1
|
Sun J, Geng W, Wang Y, Li H, Tan R, Tu Y. An innovative electrochemiluminescent immunosensor using dual amplified signals from AuNPs@CoSn(OH) 6 for the detection of the AD biomarker: amyloid beta 1-40. Analyst 2025. [PMID: 40326625 DOI: 10.1039/d5an00048c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Alzheimer's disease (AD) is a degenerative condition of the nervous system that causes severe damage to patients' daily activities and quality of life. Amyloid beta 1-40 protein (Aβ40), which is involved in the formation of cerebral plaques, is one of the crucial biomarkers related to AD. Herein, a novel and highly sensitive immunosensor for the detection of Aβ40 is developed. Using a reinforced indium tin oxide-coated glass with a nanocomposite of gold nanoparticle-enhanced CoSn(OH)6 (AuNPs@CoSn(OH)6) to trigger the electrochemiluminescence (ECL) of luminol as the sensing signal, the immunosensor is fabricated by immobilizing the Aβ40 antibody onto it. By integrating the high immune specificity, excellent conductivity and catalytic activity of the nanocomposite, the resultant immunosensor can be successfully employed to detect the target in real samples. The formation of the immune complex leads to increased steric hindrance and electron transfer resistance, which in turn causes a declined ECL output when the target Aβ40 binds to the antibody on the sensor surface. Under optimized conditions, the developed ECL immunosensor exhibits a linear response for Aβ40 ranging from 1 to 800 pg mL-1 and a low detection limit of 0.47 pg mL-1. Experimentally, it is demonstrated to be highly sensitive, specific, reproducible and stable. This work extends the application of the perovskite CoSn(OH)6 and AuNPs in the field of ECL immunosensing and provides a novel strategy for clinical research on Alzheimer's disease.
Collapse
Affiliation(s)
- Jiaojing Sun
- College of Chemistry, Chemical Engineering and Material Science, Soochow University, Suzhou, 215123, P. R. China.
| | - Wenqing Geng
- First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Yueju Wang
- First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Huiling Li
- First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
- Nursing School, Suzhou Medical College of Soochow University, Suzhou, 215006, P. R. China
| | - Rong Tan
- College of Chemistry, Chemical Engineering and Material Science, Soochow University, Suzhou, 215123, P. R. China.
- School of Material Engineering, Changshu Institute of Technology, Suzhou, 215500, P. R. China.
| | - Yifeng Tu
- College of Chemistry, Chemical Engineering and Material Science, Soochow University, Suzhou, 215123, P. R. China.
| |
Collapse
|
2
|
Bouteloup V, Villain N, Vidal JS, Gonzalez-Ortiz F, Yuksekel I, Santos C, Schraen-Maschken S, Pellegrin I, Lehmann S, Blennow K, Chêne G, Hanon O, Dufouil C, Planche V. Cognitive Phenotyping and Interpretation of Alzheimer Blood Biomarkers. JAMA Neurol 2025; 82:506-515. [PMID: 40181683 PMCID: PMC11971688 DOI: 10.1001/jamaneurol.2025.0142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/16/2025] [Indexed: 04/05/2025]
Abstract
Importance Blood phosphorylated tau 217 (p-tau217) showed good performance in predicting brain amyloidosis. However, the importance of detailed cognitive phenotyping in patients without dementia when interpreting p-tau217 results remains unclear. Objective To assess whether accuracy, negative predictive value (NPV), and positive predictive value (PPV) in predicting brain amyloidosis using p-tau217 varies across clinical presentations in patients without dementia. Design, Setting, and Participants The study design included 2 observational, prospective cohort studies: The Cohort of Outpatients From French Research Memory Centers in Order to Improve Knowledge on Alzheimer's Disease and Related Disorders (MEMENTO), with enrollment from 2011 to 2014 and 5 years of follow-up, and the Biomarker of Amyloid Peptide and Alzheimer's Disease Risk (BALTAZAR) cohort study, with enrollment from 2010 to 2015 and 3 years of follow-up. Both are multicenter cohorts conducted in French memory clinics. Participants without dementia were included for analysis if they had baseline blood p-tau217 measurement and a known amyloid status through cerebrospinal fluid amyloid β (Aβ)-42/Aβ-40 ratio or positron emission tomography. They presented with either subjective cognitive impairment (SCI), mild cognitive impairment (MCI) with a common Alzheimer disease (AD) phenotype (cAD-MCI: amnestic syndrome of hippocampal type, posterior cortical atrophy, or logopenic primary progressive aphasia), or MCI with uncommon AD or other phenotypes (uAD-MCI). Data were analyzed from May to September 2024. Exposures Blood p-tau217 concentrations. Main Outcomes and Measures Brain amyloidosis probabilities were derived from p-tau217 logistic regressions including age, gender, and APOE genotype. Published and internally developed cut points with 90% sensitivity and specificity were used. Results A total of 776 participants from the MEMENTO cohort (N = 2323 participants) and 193 participants from the BALTAZAR cohort (N = 1040) were included in this analysis. In the MEMENTO cohort (median [IQR] age, 71 [65-76] years; 444 female [57%]), brain amyloidosis prevalence was 16.5% (20 of 121) in SCI, 45.9% (78 of 170) in cAD-MCI, and 24.5% (119 of 485) in uAD-MCI. Area under the receiver operating characteristic curve for predicting brain amyloidosis with p-tau217 models was 0.78 (95% CI, 0.66-0.89), 0.91 (95% CI, 0.86-0.95), and 0.87 (95% CI, 0.84-0.91) in the SCI, cAD-MCI, and uAD-MCI subgroups, respectively. External cut points resulted in a PPV of 60.0%, 90.0%, and 74.5% in the SCI, cAD-MCI, and uAD-MCI subgroups, respectively. NPV ranged from 84.2% to 90.2%. With internally developed cut points, PPVs were 52.6%, 84.0%, and 72.3% in the SCI, cAD-MCI, and uAD-MCI subgroups, respectively. NPVs were high (91.7%-94.6%) in all subgroups. Rates of incident dementia strongly increased with the probability of brain amyloidosis in the cAD-MCI subgroup. Replicated analyses in the BALTAZAR cohort provided similar results. Conclusions and Relevance Results from 2 clinical cohorts suggest that amyloid prevalence varied across cognitive phenotypes and was associated with the diagnostic performance of blood p-tau217 models to determine brain amyloidosis. Comprehensive cognitive phenotyping beyond the basic characterization of SCI, MCI, or dementia should accompany the use of blood biomarkers in clinical practice to avoid misdiagnosis due to false positives.
