-trace substances in human body fluids, which offers promising opportunities for the early screening of high-risk populations for AD.
Collapse
Affiliation(s)
- Liting Li
- Department of Neurology, Northwest University First Hospital, Xi'an, 710043, China
- The College of Life Sciences, Northwest University, Xi'an, 710068, China
| | - Na Wei
- The College of Life Sciences, Northwest University, Xi'an, 710068, China
| | - Yiwei Guo
- Key Laboratory of Wide Band-Gap Semiconductor Materials and Devices, School of Microelectronics, Xidian University, Xi'an, 710071, China
| | - Xingyu Zhu
- The College of Life Sciences, Northwest University, Xi'an, 710068, China
| | - Lin Wang
- Key Laboratory of Wide Band-Gap Semiconductor Materials and Devices, School of Microelectronics, Xidian University, Xi'an, 710071, China
| | - Yanzhao Zhu
- Department of Neurology, Northwestern University School of Medicine, Xi'an, 710068, China
| | - Ke Fang
- Key Laboratory of Wide Band-Gap Semiconductor Materials and Devices, School of Microelectronics, Xidian University, Xi'an, 710071, China
| | - Shenhui Ma
- Key Laboratory of Wide Band-Gap Semiconductor Materials and Devices, School of Microelectronics, Xidian University, Xi'an, 710071, China
| | - Yingying Zhang
- The College of Life Sciences, Northwest University, Xi'an, 710068, China
| | - Yan Zhang
- The College of Life Sciences, Northwest University, Xi'an, 710068, China
| | - Xiaofei Zhou
- The College of Life Sciences, Northwest University, Xi'an, 710068, China
| | - Gang Zhao
- Department of Neurology, Northwest University First Hospital, Xi'an, 710043, China.
- Department of Neurology, Northwestern University School of Medicine, Xi'an, 710068, China.
| | - Yuyu Bu
- Key Laboratory of Wide Band-Gap Semiconductor Materials and Devices, School of Microelectronics, Xidian University, Xi'an, 710071, China.
| | - Linfu Zhou
- Department of Neurology, Northwest University First Hospital, Xi'an, 710043, China.
- Department of Neurology, Northwestern University School of Medicine, Xi'an, 710068, China.
| |
Collapse
|
17
|
An C, Cai H, Ren Z, Fu X, Quan S, Jia L. Biofluid biomarkers for Alzheimer's disease: past, present, and future. MEDICAL REVIEW (2021) 2024; 4:467-491. [PMID: 39664082 PMCID: PMC11629312 DOI: 10.1515/mr-2023-0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 09/04/2024] [Indexed: 12/13/2024]
Abstract
Alzheimer's disease (AD) is a gradually progressive neurodegenerative disease with tremendous social and economic burden. Therefore, early and accurate diagnosis is imperative for effective treatment or prevention of the disease. Cerebrospinal fluid and blood biomarkers emerge as favorable diagnostic tools due to their relative accessibility and potential for widespread clinical use. This review focuses on the AT(N) biomarker system, which includes biomarkers reflecting AD core pathologies, amyloid deposition, and pathological tau, as well as neurodegeneration. Novel biomarkers associated with inflammation/immunity, synaptic dysfunction, vascular pathology, and α-synucleinopathy, which might contribute to either the pathogenesis or the clinical progression of AD, have also been discussed. Other emerging candidates including non-coding RNAs, metabolites, and extracellular vesicle-based markers have also enriched the biofluid biomarker landscape for AD. Moreover, the review discusses the current challenges of biofluid biomarkers in AD diagnosis and offers insights into the prospective future development.
Collapse
Affiliation(s)
- Chengyu An
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Huimin Cai
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Ziye Ren
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Xiaofeng Fu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shuiyue Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| |
Collapse
|
18
|
Doke R, Lamkhade GJ, Vinchurkar K, Singh S. Demystifying the Role of Neuroinflammatory Mediators as Biomarkers for Diagnosis, Prognosis, and Treatment of Alzheimer's Disease: A Review. ACS Pharmacol Transl Sci 2024; 7:2987-3003. [PMID: 39416969 PMCID: PMC11475310 DOI: 10.1021/acsptsci.4c00457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
Neuroinflammatory mediators play a pivotal role in the pathogenesis of Alzheimer's Disease (AD), influencing its onset, progression, and severity. The precise mechanisms behind AD are still not fully understood, leading current treatments to focus mainly on managing symptoms rather than preventing or curing the condition. The amyloid and tau hypotheses are the most widely accepted explanations for AD pathology; however, they do not completely account for the neuronal degeneration observed in AD. Growing evidence underscores the crucial role of neuroinflammation in the pathology of AD. The neuroinflammatory hypothesis presents a promising new approach to understanding the mechanisms driving AD. This review examines the importance of neuroinflammatory biomarkers in the diagnosis, prognosis, and treatment of AD. It delves into the mechanisms underlying neuroinflammation in AD, highlighting the involvement of various mediators such as cytokines, chemokines, and ROS. Additionally, this review discusses the potential of neuroinflammatory biomarkers as diagnostic tools, prognostic indicators, and therapeutic targets for AD management. By understanding the intricate interplay between neuroinflammation and AD pathology, this review aims to help in the development of efficient diagnostic and treatment plans to fight this debilitating neurological condition. Furthermore, it elaborates recent advancements in neuroimaging techniques and biofluid analysis for the identification and monitoring of neuroinflammatory biomarkers in AD patients.
Collapse
Affiliation(s)
- Rohit
R. Doke
- Jaihind
College of Pharmacy, Vadgaon Sahani, Pune, Maharashtra 412401, India
| | | | - Kuldeep Vinchurkar
- Krishna
School of Pharmacy, Kiran and Pallavi Patel
Global University, Vadodara, Gujarat 391243, India
| | - Sudarshan Singh
- Office
of Research Administration, Chiang Mai University, Chaing Mai 50200, Thailand
- Faculty
of Pharmacy, Chiang Mai University, Chaing Mai 50200, Thailand
| |
Collapse
|
19
|
Wugalter KA, Schroeder RA, Thurston RC, Wu M, Aizenstein HJ, Cohen AD, Kamboh MI, Karikari TK, Derby CA, Maki PM. Associations of endogenous estrogens, plasma Alzheimer's disease biomarkers, and APOE4 carrier status on regional brain volumes in postmenopausal women. Front Aging Neurosci 2024; 16:1426070. [PMID: 39044806 PMCID: PMC11263297 DOI: 10.3389/fnagi.2024.1426070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/18/2024] [Indexed: 07/25/2024] Open
Abstract
Background Women carrying the APOE4 allele are at greater risk of developing Alzheimer's disease (AD) from ages 65-75 years compared to men. To better understand the elevated risk conferred by APOE4 carrier status among midlife women, we investigated the separate and interactive associations of endogenous estrogens, plasma AD biomarkers, and APOE4 carrier status on regional brain volumes in a sample of late midlife postmenopausal women. Methods Participants were enrolled in MsBrain, a cohort study of postmenopausal women (n = 171, mean age = 59.4 years, mean MoCA score = 26.9; race = 83.2% white, APOE4 carriers = 40). Serum estrone (E1) and estradiol (E2) levels were assessed using liquid chromatography-tandem mass spectrometry. APOE genotype was determined using TaqMan SNP genotyping assays. Plasma AD biomarkers were measured using single molecule array technology. Cortical volume was measured and segmented by FreeSurfer software using individual T1w MPRAGE images. Multiple linear regression models were conducted to determine whether separate and interactive associations between endogenous estrogen levels, plasma AD biomarkers (Aβ42/Aβ40, Aβ42/p-tau181), and APOE4 carrier status predict regional brain volume (21 regions per hemisphere, selected a priori); and, whether significant interactive associations between estrogens and AD biomarkers on brain volume differed by APOE4 carrier status. Results There was no main effect of APOE4 carrier status on regional brain volumes, endogenous estrogen levels, or plasma AD biomarkers. Estrogens did not associate with regional brain volumes, except for positive associations with left caudal middle frontal gyrus and fusiform volumes. The interactive association of estrogens and APOE4 carrier status on brain volume was not significant for any region. The interactive association of estrogens and plasma AD biomarkers predicted brain volume of several regions. Higher E1 and E2 were more strongly associated with greater regional brain volumes among women with a poorer AD biomarker profile (lower Aβ42/40, lower Aβ42/p-tau181 ratios). In APOE4-stratified analyses, these interactions were driven by non-APOE4 carriers. Conclusion We demonstrate that the brain volumes of postmenopausal women with poorer AD biomarker profiles benefit most from higher endogenous estrogen levels. These findings are driven by non-APOE4 carriers, suggesting that APOE4 carriers may be insensitive to the favorable effects of estrogens on brain volume in the postmenopause.