Collapse
Affiliation(s)
- Vincent Bouteloup
- Bordeaux Population Health, University of Bordeaux, Inserm, UMR1219, Bordeaux, France
- CIC 1401 EC, Pôle Santé Publique, CHU de Bordeaux, Bordeaux, France
| | - Nicolas Villain
- Sorbonne Université, INSERM U1127, CNRS 7225, Institut du Cerveau–ICM, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer’s Disease, AP-HP Sorbonne Université, Pitié-Salpêtrière Hospital, Paris, France
| | - Jean Sebastien Vidal
- Memory Resource and Research Centre of Paris-Broca-Ile de France, APHP, Hôpitaux Universitaires Paris Centre, Hospital Broca, Paris, France
- Université Paris Cité, EA 4468, Paris, France
| | - Fernando Gonzalez-Ortiz
- Inst. of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Idil Yuksekel
- Sorbonne Université, INSERM U1127, CNRS 7225, Institut du Cerveau–ICM, Paris, France
| | - Cristiano Santos
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Isabelle Pellegrin
- Laboratory of Immunology and Immunogenetics, Resources Biological Center, CHU Bordeaux, Bordeaux, France
- Univ. Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France
| | - Sylvain Lehmann
- LBPC-PPC, Univ Montpellier, INM INSERM, CHU Montpellier, Montpellier, France
| | - Kaj Blennow
- Sorbonne Université, INSERM U1127, CNRS 7225, Institut du Cerveau–ICM, Paris, France
- Inst. of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Geneviève Chêne
- Bordeaux Population Health, University of Bordeaux, Inserm, UMR1219, Bordeaux, France
- CIC 1401 EC, Pôle Santé Publique, CHU de Bordeaux, Bordeaux, France
| | - Olivier Hanon
- Memory Resource and Research Centre of Paris-Broca-Ile de France, APHP, Hôpitaux Universitaires Paris Centre, Hospital Broca, Paris, France
- Université Paris Cité, EA 4468, Paris, France
| | - Carole Dufouil
- Bordeaux Population Health, University of Bordeaux, Inserm, UMR1219, Bordeaux, France
- CIC 1401 EC, Pôle Santé Publique, CHU de Bordeaux, Bordeaux, France
| | - Vincent Planche
- Institut des Maladies Neurodégénératives, Univ. Bordeaux, CNRS, UMR 5293, Bordeaux, France
- Pôle de Neurosciences Cliniques, Centre Mémoire de Ressources et de Recherche, CHU de Bordeaux, Bordeaux, France
| |
Collapse
|
3
|
Shim JE, Kim YJ, Hahm E, Choe JH, Baek A, Kim RM, You EA. Ultrasensitive SERS nanoprobe-based multiplexed digital sensing platform for the simultaneous quantification of Alzheimer's disease biomarkers. Biosens Bioelectron 2025; 274:117216. [PMID: 39899917 DOI: 10.1016/j.bios.2025.117216] [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: 11/20/2024] [Revised: 01/18/2025] [Accepted: 01/27/2025] [Indexed: 02/05/2025]
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disease that requires early diagnosis to manage its progression. Although the simultaneous detection of multiple AD biomarkers is expected to facilitate early assessment of AD risk, the lack of multiplexed sensing platforms for accurately quantifying multiple AD biomarkers remains a challenge. Here, we present a multiplexed digital sensing platform based on bumpy core-shell (BCS) surface-enhanced Raman spectroscopy (SERS) nanoprobes for ultrasensitive, quantitative, and simultaneous detection of Aβ42 and Aβ40 as AD biomarkers, enabling the accurate determination of the Aβ42/Aβ40 ratio. We synthesized BCS SERS nanoprobes with distinct Raman reporters to generate unique, intense, and reproducible SERS signals, offering single-nanoparticle sensitivity and quantification capabilities. These nanoprobes were subsequently employed in SERS-based immunoassays combined with digital SERS analysis for multiplexed quantification. The proposed platform accurately and quantitatively detected Aβ42 and Aβ40 across a range of five orders of magnitude, with a limit of detection of 8.7× 10-17 g/mL (1.9 × 10-17 M) for Aβ42 and 1.0 × 10-15 g/mL (2.3 × 10-16 M) for Aβ40, surpassing the performance of conventional enzyme-linked immunosorbent assays. Based on the exclusive detection of Aβ42 and Aβ40 using distinct SERS nanoprobes, the proposed sensing platform can also accurately quantify Aβ42 and Aβ40 at clinically relevant levels in both cerebrospinal fluid and blood plasma. Therefore, this sensing platform can be used to accurately and reliably determine the Aβ42/Aβ40 ratio, thus serving as an effective tool for the early diagnosis of AD.
Collapse
Affiliation(s)
- Jae-Eul Shim
- Medical Metrology Group, Korea Research Institute of Standards and Science, Daejeon, 34113, Republic of Korea
| | - Young Jun Kim
- Medical Metrology Group, Korea Research Institute of Standards and Science, Daejeon, 34113, Republic of Korea
| | - Eunil Hahm
- Medical Metrology Group, Korea Research Institute of Standards and Science, Daejeon, 34113, Republic of Korea
| | - Jong-Ho Choe
- Department of Physics, Korea University, Seoul, 02841, Republic of Korea
| | - Ahruem Baek
- Nanobio Measurement Group, Korea Research Institute of Standards and Science, Daejeon, 34113, Republic of Korea
| | - Ryeong Myeong Kim
- Medical Metrology Group, Korea Research Institute of Standards and Science, Daejeon, 34113, Republic of Korea
| | - Eun-Ah You
- Medical Metrology Group, Korea Research Institute of Standards and Science, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
4
|
Leroy M, Aziz AL, Schraen S, Deramecourt V, Skrobala E, Lecerf S, Pasquier F, Huin V, Bertoux M, Lebouvier T. Comparing high and low amyloid producers in Alzheimer's disease: An in-depth analysis. Rev Neurol (Paris) 2025; 181:332-341. [PMID: 40057456 DOI: 10.1016/j.neurol.2025.02.004] [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: 11/22/2023] [Revised: 11/15/2024] [Accepted: 02/14/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND The cerebrospinal fluid (CSF) Aβ42/40 ratio has proven to be a more reliable biomarker for amyloid pathology than CSF Aβ42 in Alzheimer's disease (AD), helping to correctly classify patients with positive tau biomarkers (T+) that would otherwise have remained outside of the AD continuum. It was shown that the Aβ42/40 ratio better captures a relative decrease of Aβ42 in patients with high CSF Aβ. However, whether patients with high-amyloid (HiA) AD, in whom A+ is defined by the Aβ42/40 ratio, exactly compare with their low-amyloid (LoA) counterparts, in whom A+ is defined by Aβ42 solely, deserves further analysis. METHODS We retrospectively included patients with A+T+ AD and evidence of cognitive and neurodegenerative changes (N+). LoA patients were operationally defined as patients with T+N+ and low CSF Aβ42, while HiA patients were defined as patients with T+N+ and normal CSF Aβ42 but abnormal Aβ42/40 ratio. Tau CSF biomarkers, neuropsychological profile, rates of cognitive decline, structural and metabolic imaging, ApoE genotype and brain neuropathology were compared between the HiA and LoA groups. RESULTS At the time of the lumbar puncture, LoA patients were significantly younger than the HiA patients (68.9±8.7years vs. 71.8±9.4; P=0.0015) and had a lower Mini-Mental Status Examination (MMSE) (18.7±6.4 vs. 20.7±6.2; P=0.0005). There was no difference in the neuropsychological profile nor in the annual rates of cognitive decline between the two groups with early AD. No differences were retrieved between groups on CSF Tau and P-Tau biomarkers, atrophy and brain metabolism, distribution of the APOE4 allele and APOE4/E4 genotype, and neuropathology. CONCLUSIONS Overall, our study supports the surrogate use of the Aβ42/40 ratio as an equivalent to Aβ42 to define AD. We showed that HiA CSF profiles were not associated with differences in cognition, brain structures and metabolism, APOE genotype tau CSF biomarkers or the rates of cognitive decline, but may be the associated with later-onset and early-stage AD.
Collapse
Affiliation(s)
- Mélanie Leroy
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France; CHU de Lille, DISTALZ, Lille, France
| | - Anne Laure Aziz
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France
| | - Susanna Schraen
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France; CHU de Lille, DISTALZ, Lille, France
| | - Vincent Deramecourt
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France
| | | | - Simon Lecerf
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France
| | - Florence Pasquier
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France; CHU de Lille, DISTALZ, Lille, France
| | - Vincent Huin
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France; CHU de Lille, DISTALZ, Lille, France
| | - Maxime Bertoux
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France; CHU de Lille, DISTALZ, Lille, France
| | - Thibaud Lebouvier
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France; CHU de Lille, DISTALZ, Lille, France.
| |
Collapse
|
5
|
Gao Y, Wang L, Doeswijk T, Winblad B, Schedin‐Weiss S, Tjernberg LO. Intraneuronal Aβ accumulation causes tau hyperphosphorylation via endolysosomal leakage. Alzheimers Dement 2025; 21:e70091. [PMID: 40145397 PMCID: PMC11947758 DOI: 10.1002/alz.70091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 03/28/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD) is characterized by amyloid beta (Aβ) peptide plaques and intracellular neurofibrillary tangles formed by hyperphosphorylated tau. Many attempts have been made to clarify the link between Aβ and tau in the pathogenesis, but conclusive data describing a pathway for this connection are still lacking. METHODS We developed a neuronal model of Aβ-induced toxicity and studied downstream effects of intraneuronal Aβ42 accumulation on tau hyperphosphorylation using confocal microscopy and live cell imaging. RESULTS Aβ42 added to the medium was endocytosed into neurons, inducing the formation of endolysosomal protofibrils and endolysosomal leakage, which in turn promoted tau hyperphosphorylation. Asparaginyl endopeptidase (AEP) was released from the disrupted lysosomes, and inhibition of this peptidase activity reduced tau hyperphosphorylation. DISCUSSION The data suggest a mechanism of AD in which Aβ42 accumulates and aggregates gradually in neurons over time, leading to endolysosomal leakage and release of AEP, which subsequently triggers tau hyperphosphorylation. HIGHLIGHTS Aβ42 endocytosis leads to its endolysosomal accumulation in neurons over time. Aβ42 polymerizes into protofibrils and causes endolysosomal leakage. Tau hyperphosphorylation is induced by endolysosomal asparagine endopeptidase leakage. Tau hyperphosphorylation is inhibited by an asparagine endopeptidase inhibitor.
Collapse
Affiliation(s)
- Yang Gao
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Lisha Wang
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Tosca Doeswijk
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
- Theme Inflammation and AgingKarolinska University HospitalHuddingeSweden
| | - Sophia Schedin‐Weiss
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Lars O. Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| |
Collapse
|
6
|
Delaby C, Alcolea D, Busto G, Gabelle A, Ayrignac X, Bennys K, Muiño E, Villatoro P, Fernández-Cadenas I, Pradeilles N, Bounasri SE, Torres S, Hirtz C, Zetterberg H, Lleó A, Fortea J, Lehmann S. Plasma Hepcidin as a potential informative biomarker of Alzheimer disease and vascular dementia. Alzheimers Res Ther 2025; 17:42. [PMID: 39948603 PMCID: PMC11823057 DOI: 10.1186/s13195-025-01696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/07/2025] [Indexed: 02/16/2025]
Abstract
BACKGROUND Blood-based assays are expected to be integrated into clinical routines across various contexts, including Alzheimer's disease (AD). Vascular dementia (VaD), which is the second most common cause leading to dementia after AD, could also significantly benefit from this advancement. However, no informative fluid biomarker has been identified for VaD. Given the disruption of iron homeostasis in both AD and VaD, this study aims to characterize the potential of the iron-related hormone Hepcidin as a biomarker for these two conditions. We will compare its added value to established AT(N) blood biomarkers. METHODS Blood biomarkers (amyloid-composite, p-Tau181, Neurofilament Light Chain [NfL] and Hepcidin) levels in blood were analyzed in two independent cohorts and compared between AD patients and non-AD individuals. Additionally, blood Hepcidin and NfL were evaluated in the contexts of VaD and CADASIL, with their relative diagnostic value assessed. RESULTS Blood Hepcidin and NfL do not significantly increase the AUC obtained with both p-Tau181 and amyloid composite in the context of AD. In contrast, AUC for VaD diagnosis is significantly higher when combining these two blood biomarkers compared to NfL alone. Hepcidin was not significantly modified in CADASIL patients compared to control subjects. CONCLUSION Blood Hepcidin and NfL have limited interest in the clinical context of AD but determination of these biomarkers shows to be highly informative for the diagnosis of VaD. This result could have important implications for diagnosis and implementation of clinical trials.
Collapse
Affiliation(s)
- Constance Delaby
- Université de Montpellier, INM INSERM LBPC-PPC, IRMB CHU de Montpellier, Montpellier, France.
- Department of Neurology, Sant Pau Memory Unit, Hospital de La Santa Creu I Sant Pau - IIB Sant Pau, Barcelona, Spain.
| | - Daniel Alcolea
- Department of Neurology, Sant Pau Memory Unit, Hospital de La Santa Creu I Sant Pau - IIB Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Ciberned, Spain
| | - Germain Busto
- Department of Neurology, Université de Montpellier, Inserm INM NeuroPEPs Team, 34000, Montpellier, France
| | - Audrey Gabelle
- Department of Neurology, Université de Montpellier, Inserm INM NeuroPEPs Team, 34000, Montpellier, France
| | - Xavier Ayrignac
- Department of Neurology, Université de Montpellier, Inserm INM NeuroPEPs Team, 34000, Montpellier, France
| | - Karim Bennys
- Department of Neurology, Université de Montpellier, Inserm INM NeuroPEPs Team, 34000, Montpellier, France
| | - Elena Muiño
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Farmacogenómica y Genética Neurovascular, Sant Quintí 77-79, Barcelona, 08041, Spain
- Unidad de Epilepsia, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, Spain, 08025, Barcelona
| | - Paula Villatoro
- Farmacogenómica y Genética Neurovascular. Institut d'Investigació Biomèdica Sant Pau (IIB, SANT PAU), Sant Quintí 77-79, 08041, Barcelona, Spain
| | - Israel Fernández-Cadenas
- Farmacogenómica y Genética Neurovascular. Institut d'Investigació Biomèdica Sant Pau (IIB, SANT PAU), Sant Quintí 77-79, 08041, Barcelona, Spain
| | - Nicolas Pradeilles
- Université de Montpellier, INM INSERM LBPC-PPC, IRMB CHU de Montpellier, Montpellier, France
| | - Shaima El Bounasri
- Department of Neurology, Sant Pau Memory Unit, Hospital de La Santa Creu I Sant Pau - IIB Sant Pau, Barcelona, Spain
| | - Soraya Torres
- Department of Neurology, Sant Pau Memory Unit, Hospital de La Santa Creu I Sant Pau - IIB Sant Pau, Barcelona, Spain
| | - Christophe Hirtz
- Université de Montpellier, INM INSERM LBPC-PPC, IRMB CHU de Montpellier, Montpellier, France
| | - 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
- Dementia Research Institute at UCL, London, UK
- Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Alberto Lleó
- Department of Neurology, Sant Pau Memory Unit, Hospital de La Santa Creu I Sant Pau - IIB Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Ciberned, Spain
| | - Juan Fortea
- Department of Neurology, Sant Pau Memory Unit, Hospital de La Santa Creu I Sant Pau - IIB Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Ciberned, Spain
| | - Sylvain Lehmann
- Université de Montpellier, INM INSERM LBPC-PPC, IRMB CHU de Montpellier, Montpellier, France
| |
Collapse
|
7
|
Lee BH, Cevizci M, Lieblich SE, Galea LAM. Sex-specific influences of APOEε4 genotype on hippocampal neurogenesis and progenitor cells in middle-aged rats. Biol Sex Differ 2025; 16:10. [PMID: 39910616 PMCID: PMC11796140 DOI: 10.1186/s13293-025-00694-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) disproportionately and uniquely affects females, and these sex differences are further exacerbated by the presence of Apolipoprotein (APOE) ε4 alleles, the top genetic risk factor for late-onset AD. To expand our understanding about how late-onset AD risk might differentially influence males and females, this study explores how APOEε4 affects hippocampal neurogenesis and microglia, key neuroplastic markers involved in AD pathogenesis, differently by sex in middle-aged rats. METHODS A rat model expressing the humanized (h) APOEε4 allele was characterized to examine markers of adult neurogenesis (neural progenitor cells and new-born neurons) and immune cells (microglia) in the dentate gyrus of the hippocampus in 13 month-old male and female rats. RESULTS We observed basal sex differences in neurogenesis at middle age, as wildtype male rats had greater densities of neural progenitor cells and new-born neurons in the dentate gyrus than wildtype female rats. Male hAPOEε4 rats exhibited fewer neural progenitor cells, fewer new-born neurons, and more microglia than male wildtype rats. On the other hand, female hAPOEε4 rats exhibited more new-born neurons than female wildtype rats. Interestingly, females had more microglia than males regardless of genotype. Correlations were conducted to further elucidate any sex differences in the relationships between these biomarkers. Notably, there was a significant positive correlation between neural progenitor cells and new-born neurons, and a significant negative correlation between new-born neurons and microglia, but only in male rats. CONCLUSION In contrast to the clear pattern of effects of the hAPOEε4 risk factor on hippocampal neurogenesis in males, females had unaltered levels of neural progenitor cells and increased density of new-born neurons. Furthermore, relationships between neurogenesis and microglia were significantly correlated within males, and not females. This suggests that females may be presenting a compensatory response to the hAPOEε4 genotype at middle age. Collectively, these results exemplify the importance of thoroughly examining influences of sex on AD endophenotypes, as it may reveal sex-specific pathways and protective mechanisms relevant to AD.