Collapse
Affiliation(s)
- Katrina A. Wugalter
- Department of Psychology, University of Illinois Chicago, Chicago, IL, United States
| | - Rachel A. Schroeder
- Department of Psychology, University of Illinois Chicago, Chicago, IL, United States
| | - Rebecca C. Thurston
- Departments of Psychiatry, Epidemiology, Psychology, and Clinical and Translational Science, University of Pittsburgh, Pittsburgh, PA, United States
| | - Minjie Wu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Howard J. Aizenstein
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ann D. Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - M. Ilyas Kamboh
- Departments of Psychiatry, Human Genetics, and Epidemiology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Thomas K. Karikari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Carol A. Derby
- The Saul R. Korey Department of Neurology, Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Pauline M. Maki
- Departments of Psychiatry, Psychology and Obstetrics & Gynecology, University of Illinois Chicago, Chicago, IL, United States
| |
Collapse
|
20
|
Singh NA, Alnobani A, Graff‐Radford J, Machulda MM, Mielke MM, Schwarz CG, Senjem ML, Jack CR, Lowe VJ, Kanekiyo T, Josephs KA, Whitwell JL. Relationships between PET and blood plasma biomarkers in corticobasal syndrome. Alzheimers Dement 2024; 20:4765-4774. [PMID: 38885334 PMCID: PMC11247700 DOI: 10.1002/alz.13914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 06/20/2024]
Abstract
INTRODUCTION Corticobasal syndrome (CBS) can result from underlying Alzheimer's disease (AD) pathologies. Little is known about the utility of blood plasma metrics to predict positron emission tomography (PET) biomarker-confirmed AD in CBS. METHODS A cohort of eighteen CBS patients (8 amyloid beta [Aβ]+; 10 Aβ-) and 8 cognitively unimpaired (CU) individuals underwent PET imaging and plasma analysis. Plasma concentrations were compared using a Kruskal-Wallis test. Spearman correlations assessed relationships between plasma concentrations and PET uptake. RESULTS CBS Aβ+ group showed a reduced Aβ42/40 ratio, with elevated phosphorylated tau (p-tau)181, glial fibrillary acidic protein (GFAP), and neurofilament light (NfL) concentrations, while CBS Aβ- group only showed elevated NfL concentration compared to CU. Both p-tau181 and GFAP were able to differentiate CBS Aβ- from CBS Aβ+ and showed positive associations with Aβ and tau PET uptake. DISCUSSION This study supports use of plasma p-tau181 and GFAP to detect AD in CBS. NfL shows potential as a non-specific disease biomarker of CBS regardless of underlying pathology. HIGHLIGHTS Plasma phosphorylated tau (p-tau)181 and glial fibrillary acidic protein (GFAP) concentrations differentiate corticobasal syndrome (CBS) amyloid beta (Aβ)- from CBS Aβ+. Plasma neurofilament light concentrations are elevated in CBS Aβ- and Aβ+ compared to controls. Plasma p-tau181 and GFAP concentrations were associated with Aβ and tau positron emission tomography (PET) uptake. Aβ42/40 ratio showed a negative correlation with Aβ PET uptake.
Collapse
Affiliation(s)
| | - Alla Alnobani
- Department of Neuroscience, Mayo ClinicJacksonvilleFloridaUSA
| | | | - Mary M. Machulda
- Department of Psychiatry & Psychology, Mayo ClinicRochesterMinnesotaUSA
| | - Michelle M. Mielke
- Department of Epidemiology and PreventionWake Forest UniversityWinston‐SalemNorth CarolinaUSA
| | | | | | | | - Val J. Lowe
- Department of RadiologyMayo ClinicRochesterMinnesotaUSA
| | | | | | | |
Collapse
|
21
|
Zhang Y, Ferreira PCL, Jacobsen E, Bellaver B, Pascoal TA, Snitz BE, Chang CH, Villemagne VL, Ganguli M, Karikari TK. Association of plasma biomarkers of Alzheimer's disease and related disorders with cognition and cognitive decline: The MYHAT population-based study. Alzheimers Dement 2024; 20:4199-4211. [PMID: 38753951 PMCID: PMC11180930 DOI: 10.1002/alz.13829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION Plasma biomarkers of Alzheimer's disease and related dementias predict global cognitive performance and decline over time; it remains unclear how they associate with changes in different dementia syndromes affecting distinct cognitive domains. METHODS In a prospective study with repeated assessments of a randomly selected population-based cohort (n = 787, median age 73), we evaluated performance and decline in different cognitive domains over up to 8 years in relation to plasma concentrations of amyloid beta 42/40 (Aβ42/40) ratio, phosphorylated tau181 (p-tau181), neurofilament light chain (NfL), and glial fibrillary acidic protein (GFAP). RESULTS Cross-sectionally, memory showed the strongest associations with p-tau181, and attention, executive, and visuospatial functions with NfL. Longitudinally, memory decline was distinguishable with all biomarker profiles dichotomized according to data-driven cutoffs, most efficiently with Aβ42/40. GFAP and Aβ42/40 were the best discriminators of decline patterns in language and visuospatial functions, respectively. DISCUSSION These relatively non-invasive tests may be beneficial for clinical screening after replication in other populations and validation through neuroimaging or cerebrospinal fluid analysis. HIGHLIGHTS We performed a prospective study with up to 8 years of repeated domain-specific cognitive assessments and baseline plasma Alzheimer's disease and related dementias biomarker measurements in a randomly selected population-based cohort. We considered distinct growth curves of trajectories of different cognitive domains and survival bias induced by missing data by adding quadratic time and applying joint modeling technique. Cross-sectionally, memory showed the strongest associations with plasma phosphorylated tau181, while attention, executive, and visuospatial functions were most strongly associated with neurofilament light chain. Longitudinally, memory and visuospatial declines were most efficiently distinguished by dichotomized amyloid beta 42/40 profile among all plasma biomarkers, while language was by dichotomized glial fibrillary acidic protein. These relatively non-invasive tests may be beneficial for clinical screening; however, they will need replication in other populations and validation through neuroimaging and/or cerebrospinal fluid assessments.
Collapse
Affiliation(s)
- Yingjin Zhang
- Department of Biostatistics School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Pamela C. L. Ferreira
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Erin Jacobsen
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Bruna Bellaver
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Tharick A. Pascoal
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of NeurologySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Beth E. Snitz
- Department of NeurologySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Chung‐Chou H. Chang
- Department of Biostatistics School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of MedicineSchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Victor L. Villemagne
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Mary Ganguli
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of NeurologySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Epidemiology School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Thomas K. Karikari
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
22
|
Rudolph MD, Sutphen CL, Register TC, Whitlow CT, Solingapuram Sai KK, Hughes TM, Bateman JR, Dage JL, Russ KA, Mielke MM, Craft S, Lockhart SN. Associations among plasma, MRI, and amyloid PET biomarkers of Alzheimer's disease and related dementias and the impact of health-related comorbidities in a community-dwelling cohort. Alzheimers Dement 2024; 20:4159-4173. [PMID: 38747525 PMCID: PMC11180870 DOI: 10.1002/alz.13835] [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: 11/08/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 06/05/2024]
Abstract
INTRODUCTION We evaluated associations between plasma and neuroimaging-derived biomarkers of Alzheimer's disease and related dementias and the impact of health-related comorbidities. METHODS We examined plasma biomarkers (neurofilament light chain, glial fibrillary acidic protein, amyloid beta [Aβ] 42/40, phosphorylated tau 181) and neuroimaging measures of amyloid deposition (Aβ-positron emission tomography [PET]), total brain volume, white matter hyperintensity volume, diffusion-weighted fractional anisotropy, and neurite orientation dispersion and density imaging free water. Participants were adjudicated as cognitively unimpaired (CU; N = 299), mild cognitive impairment (MCI; N = 192), or dementia (DEM; N = 65). Biomarkers were compared across groups stratified by diagnosis, sex, race, and APOE ε4 carrier status. General linear models examined plasma-imaging associations before and after adjusting for demographics (age, sex, race, education), APOE ε4 status, medications, diagnosis, and other factors (estimated glomerular filtration rate [eGFR], body mass index [BMI]). RESULTS Plasma biomarkers differed across diagnostic groups (DEM > MCI > CU), were altered in Aβ-PET-positive individuals, and were associated with poorer brain health and kidney function. DISCUSSION eGFR and BMI did not substantially impact associations between plasma and neuroimaging biomarkers. HIGHLIGHTS Plasma biomarkers differ across diagnostic groups (DEM > MCI > CU) and are altered in Aβ-PET-positive individuals. Altered plasma biomarker levels are associated with poorer brain health and kidney function. Plasma and neuroimaging biomarker associations are largely independent of comorbidities.
Collapse
Affiliation(s)
- Marc D. Rudolph
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Courtney L. Sutphen
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Thomas C. Register
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Christopher T. Whitlow
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Kiran K. Solingapuram Sai
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Timothy M. Hughes
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - James R. Bateman
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Jeffrey L. Dage
- Department Of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kristen A. Russ
- Department Of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Michelle M. Mielke
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Suzanne Craft
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Samuel N. Lockhart
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
23
|
Nwamekang Belinga L, Espourteille J, Wepnyu Njamnshi Y, Zafack Zeukang A, Rouaud O, Kongnyu Njamnshi A, Allali G, Richetin K. Circulating Biomarkers for Alzheimer's Disease: Unlocking the Diagnostic Potential in Low- and Middle-Income Countries, Focusing on Africa. NEURODEGENER DIS 2024; 24:26-40. [PMID: 38555638 PMCID: PMC11251669 DOI: 10.1159/000538623] [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: 12/13/2023] [Accepted: 03/28/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is emerging as a significant public health challenge in Africa, with predictions indicating a tripling in incidence by 2050. The diagnosis of AD on the African continent is notably difficult, leading to late detection that severely limits treatment options and significantly impacts the quality of life for patients and their families. SUMMARY This review focuses on the potential of high-sensitivity specific blood biomarkers as promising tools for improving AD diagnosis and management globally, particularly in Africa. These advances are particularly pertinent in the continent, where access to medical and technical resources is often limited. KEY MESSAGES Identifying precise, sensitive, and specific blood biomarkers could contribute to the biological characterization and management of AD in Africa. Such advances promise to improve patient care and pave the way for new regional opportunities in pharmaceutical research and drug trials on the continent for AD.