Collapse
Affiliation(s)
- Bonnie H Lee
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Melike Cevizci
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie E Lieblich
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
8
|
Ravichandran S, Snyder PJ, Alber J, Murchison CF, Chaby LE, Jeromin A, Arthur E. Association and multimodal model of retinal and blood-based biomarkers for detection of preclinical Alzheimer's disease. Alzheimers Res Ther 2025; 17:19. [PMID: 39794837 PMCID: PMC11720872 DOI: 10.1186/s13195-024-01668-5] [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: 07/25/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
BACKGROUND The potential diagnostic value of plasma amyloidogenic beta residue 42/40 ratio (Aβ42/Aβ40 ratio), neurofilament light (NfL), tau phosphorylated at threonine-181 (p-tau181), and threonine-217 (p-tau217) has been extensively discussed in the literature. We have also previously described the association between retinal biomarkers and preclinical Alzheimer's disease (AD). The goal of this study was to evaluate the association, and a multimodal model of, retinal and plasma biomarkers for detection of preclinical AD. METHODS We included 82 cognitively unimpaired (CU) participants (141 eyes; mean age: 67 years; range: 56-80) from the Atlas of Retinal Imaging in Alzheimer's Study (ARIAS). Blood samples were assessed for concentrations of Aβ42/Aβ40 ratio, NfL, p-tau181, and p-tau217 (ALZpath, Inc.) using Single molecule array (SIMOA) technology. The Spectralis II system (Heidelberg Engineering) was used to acquire macular centered Spectral Domain Optical Coherence Tomography (SD-OCT) images for evaluation of putative retinal gliosis surface area and macular retinal nerve fiber layer (mRNFL) thickness. For all participants, correlations (adjusted for age and correlation between eyes) were assessed between retinal and blood-based biomarkers. A subgroup cohort of 57 eyes from 32 participants with recent Aβ positron emission tomography (PET) results, comprising 18 preclinical patients (Aβ PET + ve, 32 eyes) and 14 controls (Aβ PET -ve, 25 eyes) with a mean age of 69 vs. 66, p = 0.06, was included for the assessment of a multimodal model to distinguish between the two groups. For this subgroup cohort, receiver operating characteristic (ROC) analysis was performed to compare the multimodal model of retinal and plasma biomarkers vs. each biomarker alone to distinguish between the two groups. RESULTS Significant correlation was found between putative retinal gliosis and p-tau217 in the univariate mixed model (β = 0.48, p = 0.007) but not for the other plasma biomarkers (p > 0.05). This positive correlation was also retained in the multivariate mixed model (β = 0.43, p = 0.022). The multimodal ROC model based on retinal (gliosis area, inner inferior RNFL thickness, inner superior RNFL thickness, and inner nasal RNFL thickness) and plasma biomarkers (p-tau217 and Aβ42/Aβ40 ratio) had an excellent AUC of 0.97 (95% CI = 0.93-1.01; p < 0.001) compared to unimodal models of retinal and plasma biomarkers. CONCLUSIONS Our analyses show the potential of integrating retinal and blood-based biomarkers for improved detection and screening of preclinical AD.
Collapse
Affiliation(s)
- Swetha Ravichandran
- School of Optometry, University of Alabama at Birmingham, Birmingham, AL, US
| | - Peter J Snyder
- Department of Neurology, Alpert Medical School of Brown University, Providence, RI, US
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, US
| | - Jessica Alber
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, US
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, US
- Butler Hospital Memory & Aging Program, Providence, RI, US
| | - Charles F Murchison
- Alzheimer's Disease Research Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, US
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, US
| | | | | | - Edmund Arthur
- School of Optometry, University of Alabama at Birmingham, Birmingham, AL, US.
| |
Collapse
|
9
|
Lehmann S, Schraen-Maschke S, Buée L, Vidal JS, Delaby C, Hirtz C, Blanc F, Paquet C, Allinquant B, Bombois S, Gabelle A, Hanon O. Clarifying the association of CSF Aβ, tau, BACE1, and neurogranin with AT(N) stages in Alzheimer disease. Mol Neurodegener 2024; 19:66. [PMID: 39380095 PMCID: PMC11460012 DOI: 10.1186/s13024-024-00755-3] [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: 05/12/2024] [Accepted: 09/13/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Current AT(N) stratification for Alzheimer's disease (AD) accounts for complex combinations of amyloid (A), tau proteinopathy (T) and neurodegeneration (N) signatures. Understanding the transition between these different stages is a major challenge, especially in view of the recent development of disease modifying therapy. METHODS This is an observational study, CSF levels of Tau, pTau181, pTau217, Aβ38/40/42, sAPPα/β, BACE1 and neurogranin were measured in the BALTAZAR cohort of cognitively impaired patients and in the Alzheimer's Disease Neuroimaging Initiative (ADNI). Biomarkers levels were related to the AT(N) framework. (A) and (T) were defined in BALTAZAR with CSF Aβ42/40 ratio and pTau217 respectively, and in ADNI with amyloid and tau PET. (N) was defined using total CSF tau in both cohorts. RESULTS As expected, CSF Aβ42 decreased progressively with the AD continuum going from the A-T-N- to the A + T + N + profile. On the other hand, Tau and pTau181 increased progressively with the disease. The final transition from A + T + N- to A + T + N + led to a sharp increase in Aβ38, Aβ42 and sAPP levels. Synaptic CSF biomarkers BACE1 and neurogranin, were lowest in the initial A + T-N- stage and increased with T + and N + . CSF pTau181 and total tau were closely related in both cohorts. CONCLUSIONS The early transition to an A + phenotype (A + T-N-) primarily impacts synaptic function. The appearance of T + and then N + is associated with a significant and progressive increase in pathological Alzheimer's disease biomarkers. Our main finding is that CSF pTau181 is an indicator of N + rather than T + , and that N + is associated with elevated levels of BACE1 protein and beta-amyloid peptides. This increase may potentially fuel the amyloid cascade in a positive feedback loop. Overall, our data provide further insights into understanding the interconnected pathological processes of amyloid, tau, and neurodegeneration underlying Alzheimer's disease.