Collapse
Affiliation(s)
- Luc Nwamekang Belinga
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Leenaards Memory Center, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Department of Translational Neuroscience, Brain Research Africa Initiative (BRAIN), Geneva, Switzerland
- Department of Translational Neuroscience, Brain Research Africa Initiative (BRAIN), Yaoundé, Cameroon
| | - Jeanne Espourteille
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Yembe Wepnyu Njamnshi
- Department of Translational Neuroscience, Brain Research Africa Initiative (BRAIN), Geneva, Switzerland
- Department of Translational Neuroscience, Brain Research Africa Initiative (BRAIN), Yaoundé, Cameroon
- Neuroscience Lab, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, Yaoundé, Cameroon
- Division of Health Operations Research, Ministry of Public Health, Yaoundé, Cameroon
| | - Ariole Zafack Zeukang
- Department of Translational Neuroscience, Brain Research Africa Initiative (BRAIN), Geneva, Switzerland
- Department of Translational Neuroscience, Brain Research Africa Initiative (BRAIN), Yaoundé, Cameroon
| | - Olivier Rouaud
- Leenaards Memory Center, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Alfred Kongnyu Njamnshi
- Department of Translational Neuroscience, Brain Research Africa Initiative (BRAIN), Geneva, Switzerland
- Department of Translational Neuroscience, Brain Research Africa Initiative (BRAIN), Yaoundé, Cameroon
- Neuroscience Lab, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, Yaoundé, Cameroon
- Department of Clinical Neuroscience and Neurology, Yaoundé Central Hospital, Yaoundé, Cameroon
| | - Gilles Allali
- Leenaards Memory Center, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Kevin Richetin
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Leenaards Memory Center, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
24
|
Mandal PK, Maroon JC, Garg A, Arora NK, Bansal R, Kaushik A, Samkaria A, Kumaran G, Arora Y. Blood Biomarkers in Alzheimer's Disease. ACS Chem Neurosci 2023; 14:3975-3978. [PMID: 37878665 PMCID: PMC10655041 DOI: 10.1021/acschemneuro.3c00641] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/27/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder that affects millions of people worldwide. The characteristic pathological manifestation of AD includes the deposition of extracellular insoluble β amyloid plaques and intracellular neurofibrillary tangles formed from hyperphosphorylated tau protein. Cost effective and minimally invasive peripheral blood-based biomarkers are critical for early AD diagnosis. Currently, the plasma based two fraction of β amyloid peptide ratio (Aβ42/40) and phosphorylated tau (p-tau) are considered as blood-based biomarkers for AD diagnosis. Recent research indicates that oxidative stress (OS) occurs prior to amyloid plaque (Aβ) formation and abnormal tau phosphorylation in AD. The imbalance of the master antioxidant, glutathione (GSH), and prooxidants (iron, zinc, and copper)─plays a crucial role in AD neurodegeneration. We present peripheral blood-based OS related biomarkers that are mechanistically involved in the disease process and may serve as a novel screening tool for early detection of AD onset. This OS based approach may also provide a quick and cost efficient method to monitor the effects of disease-modifying therapies in AD clinical trials.
Collapse
Affiliation(s)
- Pravat K. Mandal
- Neuroimaging
and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon 122052, India
- The
Florey Institute of Neuroscience and Mental Health, Melbourne School of Medicine Campus, Melbourne 3052, VIC, Australia
- Department
of Neurosurgery, University of Pittsburgh
Medical School, Pittsburgh, Pennsylvania 15213, United States
| | - Joseph C. Maroon
- Department
of Neurosurgery, University of Pittsburgh
Medical School, Pittsburgh, Pennsylvania 15213, United States
| | - Arun Garg
- Department
of Neurology, Medanta Medicity, Gurgaon 122001, Haryana, India
| | | | - Rishu Bansal
- Department
of Neurology, Medanta Medicity, Gurgaon 122001, Haryana, India
| | - Aditi Kaushik
- Neuroimaging
and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon 122052, India
| | - Avantika Samkaria
- Neuroimaging
and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon 122052, India
- Department
of Forensic Science, Chandigarh University, Mohali 160036, Punjab, India
| | - Gayathri Kumaran
- Neuroimaging
and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon 122052, India
| | - Yashika Arora
- Neuroimaging
and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon 122052, India
| |
Collapse
|
25
|
Manolopoulos A, Delgado-Peraza F, Mustapic M, Pucha KA, Nogueras-Ortiz C, Daskalopoulos A, Knight DD, Leoutsakos JM, Oh ES, Lyketsos CG, Kapogiannis D. Comparative assessment of Alzheimer's disease-related biomarkers in plasma and neuron-derived extracellular vesicles: a nested case-control study. Front Mol Biosci 2023; 10:1254834. [PMID: 37828917 PMCID: PMC10565036 DOI: 10.3389/fmolb.2023.1254834] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023] Open
Abstract
Introduction: Alzheimer's disease (AD) is currently defined according to biomarkers reflecting the core underlying neuropathological processes: Aβ deposition, Tau, and neurodegeneration (ATN). The soluble phase of plasma and plasma neuron-derived extracellular vesicles (NDEVs) are increasingly being investigated as sources of biomarkers. The aim of this study was to examine the comparative biomarker potential of these two biofluids, as well as the association between respective biomarkers. Methods: We retrospectively identified three distinct diagnostic groups of 44 individuals who provided samples at baseline and at a mean of 3.1 years later; 14 were cognitively unimpaired at baseline and remained so (NRM-NRM), 13 had amnestic MCI that progressed to AD dementia (MCI-DEM) and 17 had AD dementia at both timepoints (DEM-DEM). Plasma NDEVs were isolated by immunoaffinity capture targeting the neuronal markers L1CAM, GAP43, and NLGN3. In both plasma and NDEVs, we assessed ATN biomarkers (Aβ42, Aβ40, total Tau, P181-Tau) alongside several other exploratory markers. Results: The Aβ42/Aβ40 ratio in plasma and NDEVs was lower in MCI-DEM than NRM-NRM at baseline and its levels in NDEVs decreased over time in all three groups. Similarly, plasma and NDEV-associated Aβ42 was lower in MCI-DEM compared to NRM-NRM at baseline and its levels in plasma decreased over time in DEM-DEM. For NDEV-associated proBDNF, compared to NRM-NRM, its levels were lower in MCI-DEM and DEM-DEM at baseline, and they decreased over time in the latter group. No group differences were found for other exploratory markers. NDEV-associated Aβ42/Aβ40 ratio and proBDNF achieved the highest areas under the curve (AUCs) for discriminating between diagnostic groups, while proBDNF was positively associated with Mini-Mental State Examination (MMSE) score. No associations were found between the two biofluids for any assessed marker. Discussion: The soluble phase of plasma and plasma NDEVs demonstrate distinct biomarker profiles both at a single time point and longitudinally. The lack of association between plasma and NDEV measures indicates that the two types of biofluids demonstrate distinct biomarker signatures that may be attributable to being derived through different biological processes. NDEV-associated proBDNF may be a useful biomarker for AD diagnosis and monitoring.
Collapse
Affiliation(s)
- Apostolos Manolopoulos
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| | - Francheska Delgado-Peraza
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| | - Maja Mustapic
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| | - Krishna Ananthu Pucha
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| | - Carlos Nogueras-Ortiz
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| | - Alexander Daskalopoulos
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| | - De’Larrian DeAnté Knight
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| | - Jeannie-Marie Leoutsakos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Esther S. Oh
- Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Constantine G. Lyketsos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Dimitrios Kapogiannis
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, United States
| |
Collapse
|
26
|
Padala SP, Newhouse PA. Blood-based biomarkers in Alzheimer's disease: a mini-review. Metab Brain Dis 2023; 38:185-193. [PMID: 36342582 PMCID: PMC10688968 DOI: 10.1007/s11011-022-01114-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and has far reaching consequences for patients and their caregivers. Early detection and treatment are key factors in limiting the impact of the disease. However, a definitive diagnosis of AD requires an examination of brain tissue during an autopsy. Although a plethora of biomarkers such as neuroimaging, electrophysiological, and cerebrospinal fluid (CSF) biomarkers are available, their utility is limited to research due to their poor reach and prohibitive cost. In order for biomarkers to be widely used, they need to be accessible, affordable, and conducive for the patient population or disease stage. Blood-based biomarkers may not only be less expensive and more accessible compared to neuroimaging or CSF tests, but they are also preferred by patients with AD as they are much less invasive. In this mini-review article, we expand on the rationale for the use of blood-based biomarkers, review currently available biomarkers and discuss the need for the standardization of these biomarkers. We contrast the blood-based biomarkers with other available biomarkers and discuss the advantages of using a panel of blood-based biomarkers to strengthen their accuracy.
Collapse
Affiliation(s)
- Sanjana P Padala
- Vanderbilt University , 2301 Vanderbilt Place, Nashville, TN, 37235, USA.
| | - Paul A Newhouse
- Vanderbilt University , 2301 Vanderbilt Place, Nashville, TN, 37235, USA
- Center for Cognitive Medicine, Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, USA
- VA-TVHS Geriatric Research Education and Clinical Center, Nashville, TN, USA
| |
Collapse
|
27
|
Connor JP, Quinn SD, Schaefer C. Sticker-and-spacer model for amyloid beta condensation and fibrillation. Front Mol Neurosci 2022; 15:962526. [PMID: 36311031 PMCID: PMC9611774 DOI: 10.3389/fnmol.2022.962526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
A major pathogenic hallmark of Alzheimer's disease is the presence of neurotoxic plaques composed of amyloid beta (Aβ) peptides in patients' brains. The pathway of plaque formation remains elusive, though some clues appear to lie in the dominant presence of Aβ1 − 42 in these plaques despite Aβ1−40 making up approximately 90% of the Aβ pool. We hypothesize that this asymmetry is driven by the hydrophobicity of the two extra amino acids that are incorporated in Aβ1−42. To investigate this hypothesis at the level of single molecules, we have developed a molecular “sticker-and-spacer lattice model” of unfolded Aβ. The model protein has a single sticker that may reversibly dimerise and elongate into semi-flexible linear chains. The growth is hampered by excluded-volume interactions that are encoded by the hydrophilic spacers but are rendered cooperative by the attractive interactions of hydrophobic spacers. For sufficiently strong hydrophobicity, the chains undergo liquid-liquid phase-separation (LLPS) into condensates that facilitate the nucleation of fibers. We find that a small fraction of Aβ1−40 in a mixture of Aβ1−40 and Aβ1−42 shifts the critical concentration for LLPS to lower values. This study provides theoretical support for the hypothesis that LLPS condensates act as a precursor for aggregation and provides an explanation for the Aβ1−42-enrichment of aggregates in terms of hydrophobic interactions.