Collapse
Affiliation(s)
- Sylvain Lehmann
- LBPC-PPC, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, 80 av Fliche, Montpellier, 34295, France.
| | - Susanna Schraen-Maschke
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, Lille, F-59000, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, Lille, F-59000, France
| | - Jean-Sébastien Vidal
- Université Paris Cité, EA 4468, APHP, Hospital Broca, Memory Resource and Research Centre of de Paris-Broca-Ile de France, Paris, F-75013, France
| | - Constance Delaby
- LBPC-PPC, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, 80 av Fliche, Montpellier, 34295, France
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Christophe Hirtz
- LBPC-PPC, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, 80 av Fliche, Montpellier, 34295, France
| | - Frédéric Blanc
- Université de Strasbourg, CHRU de Strasbourg, Memory Resource and Research Centre of Strasbourg/Colmar, French National Centre for Scientific Research (CNRS), ICube Laboratory and Fédération de Médecine Translationnelle de Strasbourg (FMTS), Team Imagerie Multimodale Intégrative en Santé (IMIS)/Neurocrypto, Strasbourg, F-67000, France
| | - Claire Paquet
- Université Paris Cité, GHU APHP Nord Lariboisière Fernand Widal, Centre de Neurologie Cognitive, Paris, F-75010, France
| | - Bernadette Allinquant
- UMR-S1266, Université Paris Cité, Institute of Psychiatry and Neuroscience, Inserm, Paris, France
| | - Stéphanie Bombois
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, Lille, F-59000, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neurologie, Centre des Maladies Cognitives et Comportementales, GH Pitié-Salpêtrière, Paris, France
| | - Audrey Gabelle
- Université de Montpellier, Memory Research and Resources Center, Department of Neurology, Inserm INM NeuroPEPs team, Montpellier, F-34000, France
| | - Olivier Hanon
- Université Paris Cité, EA 4468, APHP, Hospital Broca, Memory Resource and Research Centre of de Paris-Broca-Ile de France, Paris, F-75013, France
| |
Collapse
|
10
|
Sinha P, Turchyna Y, Mitchell SPC, Sadek M, Armagan G, Perrin F, Maesako M, Berezovska O. Glutamate Transporter 1 as a Novel Negative Regulator of Amyloid β. Cells 2024; 13:1600. [PMID: 39404364 PMCID: PMC11475981 DOI: 10.3390/cells13191600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Glutamate transporter-1 (GLT-1) dynamics are implicated in excitotoxicity and Alzheimer's disease (AD) progression. Early stages of AD are often marked by hyperactivity and increased epileptiform activity preceding cognitive decline. Previously, we identified a direct interaction between GLT-1 and Presenilin 1 (PS1) in the brain, highlighting GLT-1 as a promising target in AD research. This study reports the significance of this interaction and uncovers a novel role of GLT-1 in modulating amyloid-beta (Aβ) production. Overexpression of GLT-1 in cells reduces the levels of Aβ40 and Aβ42 by decreasing γ-secretase activity pertinent to APP processing and induces a more "open" PS1 conformation, resulting in decreased Aβ42/40 ratio. Inhibition of the GLT-1/PS1 interaction using cell-permeable peptides produced an opposing effect on Aβ, highlighting the pivotal role of this interaction in regulating Aβ levels. These findings emphasize the potential of targeting the GLT-1/PS1 interaction as a novel therapeutic strategy for AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Oksana Berezovska
- Alzheimer Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, 114, 16th Street, Charlestown, MA 02129, USA
| |
Collapse
|
11
|
Lee BH, Cevizci M, Lieblich SE, Ibrahim M, Wen Y, Eid RS, Lamers Y, Duarte-Guterman P, Galea LAM. Exploring the parity paradox: Differential effects on neuroplasticity and inflammation by APOEe4 genotype at middle age. Brain Behav Immun 2024; 120:54-70. [PMID: 38772427 DOI: 10.1016/j.bbi.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 04/20/2024] [Accepted: 05/18/2024] [Indexed: 05/23/2024] Open
Abstract
Female sex and Apolipoprotein E (APOE) ε4 genotype are top non-modifiable risk factors for Alzheimer's disease (AD). Although female-unique experiences like parity (pregnancy and motherhood) have positive effects on neuroplasticity at middle age, previous pregnancy may also contribute to AD risk. To explore these seemingly paradoxical long-term effects of parity, we investigated the impact of parity with APOEε4 genotype by examining behavioural and neural biomarkers of brain health in middle-aged female rats. Our findings show that primiparous (parous one time) hAPOEε4 rats display increased use of a non-spatial cognitive strategy and exhibit decreased number and recruitment of new-born neurons in the ventral dentate gyrus of the hippocampus in response to spatial working memory retrieval. Furthermore, primiparity and hAPOEε4 genotype synergistically modulate inflammatory markers in the ventral hippocampus. Collectively, these findings demonstrate that previous parity in hAPOEε4 rats confers an added risk to present with reduced activity and engagement of the hippocampus as well as elevated pro-inflammatory signaling, and underscore the importance of considering female-specific factors and genotype in health research.
Collapse
Affiliation(s)
- Bonnie H Lee
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Melike Cevizci
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie E Lieblich
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Muna Ibrahim
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Yanhua Wen
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Rand S Eid
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Yvonne Lamers
- Food Nutrition and Health Program, Faculty of Land and Food Systems, University of British Columbia, Vancouver, BC, Canada
| | - Paula Duarte-Guterman
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, ON, Canada.
| |
Collapse
|
12
|
Gyimesi M, Okolicsanyi RK, Haupt LM. Beyond amyloid and tau: rethinking Alzheimer's disease through less explored avenues. Open Biol 2024; 14:240035. [PMID: 38862019 DOI: 10.1098/rsob.240035] [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: 02/12/2024] [Accepted: 04/25/2024] [Indexed: 06/13/2024] Open
Abstract
Neurodegenerative diseases, particularly Alzheimer's disease (AD), pose a significant challenge in ageing populations. Our current understanding indicates that the onset of toxic amyloid and tau protein pathologies initiates disease progression. However, existing treatments targeting these hallmark symptoms offer symptomatic relief without halting disease advancement. This review offers an alternative perspective on AD, centring on impaired adult hippocampal neurogenesis (AHN) as a potential early aetiological factor. By delving into the intricate molecular events during the initial stages of AD (Braak Stages I-III), a novel hypothesis is presented, interweaving the roles of Notch signalling and heparan sulfate proteoglycans (HSPGs) in compromised AHN. While acknowledging the significance of the amyloid and tau hypotheses, it calls for further exploration beyond these paradigms, suggesting the potential of altered HS sulfation patterns in AD initiation. Future directions propose more detailed investigations into early HS aggregation, aberrant sulfation patterns and examination of their temporal relationship with tau hyperphosphorylation. In challenging the conventional 'triggers' of AD and urging their reconsideration as symptoms, this review advocates an alternative approach to understanding this disease, offering new avenues of investigation into the intricacies of AD pathogenesis.
Collapse
Affiliation(s)
- M Gyimesi
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave , Kelvin Grove, Queensland 4059, Australia
| | - R K Okolicsanyi
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave , Kelvin Grove, Queensland 4059, Australia
- Max Planck Queensland Centre for the Materials Sciences of Extracellular Matrices , Brisbane, QLD 4059, Australia
| | - L M Haupt
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave , Kelvin Grove, Queensland 4059, Australia
- Max Planck Queensland Centre for the Materials Sciences of Extracellular Matrices , Brisbane, QLD 4059, Australia
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), 60 Musk Ave , Kelvin Grove, Queensland 4059, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies , Brisbane, QLD 4059, Australia
| |
Collapse
|
13
|
Shahbaz MA, Kuivanen S, Mussalo L, Afonin AM, Kumari K, Behzadpour D, Kalapudas J, Koivisto AM, Penttilä E, Löppönen H, Jalava P, Vapalahti O, Balistreri G, Lampinen R, Kanninen KM. Exposure to urban particulate matter alters responses of olfactory mucosal cells to SARS-CoV-2 infection. ENVIRONMENTAL RESEARCH 2024; 249:118451. [PMID: 38341073 DOI: 10.1016/j.envres.2024.118451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Respiratory viruses have a significant impact on health, as highlighted by the COVID-19 pandemic. Exposure to air pollution can contribute to viral susceptibility and be associated with severe outcomes, as suggested by recent epidemiological studies. Furthermore, exposure to particulate matter (PM), an important constituent of air pollution, is linked to adverse effects on the brain, including cognitive decline and Alzheimer's disease (AD). The olfactory mucosa (OM), a tissue located at the rooftop of the nasal cavity, is directly exposed to inhaled air and in direct contact with the brain. Increasing evidence of OM dysfunction related to neuropathogenesis and viral infection demonstrates the importance of elucidating the interplay between viruses and air pollutants at the OM. This study examined the effects of subacute exposure to urban PM 0.2 and PM 10-2.5 on SARS-CoV-2 infection using primary human OM cells obtained from cognitively healthy individuals and individuals diagnosed with AD. OM cells were exposed to PM and subsequently infected with the SARS-CoV-2 virus in the presence of pollutants. SARS-CoV-2 entry receptors and replication, toxicological endpoints, cytokine release, oxidative stress markers, and amyloid beta levels were measured. Exposure to PM did not enhance the expression of viral entry receptors or cellular viral load in human OM cells. However, PM-exposed and SARS-CoV-2-infected cells showed alterations in cellular and immune responses when compared to cells infected only with the virus or pollutants. These changes are highly pronounced in AD OM cells. These results suggest that exposure of human OM cells to PM does not increase susceptibility to SARS-CoV-2 infection in vitro, but it can alter cellular immune responses to the virus, particularly in AD. Understanding the interplay of air pollutants and COVID-19 can provide important insight for the development of public health policies and interventions to reduce the negative influences of air pollution exposure.
Collapse
Affiliation(s)
- Muhammad Ali Shahbaz
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Kuopio, Finland
| | - Suvi Kuivanen
- University of Helsinki, Department of Virology, Faculty of Medicine, Helsinki, Finland; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany
| | - Laura Mussalo
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Kuopio, Finland
| | - Alexey M Afonin
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Kuopio, Finland
| | - Kajal Kumari
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Kuopio, Finland
| | - Donya Behzadpour
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Kuopio, Finland
| | - Juho Kalapudas
- University of Eastern Finland, Brain Research Unit, Department of Neurology, School of Medicine, Kuopio, Finland
| | - Anne M Koivisto
- University of Eastern Finland, Brain Research Unit, Department of Neurology, School of Medicine, Kuopio, Finland; Kuopio University Hospital, Department of Neurology, Neuro Centre, Kuopio, Finland; University of Helsinki, Faculty of Medicine, Department of Neurology and Geriatrics, Helsinki University Hospital and Neurosciences, Helsinki, Finland
| | - Elina Penttilä
- University of Eastern Finland and Kuopio University Hospital, Department of Otorhinolaryngology, Kuopio, Finland
| | - Heikki Löppönen
- University of Eastern Finland and Kuopio University Hospital, Department of Otorhinolaryngology, Kuopio, Finland
| | - Pasi Jalava
- University of Eastern Finland, Inhalation Toxicology Laboratory, Department of Environmental and Biological Sciences, Kuopio, Finland
| | - Olli Vapalahti
- University of Helsinki, Department of Virology, Faculty of Medicine, Helsinki, Finland
| | - Giuseppe Balistreri
- University of Helsinki, Department of Virology, Faculty of Medicine, Helsinki, Finland
| | - Riikka Lampinen
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Kuopio, Finland
| | - Katja M Kanninen
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Kuopio, Finland.
| |
Collapse
|
14
|
Mantellatto Grigoli M, Pelegrini LNC, Whelan R, Cominetti MR. Present and Future of Blood-Based Biomarkers of Alzheimer's Disease: Beyond the Classics. Brain Res 2024; 1830:148812. [PMID: 38369085 DOI: 10.1016/j.brainres.2024.148812] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/13/2023] [Accepted: 02/13/2024] [Indexed: 02/20/2024]
Abstract
The field of blood-based biomarkers for Alzheimer's disease (AD) has advanced at an incredible pace, especially after the development of sensitive analytic platforms that can facilitate large-scale screening. Such screening will be important when more sophisticated diagnostic methods are scarce and expensive. Thus, blood-based biomarkers can potentially reduce diagnosis inequities among populations from different socioeconomic contexts. This large-scale screening can be performed so that older adults at risk of cognitive decline assessed using these methods can then undergo more complete assessments with classic biomarkers, increasing diagnosis efficiency and reducing costs to the health systems. Blood-based biomarkers can also aid in assessing the effect of new disease-modifying treatments. This paper reviews recent advances in the area, focusing on the following leading candidates for blood-based biomarkers: amyloid-beta (Aβ), phosphorylated tau isoforms (p-tau), neurofilament light (NfL), and glial fibrillary acidic (GFAP) proteins, as well as on new candidates, Neuron-Derived Exosomes contents (NDEs) and Transactive response DNA-binding protein-43 (TDP-43), based on data from longitudinal observational cohort studies. The underlying challenges of validating and incorporating these biomarkers into routine clinical practice and primary care settings are also discussed. Importantly, challenges related to the underrepresentation of ethnic minorities and socioeconomically disadvantaged persons must be considered. If these challenges are overcome, a new time of cost-effective blood-based biomarkers for AD could represent the future of clinical procedures in the field and, together with continued prevention strategies, the beginning of an era with a lower incidence of dementia worldwide.
Collapse
Affiliation(s)
| | | | - Robert Whelan
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland; School of Psychology, Trinity College Dublin, Dublin, Ireland
| | - Marcia R Cominetti
- Department of Gerontology, Federal University of São Carlos, Brazil; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
15
|
Delaby C, Lehmann S. [On the road to biological blood diagnosis of Alzheimer's disease?]. Med Sci (Paris) 2024; 40:351-360. [PMID: 38651960 DOI: 10.1051/medsci/2024037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
The growing number of people suffering from Alzheimer's disease (AD) represents a major public health problem. The diagnosis of AD is multidisciplinary and involves the use of amyloid and tau biomarkers measured in cerebrospinal fluid. Recent advances in analytical techniques now allow us to measure these biomarkers in blood. Blood biomarkers offer particularly promising potential for early, minimally invasive detection of AD, as well as for differential diagnosis of dementia and patient follow-up. The aim of this review is to provide an overview of current and candidate blood biomarkers for AD, their informative value, and their potential to be integrated into clinical practice in the near future.