Collapse
Affiliation(s)
- Jack P. Connor
- Department of Biology, University of York, York, United Kingdom
- School of Physics, Engineering and Technology, University of York, York, United Kingdom
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
- *Correspondence: Jack P. Connor
| | - Steven D. Quinn
- School of Physics, Engineering and Technology, University of York, York, United Kingdom
- York Biomedical Research Institute, University of York, York, United Kingdom
| | - Charley Schaefer
- School of Physics, Engineering and Technology, University of York, York, United Kingdom
- Charley Schaefer
| |
Collapse
|
28
|
Zhang P, Zhou Y, Chen G, Li J, Wang B, Lu X. Potential association of bone mineral density loss with cognitive impairment and central and peripheral amyloid-β changes: a cross-sectional study. BMC Musculoskelet Disord 2022; 23:626. [PMID: 35773707 PMCID: PMC9245236 DOI: 10.1186/s12891-022-05580-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 06/21/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND There is some evidence in the literature that older adults with cognitive impairments have a higher risk for falls and osteoporotic hip fractures. Currently, the associations between bone health and cognitive health have not been extensively studied. Thus, the present cross-sectional study aims to investigate the relationship between markers of bone loss and cognitive performance in older adults with and without osteopenia as well as older adults with cognitive impairments (i.e., Alzheimer's disease [AD]). METHODS Sixty-two non-osteopenia participants and one hundred three osteopenia participants as the cohort 1 and 33 cognitively normal non-AD participants and 39 AD participants as the cohort 2 were recruited. To assess cognitive and bone health, hip bone mineral density (BMD) and cognitive performance (via Minimal Mental State Examination [MMSE] and/or Auditory Verbal Learning Test-delayed recall [AVLT-DR]) were assessed. Furthermore, in cohort 1, plasma amyloid-β (Aβ) levels, and in cohort 2, cerebrospinal fluid (CSF) Aβ levels were determined. RESULTS We observed that (1) compared with non-osteopenia participants, BMD values (t = - 22.806; 95%CI: - 1.801, - 1.484; p < 0.001), MMSE scores (t = - 5.392; 95%CI: - 3.260, - 1.698; p < 0.001), and AVLT-DR scores (t = - 4.142; 95%CI: - 2.181, - 0.804; p < 0.001), plasma Aβ42 levels (t = - 2.821; 95%CI: - 1.737, - 0.305; p = 0.01), and Aβ42/40 ratio (t = - 2.020; 95%CI: - 0.009, - 0.001; p = 0.04) were significantly lower in osteopenia participants; (2) plasma Aβ42/40 ratio showed a mediate effect for the association between BMD values and the performance of cognitive function in osteopenia participants by mediation analysis, adjusting age, sex, years of education, and body mass index (BMI); (3) BMD values (95%CI: - 1.085, 0.478; p < 0.001) were significantly reduced in AD participants as compared with cognitively normal non-AD participants; (4) in AD participants, the interactive effects of BMD and CSF Aβ42/40 ratio on MMSE scores was found by regression analysis, controlling age, sex, years of education, and BMI; (5) BMD can distinguish AD participants from cognitively normal non-AD participants with AUC of 0.816 and distinguish participants with the cognitive impairment from cognitively normal participants with AUC of 0.794. CONCLUSION Our findings suggest a relationship between bone health and cognitive health. Given the correlations between BMD and important markers of cognitive health (e.g., central and peripheral pathological change of Aβ), BMD might serve as a promising and easy-accessible biomarker. However, more research is needed to further substantiate our findings.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No. 136 Jingzhou Street, Xiangcheng District, Xiangyang, 441021, China
| | - Yi Zhou
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No. 136 Jingzhou Street, Xiangcheng District, Xiangyang, 441021, China
| | - Gang Chen
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No. 136 Jingzhou Street, Xiangcheng District, Xiangyang, 441021, China
| | - Jun Li
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No. 136 Jingzhou Street, Xiangcheng District, Xiangyang, 441021, China
| | - Bangjun Wang
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No. 136 Jingzhou Street, Xiangcheng District, Xiangyang, 441021, China
| | - Xinyan Lu
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No. 136 Jingzhou Street, Xiangcheng District, Xiangyang, 441021, China.
| |
Collapse
|
29
|
Noveir SD, Kerman BE, Xian H, Meuret C, Smadi S, Martinez AE, Johansson J, Zetterberg H, Parks BA, Kuklenyik Z, Mack WJ, Johansson JO, Yassine HN. Effect of the ABCA1 agonist CS-6253 on amyloid-β and lipoprotein metabolism in cynomolgus monkeys. Alzheimers Res Ther 2022; 14:87. [PMID: 35751102 PMCID: PMC9229758 DOI: 10.1186/s13195-022-01028-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 06/07/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Inducing brain ATP-binding cassette 1 (ABCA1) activity in Alzheimer's disease (AD) mouse models is associated with improvement in AD pathology. The purpose of this study was to investigate the effects of the ABCA1 agonist peptide CS-6253 on amyloid-β peptides (Aβ) and lipoproteins in plasma and cerebrospinal fluid (CSF) of cynomolgus monkeys, a species with amyloid and lipoprotein metabolism similar to humans. METHODS CS-6253 peptide was injected intravenously into cynomolgus monkeys at various doses in three different studies. Plasma and CSF samples were collected at several time points before and after treatment. Levels of cholesterol, triglyceride (TG), lipoprotein particles, apolipoproteins, and Aβ were measured using ELISA, ion-mobility analysis, and asymmetric-flow field-flow fractionation (AF4). The relationship between the change in levels of these biomarkers was analyzed using multiple linear regression models and linear mixed-effects models. RESULTS Following CS-6253 intravenous injection, within minutes, small plasma high-density lipoprotein (HDL) particles were increased. In two independent experiments, plasma TG, apolipoprotein E (apoE), and Aβ42/40 ratio were transiently increased following CS-6253 intravenous injection. This change was associated with a non-significant decrease in CSF Aβ42. Both plasma total cholesterol and HDL-cholesterol levels were reduced following treatment. AF4 fractionation revealed that CS-6253 treatment displaced apoE from HDL to intermediate-density- and low density-lipoprotein (IDL/LDL)-sized particles in plasma. In contrast to plasma, CS-6253 had no effect on the assessed CSF apolipoproteins or lipids. CONCLUSIONS Treatment with the ABCA1 agonist CS-6253 appears to favor Aβ clearance from the brain.
Collapse
Affiliation(s)
- Sasan D Noveir
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Bilal E Kerman
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Haotian Xian
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Cristiana Meuret
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Sabrina Smadi
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Ashley E Martinez
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Bryan A Parks
- Centers for Disease Control and Prevention, Atlanta, GA, 30341, USA
| | | | - Wendy J Mack
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, 90033, USA
| | | | - Hussein N Yassine
- Departments of Medicine and Neurology, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
30
|
Wang J, Gao L, Liu J, Dang L, Wei S, Hu N, Gao Y, Peng W, Shang S, Huo K, Wang J, Qu Q. The Association of Plasma Amyloid-β and Cognitive Decline in Cognitively Unimpaired Population. Clin Interv Aging 2022; 17:555-565. [PMID: 35480964 PMCID: PMC9035463 DOI: 10.2147/cia.s357994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022] Open
Abstract
Purpose This study investigates the relationship between baseline plasma Aβ and cognitive decline during follow-up in cognitively unimpaired population. Materials and Methods Cognitively unimpaired population was selected from people who lived in the suburbs of Xi’an, China. The levels of plasma Aβ1-42 and Aβ1-40 were tested using commercial enzyme-linked immunosorbent assay (ELISA). The mini-mental state examination (MMSE) and neuropsychological battery were used to assess cognition. Two years later, MMSE was tested again, and significant cognitive decline was defined as a decrease in MMSE scores ≥5 points. Logistic regression analysis was performed to analyze the relationship between baseline plasma Aβ and cognitive change during the two-year follow-up. Results A total of 1144 participants completed the study, among whom 59 subjects (5.2%) presented significant cognitive decline. The high plasma Aβ1-42 level group had more significant cognitive decline (P = 0.023). Multivariable logistic regression analysis showed that significant cognitive decline was associated with the high levels of baseline plasma Aβ1-42 (OR = 1.043, 95% CI: 1.005–1.083, P = 0.026). However, significant cognitive decline was not associated with baseline plasma Aβ1-40 levels and Aβ1-42 /Aβ1-40 ratio. Conclusion Population with high level of baseline plasma Aβ1-42 manifested significant cognitive decline over 2 years; however, further investigation on the dynamics of plasma Aβ and long-term follow-up are needed.