Collapse
Affiliation(s)
- Constance Delaby
- LBPC-PPC, Université Montpellier, CHU Montpellier, INM Inserm U1298, Montpellier, France - Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau - Universitat Autònoma de Barcelona, Barcelone, Espagne
| | - Sylvain Lehmann
- LBPC-PPC, Université Montpellier, CHU Montpellier, INM Inserm U1298, Montpellier, France
| |
Collapse
|
16
|
Jia Z, Maghaydah Y, Zdanys K, Kuchel GA, Diniz BS, Liu C. CRISPR-Powered Aptasensor for Diagnostics of Alzheimer's Disease. ACS Sens 2024; 9:398-405. [PMID: 38154140 DOI: 10.1021/acssensors.3c02167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and the most common cause of dementia, characterized by the accumulation of amyloid beta (Aβ) peptides in the brain. Here, we present a simple, rapid, and affordable CRISPR-powered aptasensor for the quantitative detection of Aβ40 and Aβ42 biomarkers in cerebrospinal fluid (CSF) samples, enabling early and accurate diagnostics of AD patients. The aptasensor couples the high specificity of aptamers for Aβ biomarkers with CRISPR-Cas12a-based fluorescence detection. The CRISPR-powered aptasensor enables us to detect Aβ40 and Aβ42 in CSF samples within 60 min, achieving a detection sensitivity of 1 pg/mL and 0.1 pg/mL, respectively. To validate its clinical utility, we quantitatively detected Aβ40 and Aβ42 biomarkers in clinical CSF samples. Furthermore, by combining CSF Aβ42 levels with the c(Aβ42)/c(Aβ40) ratio, we achieved an accurate diagnostic classification of AD patients and healthy individuals, showing superior performance over the conventional ELISA method. We believe that our innovative aptasensor approach holds promise for the early diagnostic classification of AD patients.
Collapse
Affiliation(s)
- Zhengyang Jia
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
| | - Yazeed Maghaydah
- Center on Aging, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
| | - Kristina Zdanys
- Department of Psychiatry, Division of Geriatric Psychiatry and Behavioral Health, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
| | - George A Kuchel
- Center on Aging, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
| | - Breno Satler Diniz
- Center on Aging, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
- Department of Psychiatry, Division of Geriatric Psychiatry and Behavioral Health, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
| | - Changchun Liu
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
| |
Collapse
|
17
|
Appleman ML, Thomas JL, Weiss AR, Nilaver BI, Cervera-Juanes R, Kohama SG, Urbanski HF. Effect of hormone replacement therapy on amyloid beta (Aβ) plaque density in the rhesus macaque amygdala. Front Aging Neurosci 2024; 15:1326747. [PMID: 38274989 PMCID: PMC10808750 DOI: 10.3389/fnagi.2023.1326747] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/27/2023] [Indexed: 01/27/2024] Open
Abstract
Background Amyloid beta (Aβ) plaque density was examined in the amygdala of rhesus macaques, to elucidate the influence of age, diet and hormonal environment. Methods Luminex technology was used to measure cerebrospinal fluid (CSF) concentrations of Aβ40 and Aβ42 across three decades, while immunohistochemistry was used to examine Aβ plaque density in the amygdala. Results Aβ40 was found to be the predominant isoform of Aβ in the CSF, but neither Aβ40 or Aβ42 concentrations showed an age-related change, and the ratio of Aβ42 to Aβ40 showed only a marginal increase. Significantly fewer Aβ plaques were detected in the amygdala of old ovariectomized animals if they received estradiol HRT (p < 0.001); similar results were obtained regardless of whether they had been maintained on a regular monkey chow for ∼48 months or on a high-fat, high-sugar, Western-style diet for ∼30 months. Conclusion The results demonstrate that HRT involving estrogen can reduce Aβ plaque load in a cognitive brain region of aged non-human primates. The results from this translational animal model may therefore have clinical relevance to the treatment of AD in post-menopausal women, whether used alone, or as a supplement to current pharmacological and monoclonal antibody-based interventions.
Collapse
Affiliation(s)
- Maria-Luisa Appleman
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Jeremy L. Thomas
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Alison R. Weiss
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Benjamin I. Nilaver
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Rita Cervera-Juanes
- Department of Physiology and Pharmacology, Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC, United States
| | - Steven G. Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Henryk F. Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, United States
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
18
|
Sánchez‐Soblechero A, Berbel A, Villarejo A, Palmí‐Cortés I, Vieira A, Gil‐Moreno MJ, Fernández C, Martín‐Montes Ã, Carreras MT, Fernández Y, Puertas C, Blanco‐Palmero V, Llamas S, González‐Sánchez M, Lapeña T, de Luis P, Manzano S, Olazarán J. Translating NIA-AA criteria into usual practice: Report from the ReDeMa Project. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12451. [PMID: 38505833 PMCID: PMC10948948 DOI: 10.1002/trc2.12451] [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: 10/06/2023] [Revised: 12/23/2023] [Accepted: 12/30/2023] [Indexed: 03/21/2024]
Abstract
INTRODUCTION Biomarker-informed criteria were proposed for the diagnosis of Alzheimer's disease (AD) by the National Institute on Aging and the Alzheimer's Association (NIA-AA) in 2011; however, the adequacy of this criteria has not been sufficiently evaluated. METHODS ReDeMa (Red de Demencias de Madrid) is a regional cohort of patients attending memory and neurology clinics. Core cerebrospinal fluid biomarkers were obtained, NIA-AA diagnostic criteria were considered, and changes in diagnosis and management were evaluated. RESULTS A total of 233 patients were analyzed (mean age 70 years, 50% women, 73% AD). The diagnostic language was modified significantly, with a majority assumption of NIA-AA definitions (69%). Confidence in diagnosis increased from 70% to 92% (p < 0.0005) and management was changed in 71% of patient/caregivers. The influence of neurologist's age or expertise on study results was minimal. DISCUSSION The NIA-AA criteria are adequate and utile for usual practice in memory and neurology clinics, improving diagnostic confidence and significantly modifying patient management. HIGHLIGHTS Alzheimer's disease (AD) cerebrospinal fluid (CSF) biomarkers increase diagnostic certainty regardless of the neurologist.AD CSF biomarkers lead to changes in disease management .Biomarker-enriched, 2011 NIA-AA diagnostic criteria are adequate for usual practice.
Collapse
Affiliation(s)
| | | | | | - Itziar Palmí‐Cortés
- Neurology ServiceUniversity Hospital Infanta Sofía, San Sebastián de los ReyesMadridSpain
| | - Alba Vieira
- Neurology ServiceUniversity Hospital la PrincesaMadridSpain
| | | | | | - Ãngel Martín‐Montes
- Hospital La Paz Institute for Health Research – IdiPAZ (La Paz University Hospital – Universidad Autónoma de Madrid)MadridSpain
| | | | - Yolanda Fernández
- Memory Disorders Clinic ‐ HM Hospitals and Neurology Service ‐ University Hospital Gregorio MarañónMadridSpain
| | - Carolina Puertas
- Clinical Biochemistry ServiceUniversity Hospital Gregorio MarañónMadridSpain
| | | | - Sara Llamas
- Neurology ServiceUniversity Hospital 12 de OctubreMadridSpain
| | - Marta González‐Sánchez
- Neurology ServiceUniversity Hospital 12 de OctubreMadridSpain
- Group of Neurodegenerative DiseasesUniversity Hospital 12 de Octubre Research Institute (imas12), and Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED)MadridSpain
| | | | | | | | - Javier Olazarán
- Memory Disorders Clinic ‐ HM HospitalsNeurology Service ‐ University Hospital Gregorio Marañón, and Maria Wolff FoundationMadridSpain
| |
Collapse
|
19
|
Jones KT, Gallen CL, Ostrand AE, Rojas JC, Wais P, Rini J, Chan B, Lago AL, Boxer A, Zhao M, Gazzaley A, Zanto TP. Gamma neuromodulation improves episodic memory and its associated network in amnestic mild cognitive impairment: a pilot study. Neurobiol Aging 2023; 129:72-88. [PMID: 37276822 PMCID: PMC10583532 DOI: 10.1016/j.neurobiolaging.2023.04.005] [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: 09/06/2022] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 06/07/2023]
Abstract
Amnestic mild cognitive impairment (aMCI) is a predementia stage of Alzheimer's disease associated with dysfunctional episodic memory and limited treatment options. We aimed to characterize feasibility, clinical, and biomarker effects of noninvasive neurostimulation for aMCI. 13 individuals with aMCI received eight 60-minute sessions of 40-Hz (gamma) transcranial alternating current stimulation (tACS) targeting regions related to episodic memory processing. Feasibility, episodic memory, and plasma Alzheimer's disease biomarkers were assessed. Neuroplastic changes were characterized by resting-state functional connectivity (RSFC) and neuronal excitatory/inhibitory balance. Gamma tACS was feasible and aMCI participants demonstrated improvement in multiple metrics of episodic memory, but no changes in biomarkers. Improvements in episodic memory were most pronounced in participants who had the highest modeled tACS-induced electric fields and exhibited the greatest changes in RSFC. Increased RSFC was also associated with greater hippocampal excitability and higher baseline white matter integrity. This study highlights initial feasibility and the potential of gamma tACS to rescue episodic memory in an aMCI population by modulating connectivity and excitability within an episodic memory network.