Collapse
Affiliation(s)
- Jin Wang
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Ling Gao
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Jie Liu
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Liangjun Dang
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Shan Wei
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Ningwei Hu
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yao Gao
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Wei Peng
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Suhang Shang
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Kang Huo
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Jingyi Wang
- Huyi Hospital of Traditional Chinese Medicine, Xi’an, People’s Republic of China
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Correspondence: Qiumin Qu, Department of Neurology, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an, 710061, People’s Republic of China, Tel/Fax +86 29 8532 4083, Email
| |
Collapse
|
31
|
Bogolepova A, Makhnovich E, Kovalenko E, Osinovskaya N. Potential biomarkers of early diagnosis of Alzheimer’s disease. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:7-14. [DOI: 10.17116/jnevro20221220917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
32
|
Bhargavan B, Woollard SM, McMillan JE, Kanmogne GD. CCR5 antagonist reduces HIV-induced amyloidogenesis, tau pathology, neurodegeneration, and blood-brain barrier alterations in HIV-infected hu-PBL-NSG mice. Mol Neurodegener 2021; 16:78. [PMID: 34809709 PMCID: PMC8607567 DOI: 10.1186/s13024-021-00500-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 11/03/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Neurocognitive impairment is present in 50% of HIV-infected individuals and is often associated with Alzheimer's Disease (AD)-like brain pathologies, including increased amyloid-beta (Aβ) and Tau hyperphosphorylation. Here, we aimed to determine whether HIV-1 infection causes AD-like pathologies in an HIV/AIDS humanized mouse model, and whether the CCR5 antagonist maraviroc alters HIV-induced pathologies. METHODS NOD/scid-IL-2Rγcnull mice engrafted with human blood leukocytes were infected with HIV-1, left untreated or treated with maraviroc (120 mg/kg twice/day). Human cells in animal's blood were quantified weekly by flow cytometry. Animals were sacrificed at week-3 post-infection; blood and tissues viral loads were quantified using p24 antigen ELISA, RNAscope, and qPCR. Human (HLA-DR+) cells, Aβ-42, phospho-Tau, neuronal markers (MAP 2, NeuN, neurofilament-L), gamma-secretase activating protein (GSAP), and blood-brain barrier (BBB) tight junction (TJ) proteins expression and transcription were quantified in brain tissues by immunohistochemistry, immunofluorescence, immunoblotting, and qPCR. Plasma Aβ-42, Aβ-42 cellular uptake, release and transendothelial transport were quantified by ELISA. RESULTS HIV-1 significantly decreased human (h)CD4+ T-cells and hCD4/hCD8 ratios; decreased the expression of BBB TJ proteins claudin-5, ZO-1, ZO-2; and increased HLA-DR+ cells in brain tissues. Significantly, HIV-infected animals showed increased plasma and brain Aβ-42 and phospho-Tau (threonine181, threonine231, serine396, serine199), associated with transcriptional upregulation of GSAP, an enzyme that catalyzes Aβ formation, and loss of MAP 2, NeuN, and neurofilament-L. Maraviroc treatment significantly reduced blood and brain viral loads, prevented HIV-induced loss of neuronal markers and TJ proteins; decreased HLA-DR+ cells infiltration in brain tissues, significantly reduced HIV-induced increase in Aβ-42, GSAP, and phospho-Tau. Maraviroc also reduced Aβ retention and increased Aβ release in human macrophages; decreased the receptor for advanced glycation end products (RAGE) and increased low-density lipoprotein receptor-related protein-1 (LRP1) expression in human brain endothelial cells. Maraviroc induced Aβ transendothelial transport, which was blocked by LRP1 antagonist but not RAGE antagonist. CONCLUSIONS Maraviroc significantly reduced HIV-induced amyloidogenesis, GSAP, phospho-Tau, neurodegeneration, BBB alterations, and leukocytes infiltration into the CNS. Maraviroc increased cellular Aβ efflux and transendothelial Aβ transport via LRP1 pathways. Thus, therapeutically targeting CCR5 could reduce viremia, preserve the BBB and neurons, increased brain Aβ efflux, and reduce AD-like neuropathologies.
Collapse
Affiliation(s)
- Biju Bhargavan
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, 985800 Nebraska Medical Center, Omaha, NE 68198-5800 USA
| | - Shawna M. Woollard
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, 985800 Nebraska Medical Center, Omaha, NE 68198-5800 USA
- Huvepharma, 421 W Industrial Lake Drive, Lincoln, NE 68528 USA
| | - Jo Ellyn McMillan
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, 985800 Nebraska Medical Center, Omaha, NE 68198-5800 USA
| | - Georgette D. Kanmogne
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, 985800 Nebraska Medical Center, Omaha, NE 68198-5800 USA
| |
Collapse
|
33
|
Pannee J, Shaw LM, Korecka M, Waligorska T, Teunissen CE, Stoops E, Vanderstichele HMJ, Mauroo K, Verberk IMW, Keshavan A, Pesini P, Sarasa L, Pascual‐Lucas M, Fandos N, Allué J, Portelius E, Andreasson U, Yoda R, Nakamura A, Kaneko N, Yang S, Liu H, Palme S, Bittner T, Mawuenyega KG, Ovod V, Bollinger J, Bateman RJ, Li Y, Dage JL, Stomrud E, Hansson O, Schott JM, Blennow K, Zetterberg H. The global Alzheimer's Association round robin study on plasma amyloid β methods. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12242. [PMID: 34692980 PMCID: PMC8515356 DOI: 10.1002/dad2.12242] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 07/07/2021] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Blood-based assays to measure brain amyloid beta (Aβ) deposition are an attractive alternative to the cerebrospinal fluid (CSF)-based assays currently used in clinical settings. In this study, we examined different blood-based assays to measure Aβ and how they compare among centers and assays. METHODS Aliquots from 81 plasma samples were distributed to 10 participating centers. Seven immunological assays and four mass-spectrometric methods were used to measure plasma Aβ concentrations. RESULTS Correlations were weak for Aβ42 while Aβ40 correlations were stronger. The ratio Aβ42/Aβ40 did not improve the correlations and showed weak correlations. DISCUSSION The poor correlations for Aβ42 in plasma might have several potential explanations, such as the high levels of plasma proteins (compared to CSF), sensitivity to pre-analytical sample handling and specificity, and cross-reactivity of different antibodies. Different methods might also measure different pools of plasma Aβ42. We, however, hypothesize that greater correlations might be seen in future studies because many of the methods have been refined during completion of this study.
Collapse
Affiliation(s)
- Josef Pannee
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy, University of GothenburgMölndalSweden
- Clinical Neurochemistry LabSahlgrenska University HospitalMölndalSweden
| | - Leslie M. Shaw
- Perelman School of MedicineDepartment of Pathology and Laboratory MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Magdalena Korecka
- Perelman School of MedicineDepartment of Pathology and Laboratory MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Teresa Waligorska
- Perelman School of MedicineDepartment of Pathology and Laboratory MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Charlotte E. Teunissen
- Neurochemistry LaboratoryDepartment of Clinical ChemistryAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamthe Netherlands
| | | | | | | | - Inge M. W. Verberk
- Neurochemistry LaboratoryDepartment of Clinical ChemistryAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamthe Netherlands
| | - Ashvini Keshavan
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | | | | | | | | | | | - Erik Portelius
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy, University of GothenburgMölndalSweden
- Clinical Neurochemistry LabSahlgrenska University HospitalMölndalSweden
| | - Ulf Andreasson
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy, University of GothenburgMölndalSweden
- Clinical Neurochemistry LabSahlgrenska University HospitalMölndalSweden
| | - Ritsuko Yoda
- Koichi Tanaka Mass Spectrometry Research LaboratoryShimadzu CorporationKyotoJapan
| | - Akinori Nakamura
- Department of Biomarker ResearchNational Center for Geriatrics and GerontologyObuAichiJapan
| | - Naoki Kaneko
- Koichi Tanaka Mass Spectrometry Research LaboratoryShimadzu CorporationKyotoJapan
| | | | | | | | | | - Kwasi G. Mawuenyega
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Vitaliy Ovod
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - James Bollinger
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Randall J. Bateman
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Yan Li
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | | | - Erik Stomrud
- Clinical Memory Research UnitFaculty of MedicineLund UniversityLundSweden
- Memory ClinicSkåne University HospitalMalmöSweden
| | - Oskar Hansson
- Clinical Memory Research UnitFaculty of MedicineLund UniversityLundSweden
- Memory ClinicSkåne University HospitalMalmöSweden
| | | | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy, University of GothenburgMölndalSweden
- Clinical Neurochemistry LabSahlgrenska University HospitalMölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy, University of GothenburgMölndalSweden
- Clinical Neurochemistry LabSahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research InstituteLondonUK
| |
Collapse
|
34
|
Feinkohl I, Schipke CG, Kruppa J, Menne F, Winterer G, Pischon T, Peters O. Plasma Amyloid Concentration in Alzheimer's Disease: Performance of a High-Throughput Amyloid Assay in Distinguishing Alzheimer's Disease Cases from Controls. J Alzheimers Dis 2021; 74:1285-1294. [PMID: 32176645 PMCID: PMC7242850 DOI: 10.3233/jad-200046] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background: Collection of cerebrospinal fluid (CSF) for measurement of amyloid-β (Aβ) species is a gold standard in Alzheimer’s disease (AD) diagnosis, but has risks. Thus, establishing a low-risk blood Aβ test with high AD sensitivity and specificity is of outmost interest. Objective: We evaluated the ability of a commercially available plasma Aβ assay to distinguish AD patients from biomarker-healthy controls. Method: In a case-control design, we examined plasma samples from 44 AD patients (A + N+) and 49 controls (A–N–) from a memory clinic. AD was diagnosed using a combination of neuropsychological examination, CSF biomarker analysis and brain imaging. Total Aβ40 and total Aβ42 in plasma were measured through enzyme-linked immunosorbent assay (ELISA) technology using ABtest40 and ABtest42 test kits (Araclon Biotech Ltd.). Receiver operating characteristic (ROC) analyses with outcome AD were performed, and sensitivity and specificity were calculated. Results: Plasma Aβ42/40 was weakly positively correlated with CSF Aβ42/40 (Spearman’s rho 0.22; p = 0.037). Plasma Aβ42/40 alone was not able to statistically significantly distinguish between AD patients and controls (AUC 0.58; 95% CI 0.46, 0.70). At a cut-point of 0.076 maximizing sensitivity and specificity, plasma Aβ42/40 had a sensitivity of 61.2% and a specificity of 63.6%. Conclusion: In this sample, the high-throughput blood Aβ assay was not able to distinguish well between AD patients and controls. Whether or not the assay may be useful in large-scale epidemiological settings remains to be seen.
Collapse
Affiliation(s)
- Insa Feinkohl
- Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Carola G Schipke
- Berlin Institute of Health (BIH), Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health (BIH), Experimental & Clinical Research Center (ECRC), Berlin, Germany
| | - Jochen Kruppa
- Berlin Institute of Health (BIH), Berlin, Germany.,Institut für Biometrie und Klinische Epidemiologie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Felix Menne
- Berlin Institute of Health (BIH), Berlin, Germany.,Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Georg Winterer
- Berlin Institute of Health (BIH), Berlin, Germany.,Pharmaimage Biomarker Solutions GmbH, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Tobias Pischon
- Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany.,MDC/BIH Biobank, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), and Berlin Institute of Health (BIH), Berlin, Germany
| | - Oliver Peters
- Berlin Institute of Health (BIH), Berlin, Germany.,Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
35
|
Highly specific and ultrasensitive plasma test detects Abeta(1-42) and Abeta(1-40) in Alzheimer's disease. Sci Rep 2021; 11:9736. [PMID: 33958661 PMCID: PMC8102604 DOI: 10.1038/s41598-021-89004-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
Plasma biomarkers that reflect specific amyloid beta (Abeta) proteoforms provide an insight in the treatment effects of Alzheimer’s disease (AD) therapies. Our aim was to develop and validate ready-to-use Simoa ‘Amyblood’ assays that measure full length Abeta1-42 and Abeta1-40 and compare their performance with two commercial assays. Linearity, intra- and inter-assay %CV were compared between Amyblood, Quanterix Simoa triplex, and Euroimmun ELISA. Sensitivity and selectivity were assessed for Amyblood and the Quanterix triplex. Clinical performance was assessed in CSF biomarker confirmed AD (n = 43, 68 ± 6 years) and controls (n = 42, 62 ± 5 years). Prototype and Amyblood showed similar calibrator curves and differentiation (20 AD vs 20 controls, p < 0.001). Amyblood, Quanterix triplex, and ELISA showed similar linearity (96%-122%) and intra-assay %CVs (≤ 3.1%). A minor non-specific signal was measured with Amyblood of + 2.4 pg/mL Abeta1-42 when incubated with 60 pg/mL Abeta1-40. A substantial non-specific signal of + 24.7 pg/mL Abetax-42 was obtained when 40 pg/mL Abeta3-42 was measured with the Quanterix triplex. Selectivity for Abeta1-42 at physiological Abeta1-42 and Abeta1-40 concentrations was 125% for Amyblood and 163% for Quanterix. Amyblood and Quanterix ratios (p < 0.001) and ELISA Abeta1-42 concentration (p = 0.025) could differentiate AD from controls. We successfully developed and upscaled a prototype to the Amyblood assays with similar technical and clinical performance as the Quanterix triplex and ELISA, but better specificity and selectivity than the Quanterix triplex assay. These results suggest leverage of this specific assay for monitoring treatment response in trials.
Collapse
|
36
|
Cano A, Turowski P, Ettcheto M, Duskey JT, Tosi G, Sánchez-López E, García ML, Camins A, Souto EB, Ruiz A, Marquié M, Boada M. Nanomedicine-based technologies and novel biomarkers for the diagnosis and treatment of Alzheimer's disease: from current to future challenges. J Nanobiotechnology 2021; 19:122. [PMID: 33926475 PMCID: PMC8086346 DOI: 10.1186/s12951-021-00864-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/17/2021] [Indexed: 02/07/2023] Open
Abstract
Increasing life expectancy has led to an aging population, which has consequently increased the prevalence of dementia. Alzheimer's disease (AD), the most common form of dementia worldwide, is estimated to make up 50-80% of all cases. AD cases are expected to reach 131 million by 2050, and this increasing prevalence will critically burden economies and health systems in the next decades. There is currently no treatment that can stop or reverse disease progression. In addition, the late diagnosis of AD constitutes a major obstacle to effective disease management. Therefore, improved diagnostic tools and new treatments for AD are urgently needed. In this review, we investigate and describe both well-established and recently discovered AD biomarkers that could potentially be used to detect AD at early stages and allow the monitoring of disease progression. Proteins such as NfL, MMPs, p-tau217, YKL-40, SNAP-25, VCAM-1, and Ng / BACE are some of the most promising biomarkers because of their successful use as diagnostic tools. In addition, we explore the most recent molecular strategies for an AD therapeutic approach and nanomedicine-based technologies, used to both target drugs to the brain and serve as devices for tracking disease progression diagnostic biomarkers. State-of-the-art nanoparticles, such as polymeric, lipid, and metal-based, are being widely investigated for their potential to improve the effectiveness of both conventional drugs and novel compounds for treating AD. The most recent studies on these nanodevices are deeply explained and discussed in this review.
Collapse
Affiliation(s)
- Amanda Cano
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain.
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
- Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain.
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain.
| | - Patric Turowski
- UCL Institute of Ophthalmology, University College of London, London, UK
| | - Miren Ettcheto
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Jason Thomas Duskey
- Nanotech Lab, Te.Far.T.I, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Umberto Veronesi Foundation, 20121, Milano, Italy
| | - Giovanni Tosi
- Nanotech Lab, Te.Far.T.I, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Sánchez-López
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Maria Luisa García
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Antonio Camins
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Eliana B Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Agustín Ruiz
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Marta Marquié
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
37
|
Giudici KV, de Souto Barreto P, Guyonnet S, Li Y, Bateman RJ, Vellas B. Assessment of Plasma Amyloid-β42/40 and Cognitive Decline Among Community-Dwelling Older Adults. JAMA Netw Open 2020; 3:e2028634. [PMID: 33331917 PMCID: PMC7747018 DOI: 10.1001/jamanetworkopen.2020.28634] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
IMPORTANCE Plasma measurement of amyloid-β (Aβ) peptides has been associated with cognitive function, but evidence of its ability to identify cognitive decline is still scarce. OBJECTIVE To investigate the associations between plasma Aβ42/40 and cognitive decline over time among community-dwelling older adults with subjective memory concerns. DESIGN, SETTING, AND PARTICIPANTS This multicenter cohort study used data from volunteers in the 5-year study Multidomain Alzheimer Preventive Trial (MAPT). Participants were aged 70 years or older and observed for a median (interquartile range) of 3.9 (2.0-4.0) years. Recruitment of participants started in May 2008 and ended in February 2011. Follow-up ended in April 2016. Data analysis was conducted from April to October 2020. EXPOSURE Plasma Aβ42 and Aβ40 were measured at 12 months for 448 participants (92.8%) and at 24 months for the rest. The moment of Aβ assessment was defined as the baseline for this study. MAIN OUTCOMES AND MEASURES Cognitive function was assessed at 12, 24, 36, 48, and 60 months by a composite cognitive score based on 4 tests; Mini Mental State Examination (MMSE); Clinical Dementia Rating, sum of boxes; and Alzheimer Disease Cooperative Study-Activities of Daily Living. Mixed-effect linear regressions were performed. RESULTS A total of 483 participants (median [IQR] age, 76.0 [73.0-80.0]; 286 [59.2%] women) were analyzed. Of them, 161 (33.3%) were classified as low plasma Aβ42/40 (≤0.107). After adjusting for age, sex, education, body mass index, Geriatric Depression Scale score, apolipoprotein E ε4 genotype, and MAPT intervention groups, low plasma Aβ42/40 was associated with more pronounced decline in composite cognitive score (adjusted between-group mean difference: -0.20, 95% CI, -0.34 to -0.07; P = .004) and decline in MMSE score (adjusted between-group mean difference: -0.59; 95% CI, -1.07 to -0.11; P = .02) during the follow-up period compared with the group with an Aβ42/40 ratio greater than 0.107. CONCLUSIONS AND RELEVANCE In this study, low plasma Aβ42/40 was associated with more pronounced decline in cognitive function (measured by multiple outcomes) over time. Findings suggest that plasma Aβ42/40 may be used to identify people at risk of cognitive decline, being an alternative to more complex and expensive measures, such as positron emission tomography imaging or cerebrospinal fluid measurement.
Collapse
Affiliation(s)
| | - Philipe de Souto Barreto
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital, Toulouse, France
- UPS/Inserm UMR1027, University of Toulouse III, Toulouse, France
| | - Sophie Guyonnet
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital, Toulouse, France
- UPS/Inserm UMR1027, University of Toulouse III, Toulouse, France
| | - Yan Li
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, Missouri
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Randall John Bateman
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Bruno Vellas
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital, Toulouse, France
- UPS/Inserm UMR1027, University of Toulouse III, Toulouse, France
| |
Collapse
|
38
|
Alonso-Lana S, Marquié M, Ruiz A, Boada M. Cognitive and Neuropsychiatric Manifestations of COVID-19 and Effects on Elderly Individuals With Dementia. Front Aging Neurosci 2020; 12:588872. [PMID: 33192483 PMCID: PMC7649130 DOI: 10.3389/fnagi.2020.588872] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly spread worldwide and has had unprecedented effects in healthcare systems, economies and society. COVID-19 clinical presentation primarily affects the respiratory system causing bilateral pneumonia, but it is increasingly being recognized as a systemic disease, with neurologic manifestations reported in patients with mild symptoms but, most frequently, in those in a severe condition. Elderly individuals are at high risk of developing severe forms of COVID-19 due to factors associated with aging and a higher prevalence of medical comorbidities and, therefore, they are more vulnerable to possible lasting neuropsychiatric and cognitive impairments. Several reports have described insomnia, depressed mood, anxiety, post-traumatic stress disorder and cognitive impairment in a proportion of patients after discharge from the hospital. The potential mechanisms underlying these symptoms are not fully understood but are probably multifactorial, involving direct neurotrophic effect of SARS-CoV-2, consequences of long intensive care unit stays, the use of mechanical ventilation and sedative drugs, brain hypoxia, systemic inflammation, secondary effects of medications used to treat COVID-19 and dysfunction of peripheral organs. Chronic diseases such as dementia are a particular concern not only because they are associated with higher rates of hospitalization and mortality but also because COVID-19 further exacerbates the vulnerability of those with cognitive impairment. In patients with dementia, COVID-19 frequently has an atypical presentation with mental status changes complicating the early identification of cases. COVID-19 has had a dramatical impact in long-term care facilities, where rates of infection and mortality have been very high. Community measures implemented to slow the spread of the virus have forced to social distancing and cancelation of cognitive stimulation programs, which may have contributed to generate loneliness, behavioral symptoms and worsening of cognition in patients with dementia. COVID-19 has impacted the functioning of Memory Clinics, research programs and clinical trials in the Alzheimer’s field, triggering the implementation of telemedicine. COVID-19 survivors should be periodically evaluated with comprehensive cognitive and neuropsychiatric assessments, and specific mental health and cognitive rehabilitation programs should be provided for those suffering long-term cognitive and psychiatric sequelae.
Collapse
Affiliation(s)
- Silvia Alonso-Lana
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Marta Marquié
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Agustín Ruiz
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
39
|
Zhang T, Kong W, Wang S, Mou X. Association Analysis of Peripheral and CSF Biomarkers in Late Mild Cognitive Impairment. Front Genet 2020; 11:834. [PMID: 32903489 PMCID: PMC7437457 DOI: 10.3389/fgene.2020.00834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/10/2020] [Indexed: 01/13/2023] Open
Abstract
Research shows that late mild cognitive impairment (LMCI) has a high risk of turning into Alzheimer's disease (AD). Due to the invasion of detection methods and physical damage to the patients, it is not a convenient way to diagnose and detect early AD and LMCI by cerebrospinal fluid (CSF) data. So there is an urgent need to find the correlation between peripheral biological data and CSF data in the brain, and to find new diagnostic methods through changes in the peripheral biological data. Studies have shown that during the pathogenesis of LMCI and AD, peripheral immune cells specifically infiltrate into the brain through the blood-brain barrier, causing an imbalance in the brain's immune response and dysregulating the clearance of Aβ in CSF. Therefore, in this paper, canonical correlation analysis (CCA) algorithm is presented to derive the correlation between peripheral and CSF biomarkers based on LMCI peripheral gene expression data and plasma marker information. Firstly, to explore the influence of the infiltration of peripheral blood immune cells on the brain, the abundance of 28 immune cells were calculated by using the gene set enrichment analysis algorithm of LMCI samples. Then, to identify the correlation between biomarkers inside and outside of the brain, we performed CCA to calculate the relationship between CSF and peripheral biomarkers. Results of CCA showed significant correlations between the variable sets of 8 peripheral biomarkers and the variable sets of CSF biomarkers (at 0.794). Finally, according to Kyoto Encyclopedia of Genes and Genomes and Gene Ontology analysis, it was found that the obtained peripheral biomarkers are involved in many immune-related pathways and functions which can be activated in peripheral blood of LMCI patients. Most related genes enriched in immune-related pathways and functions were up-regulated. Through receiver operating characteristic curve (ROC) analysis, it was also found that FP40/FP42 and type 1 T helper can accurately predict the pathological changes of LMCI (at 0.747).
Collapse
Affiliation(s)
- Tao Zhang
- College of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Wei Kong
- College of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Shuaiqun Wang
- College of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Xiaoyang Mou
- Department of Biochemistry, Rowan University and Guava Medicine, Glassboro, NJ, United States
| |
Collapse
|
40
|
Current Biomarkers for Alzheimer's Disease: From CSF to Blood. J Pers Med 2020; 10:jpm10030085. [PMID: 32806668 PMCID: PMC7564023 DOI: 10.3390/jpm10030085] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 12/23/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia and affects a large portion of the elderly population worldwide. Currently, a diagnosis of AD depends on the clinical symptoms of dementia, magnetic resonance imaging to determine brain volume, and positron emission tomography imaging to detect brain amyloid or tau deposition. The best characterized biological fluid markers for AD are decreased levels of amyloid β-protein (Aβ) 42 and increased levels of phosphorylated tau and total tau in cerebrospinal fluid (CSF). However, less invasive and easily detectable biomarkers for the diagnosis of AD, especially at the early stage, are still under development. Here, we provide an overview of various biomarkers identified in CSF and blood for the diagnostics of AD over the last 25 years. CSF biomarkers that reflect the three hallmarks of AD, amyloid deposition, neurofibrillary tangles, and neurodegeneration, are well established. Based on the need to start treatment in asymptomatic people with AD and to screen for AD risk in large numbers of young, healthy individuals, the development of biomarkers for AD is shifting from CSF to blood. Elements of the core pathogenesis of AD in blood, including Aβ42, tau proteins, plasma proteins, or lipids have shown their usefulness and capabilities in AD diagnosis. We also highlight some novel identified blood biomarkers (including Aβ42/Aβ43, p-tau 181, Aβ42/APP669-711, structure of Aβ in blood, and flotillin) for AD.
Collapse
|
41
|
Stylianaki I, Polizopoulou ZS, Theodoridis A, Koutouzidou G, Baka R, Papaioannou NG. Amyloid-beta plasma and cerebrospinal fluid biomarkers in aged dogs with cognitive dysfunction syndrome. J Vet Intern Med 2020; 34:1532-1540. [PMID: 32557873 PMCID: PMC7379053 DOI: 10.1111/jvim.15812] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/02/2020] [Accepted: 05/08/2020] [Indexed: 01/03/2023] Open
Abstract
Background Cognitive dysfunction syndrome (CDS) is a common progressive neurodegenerative disease that is poorly defined. Specific multitargeted protocols do not exist for setting the diagnosis and the prognosis of the syndrome. Hypothesis/Objectives To quantify Aβ42 and Aβ40 peptides in blood and cerebrospinal fluid (CSF) and to investigate their contribution to CCDS. Animals A total of 61 dogs from a hospital population. Methods Case‐control study. Six young (YG: 0‐4 years old), 8 middle‐aged (4‐8 years old), 17 cognitively unimpaired and aged (CU: 8‐20 years old), and 30 cognitively impaired and aged (CI: 8‐17 years). From the CI group, 10 dogs exhibited mild impairment (CI‐MCI) and 20 exhibited severe impairment (CI‐SCI). Cognitive status was assessed using a validated owner‐based questionnaire. Direct and indirect Aβ markers were determined in plasma fractions (total‐TP, free‐FP, bound to plasma components‐CP) and CSF using commercial ELISA assays (AΒtest, Araclon Biotech). Results TPAβ42/40 facilitated discrimination between CI‐MCI and CU aged dogs with area under curve ≥ 0.79. CSFAβ42 levels were higher (P = .09) in CU (1.25 ± 0.28 ng/mL) than in MCI (1.04 ± 0.32 ng/mL) dogs. CSF Aβ42 levels were correlated with the CP fragment (CPAβ40: P = .02, CPAβ42: P = .02). CPAβ42 was higher in the CI‐MCI (23.03 ± 11.79 pg/μL) group compared to the other aged dogs (CU: 10.42 ± 7.18 pg/μL, P = .02, SCI: 11.40 ± 12.98 pg/μL, P = .26). Conclusion and Clinical Importance The Aβ should be determined in all of the 3 plasma fractions (TP, FP, CP). In the clinical approach, TPAβ42/40 could be used as an efficient preselection tool for the aged canine population targeting dogs with mild cognitive impairment.
Collapse
Affiliation(s)
- Ioanna Stylianaki
- Department of Pathology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Zoe S Polizopoulou
- Diagnostic Laboratory, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alexandros Theodoridis
- Laboratory of Animal Production Economics, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgia Koutouzidou
- Department of Applied Informatics, University of Macedonia, Thessaloniki, Greece
| | - Rania Baka
- Diagnostic Laboratory, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos G Papaioannou
- Department of Pathology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
42
|
Doecke JD, Pérez-Grijalba V, Fandos N, Fowler C, Villemagne VL, Masters CL, Pesini P, Sarasa M. Total Aβ 42/Aβ 40 ratio in plasma predicts amyloid-PET status, independent of clinical AD diagnosis. Neurology 2020; 94:e1580-e1591. [PMID: 32179698 PMCID: PMC7251518 DOI: 10.1212/wnl.0000000000009240] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 10/24/2019] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE To explore whether the plasma total β-amyloid (Aβ) Aβ42/Aβ40 ratio is a reliable predictor of the amyloid-PET status by exploring the association between these 2 variables in a subset of the Australian Imaging, Biomarkers and Lifestyle (AIBL) study of aging cohort. METHODS Taking plasma samples at 3 separate time points, month 18 (n = 176), month 36 (n = 169), and month 54 (n = 135), we assessed the total Aβ42/Aβ40 ratio in plasma (TP42/40) with regard to neocortical Aβ burden via PET standardized uptake value ratio (SUVR) and investigated both association with Aβ-PET status and correlation (and agreement) with SUVR. RESULTS The TP42/40 plasma ratio was significantly reduced in amyloid-PET-positive participants at all time points (p < 0.0001). Adjusting for covariates age, gender, APOE ε4 allele status, and clinical classification clearly affects the significance, with p values reduced and only comparisons at 54 months retaining significance (p = 0.006). Correlations with SUVR were similar across each time point, with Spearman ρ reaching -0.64 (p < 0.0001). Area under the curve values were highly reproducible over time points, with values ranging from 0.880 at 36 months to 0.913 at 54 months. In assessments of the healthy control group only, the same relationships were found. CONCLUSIONS The current study demonstrates reproducibility of the plasma assay to discriminate between amyloid-PET positive and negative over 3 time points, which can help to substantially reducing the screening rate of failure for clinical trials targeting preclinical or prodromal disease. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that plasma total Aβ42/Aβ40 ratio is associated with neocortical amyloid burden as measured by PET SUVR.
Collapse
Affiliation(s)
- James D Doecke
- From the CSIRO Health and Biosecurity/Australian E-Health Research Centre (J.D.D.), Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; R&D Department (V.P.-G., N.F., P.P., M.S.), Araclon Biotech Ltd, Zaragoza, Spain; and The Florey Institute of Neuroscience and Mental Health (C.F., V.L.V., C.L.M.), University of Melbourne, Parkville, Victoria, Australia
| | - Virginia Pérez-Grijalba
- From the CSIRO Health and Biosecurity/Australian E-Health Research Centre (J.D.D.), Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; R&D Department (V.P.-G., N.F., P.P., M.S.), Araclon Biotech Ltd, Zaragoza, Spain; and The Florey Institute of Neuroscience and Mental Health (C.F., V.L.V., C.L.M.), University of Melbourne, Parkville, Victoria, Australia
| | - Noelia Fandos
- From the CSIRO Health and Biosecurity/Australian E-Health Research Centre (J.D.D.), Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; R&D Department (V.P.-G., N.F., P.P., M.S.), Araclon Biotech Ltd, Zaragoza, Spain; and The Florey Institute of Neuroscience and Mental Health (C.F., V.L.V., C.L.M.), University of Melbourne, Parkville, Victoria, Australia
| | - Christopher Fowler
- From the CSIRO Health and Biosecurity/Australian E-Health Research Centre (J.D.D.), Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; R&D Department (V.P.-G., N.F., P.P., M.S.), Araclon Biotech Ltd, Zaragoza, Spain; and The Florey Institute of Neuroscience and Mental Health (C.F., V.L.V., C.L.M.), University of Melbourne, Parkville, Victoria, Australia
| | - Victor L Villemagne
- From the CSIRO Health and Biosecurity/Australian E-Health Research Centre (J.D.D.), Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; R&D Department (V.P.-G., N.F., P.P., M.S.), Araclon Biotech Ltd, Zaragoza, Spain; and The Florey Institute of Neuroscience and Mental Health (C.F., V.L.V., C.L.M.), University of Melbourne, Parkville, Victoria, Australia
| | - Colin L Masters
- From the CSIRO Health and Biosecurity/Australian E-Health Research Centre (J.D.D.), Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; R&D Department (V.P.-G., N.F., P.P., M.S.), Araclon Biotech Ltd, Zaragoza, Spain; and The Florey Institute of Neuroscience and Mental Health (C.F., V.L.V., C.L.M.), University of Melbourne, Parkville, Victoria, Australia
| | - Pedro Pesini
- From the CSIRO Health and Biosecurity/Australian E-Health Research Centre (J.D.D.), Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; R&D Department (V.P.-G., N.F., P.P., M.S.), Araclon Biotech Ltd, Zaragoza, Spain; and The Florey Institute of Neuroscience and Mental Health (C.F., V.L.V., C.L.M.), University of Melbourne, Parkville, Victoria, Australia.
| | - Manuel Sarasa
- From the CSIRO Health and Biosecurity/Australian E-Health Research Centre (J.D.D.), Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; R&D Department (V.P.-G., N.F., P.P., M.S.), Araclon Biotech Ltd, Zaragoza, Spain; and The Florey Institute of Neuroscience and Mental Health (C.F., V.L.V., C.L.M.), University of Melbourne, Parkville, Victoria, Australia
| | | |
Collapse
|
43
|
Pérez-Grijalba V, Arbizu J, Romero J, Prieto E, Pesini P, Sarasa L, Guillen F, Monleón I, San-José I, Martínez-Lage P, Munuera J, Hernández I, Buendía M, Sotolongo-Grau O, Alegret M, Ruiz A, Tárraga L, Boada M, Sarasa M. Plasma Aβ42/40 ratio alone or combined with FDG-PET can accurately predict amyloid-PET positivity: a cross-sectional analysis from the AB255 Study. Alzheimers Res Ther 2019; 11:96. [PMID: 31787105 PMCID: PMC6886187 DOI: 10.1186/s13195-019-0549-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/22/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND To facilitate population screening and clinical trials of disease-modifying therapies for Alzheimer's disease, supportive biomarker information is necessary. This study was aimed to investigate the association of plasma amyloid-beta (Aβ) levels with the presence of pathological accumulation of Aβ in the brain measured by amyloid-PET. Both plasma Aβ42/40 ratio alone or combined with an FDG-PET-based biomarker of neurodegeneration were assessed as potential AD biomarkers. METHODS We included 39 cognitively normal subjects and 20 patients with mild cognitive impairment from the AB255 Study who had undergone PiB-PET scans. Total Aβ40 and Aβ42 levels in plasma (TP42/40) were quantified using ABtest kits. Subjects were dichotomized as Aβ-PET positive or negative, and the ability of TP42/40 to detect Aβ-PET positivity was assessed by logistic regression and receiver operating characteristic analyses. Combination of plasma Aβ biomarkers and FDG-PET was further assessed as an improvement for brain amyloidosis detection and diagnosis classification. RESULTS Eighteen (30.5%) subjects were Aβ-PET positive. TP42/40 ratio alone identified Aβ-PET status with an area under the curve (AUC) of 0.881 (95% confidence interval [CI] = 0.779-0.982). Discriminating performance of TP42/40 to detect Aβ-PET-positive subjects yielded sensitivity and specificity values at Youden's cutoff of 77.8% and 87.5%, respectively, with a positive predictive value of 0.732 and negative predictive value of 0.900. All these parameters improved after adjusting the model for significant covariates. Applying TP42/40 as the first screening tool in a sequential diagnostic work-up would reduce the number of Aβ-PET scans by 64%. Combination of both FDG-PET scores and plasma Aβ biomarkers was found to be the most accurate Aβ-PET predictor, with an AUC of 0.965 (95% CI = 0.913-0.100). CONCLUSIONS Plasma TP42/40 ratio showed a relevant and significant potential as a screening tool to identify brain Aβ positivity in preclinical and prodromal stages of Alzheimer's disease.
Collapse
Affiliation(s)
| | - Javier Arbizu
- Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain
| | - Judith Romero
- Araclon Biotech S.L., Vía Hispanidad 21, 50009, Zaragoza, Spain
| | - Elena Prieto
- Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain
| | - Pedro Pesini
- Araclon Biotech S.L., Vía Hispanidad 21, 50009, Zaragoza, Spain.
| | - Leticia Sarasa
- Araclon Biotech S.L., Vía Hispanidad 21, 50009, Zaragoza, Spain
| | - Fernando Guillen
- Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Itziar San-José
- Araclon Biotech S.L., Vía Hispanidad 21, 50009, Zaragoza, Spain
| | - Pablo Martínez-Lage
- Center for Research and Advanced Therapies and Memory Clinic, Fundación CITA-Alzheimer, San Sebastián, Spain
| | - Josep Munuera
- Institut de recerca Sant Joan de Déu, Hospital Infantil Sant Joan de Déu, Barcelona, Spain
| | - Isabel Hernández
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mar Buendía
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
| | - Oscar Sotolongo-Grau
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
| | - Montserrat Alegret
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Agustín Ruiz
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Lluis Tárraga
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Sarasa
- Araclon Biotech S.L., Vía Hispanidad 21, 50009, Zaragoza, Spain
| |
Collapse
|
44
|
Risacher SL, Fandos N, Romero J, Sherriff I, Pesini P, Saykin AJ, Apostolova LG. Plasma amyloid beta levels are associated with cerebral amyloid and tau deposition. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2019; 11:510-519. [PMID: 31384662 PMCID: PMC6661419 DOI: 10.1016/j.dadm.2019.05.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction We investigated the relationship of plasma amyloid beta (Aβ) with cerebral deposition of Aβ and tau on positron emission tomography (PET). Methods Forty-four participants (18 cognitively normal older adults [CN], 10 mild cognitive impairment, 16 Alzheimer's disease [AD]) underwent amyloid PET and a blood draw. Free and total plasma Aβ40 and Aβ42 were assessed using a validated assay. Thirty-seven participants (17 CN, 8 mild cognitive impairment, 12 AD) also underwent a [18F]flortaucipir scan. Scans were preprocessed by standard techniques, and mean global and regional amyloid and tau values were extracted. Free Aβ42/Aβ40 (Aβ F42:F40) and total Aβ42/Aβ40 (Aβ T42:T40) were evaluated for differences by diagnosis and relation to PET Aβ positivity. Relationships between these measures and cerebral Aβ and tau on both regional and voxel-wise basis were also evaluated. Results Lower Aβ T42:T40 was associated with diagnosis and PET Aβ positivity. Lower plasma Aβ T42:T40 ratios predicted cerebral Aβ positivity, both across the full sample and in CN only. Finally, lower plasma Aβ T42:T40 ratios were associated with increased cortical Aβ and tau in AD-related regions on both regional and voxel-wise analyses. Discussion Plasma Aβ measures may be useful biomarkers for predicting cerebral Aβ and tau. Additional studies in larger samples are warranted.
Collapse
Affiliation(s)
- Shannon L Risacher
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Liana G Apostolova
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|