Collapse
Affiliation(s)
- Kevin T Jones
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA.
| | - Courtney L Gallen
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA
| | - Avery E Ostrand
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA
| | - Julio C Rojas
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Weill Institute for Neurosciences, Memory and Aging Center, University of California-San Francisco, San Francisco, CA
| | - Peter Wais
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA
| | - James Rini
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA
| | - Brandon Chan
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Weill Institute for Neurosciences, Memory and Aging Center, University of California-San Francisco, San Francisco, CA
| | - Argentina Lario Lago
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Weill Institute for Neurosciences, Memory and Aging Center, University of California-San Francisco, San Francisco, CA
| | - Adam Boxer
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Weill Institute for Neurosciences, Memory and Aging Center, University of California-San Francisco, San Francisco, CA
| | - Min Zhao
- Departments of Ophthalmology and Vision Science and Dermatology, Institute for Regenerative Cures, University of California-Davis, Davis, CA
| | - Adam Gazzaley
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA; Departments of Physiology and Psychiatry, University of California-San Francisco, San Francisco, CA
| | - Theodore P Zanto
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA.
| |
Collapse
|
20
|
Vadukul D, Papp M, Thrush RJ, Wang J, Jin Y, Arosio P, Aprile FA. α-Synuclein Aggregation Is Triggered by Oligomeric Amyloid-β 42 via Heterogeneous Primary Nucleation. J Am Chem Soc 2023; 145:18276-18285. [PMID: 37556728 PMCID: PMC10450681 DOI: 10.1021/jacs.3c03212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Indexed: 08/11/2023]
Abstract
An increasing number of cases where amyloids of different proteins are found in the same patient are being reported. This observation complicates diagnosis and clinical intervention. Amyloids of the amyloid-β peptide or the protein α-synuclein are traditionally considered hallmarks of Alzheimer's and Parkinson's diseases, respectively. However, the co-occurrence of amyloids of these proteins has also been reported in patients diagnosed with either disease. Here, we show that soluble species containing amyloid-β can induce the aggregation of α-synuclein. Fibrils formed under these conditions are solely composed of α-synuclein to which amyloid-β can be found associated but not as part of the core of the fibrils. Importantly, by global kinetic analysis, we found that the aggregation of α-synuclein under these conditions occurs via heterogeneous primary nucleation, triggered by soluble aggregates containing amyloid-β.
Collapse
Affiliation(s)
- Devkee
M. Vadukul
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.
| | - Marcell Papp
- Department
of Chemistry and Applied Biosciences, Institute
for Chemical and Bioengineering, Swiss Federal Institute of Technology, 8093 Zurich, Switzerland
| | - Rebecca J. Thrush
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.
- Institute
of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.
| | - Jielei Wang
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.
| | - Yiyun Jin
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.
| | - Paolo Arosio
- Department
of Chemistry and Applied Biosciences, Institute
for Chemical and Bioengineering, Swiss Federal Institute of Technology, 8093 Zurich, Switzerland
| | - Francesco A. Aprile
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.
- Institute
of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.
| |
Collapse
|
21
|
Yosypyshyn D, Kučikienė D, Ramakers I, Schulz JB, Reetz K, Costa AS. Clinical characteristics of patients with suspected Alzheimer's disease within a CSF Aß-ratio grey zone. Neurol Res Pract 2023; 5:40. [PMID: 37533121 PMCID: PMC10398972 DOI: 10.1186/s42466-023-00262-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/28/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND The AT(N) research framework for Alzheimer's disease (AD) remains unclear on how to best deal with borderline cases. Our aim was to characterise patients with suspected AD with a borderline Aß1-42/Aß1-40 ratio in cerebrospinal fluid. METHODS We analysed retrospective data from two cohorts (memory clinic cohort and ADNI) of patients (n = 63) with an Aß1-42/Aß1-40 ratio within a predefined borderline area-Q1 above the validated cut-off value(grey zone). We compared demographic, clinical, neuropsychological and neuroimaging features between grey zone patients and patients with low Aß1-42 (normal Aß ratio but pathological Aß1-42, n = 42) and patients with AD (pathological Aß, P-Tau, und T-Tau, n = 80). RESULTS Patients had mild cognitive impairment or mild dementia and a median age of 72 years. Demographic and general clinical characteristics did not differ between the groups. Patients in the grey zone group were the least impaired in cognition. However, they overlapped with the low Aß1-42 group in verbal episodic memory performance, especially in delayed recall and recognition. The grey zone group had less severe medial temporal atrophy, but mild posterior atrophy and mild white matter hyperintensities, similar to the low Aß1-42 group. CONCLUSIONS Patients in the Aß ratio grey zone were less impaired, but showed clinical overlap with patients on the AD continuum. These borderline patients may be at an earlier disease stage. Assuming an increased risk of AD and progressive cognitive decline, careful consideration of clinical follow-up is recommended when using dichotomous approaches to classify Aß status.
Collapse
Affiliation(s)
- Dariia Yosypyshyn
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Domantė Kučikienė
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Inez Ramakers
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Jörg B Schulz
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
- JARA Institute Molecular Neuroscience and Neuroimaging, RWTH Aachen & Forschungszentrum Jülich, Aachen, Germany
| | - Kathrin Reetz
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany.
- JARA Institute Molecular Neuroscience and Neuroimaging, RWTH Aachen & Forschungszentrum Jülich, Aachen, Germany.
| | - Ana Sofia Costa
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
- JARA Institute Molecular Neuroscience and Neuroimaging, RWTH Aachen & Forschungszentrum Jülich, Aachen, Germany
| |
Collapse
|
22
|
Xu Y, Jiang H, Zhu B, Cao M, Feng T, Sun Z, Du G, Zhao Z. Advances and applications of fluids biomarkers in diagnosis and therapeutic targets of Alzheimer's disease. CNS Neurosci Ther 2023. [PMID: 37144603 DOI: 10.1111/cns.14238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/25/2023] [Accepted: 04/12/2023] [Indexed: 05/06/2023] Open
Abstract
AIMS Alzheimer's disease (AD) is a neurodegenerative disease with challenging early diagnosis and effective treatments due to its complex pathogenesis. AD patients are often diagnosed after the appearance of the typical symptoms, thereby delaying the best opportunity for effective measures. Biomarkers could be the key to resolving the challenge. This review aims to provide an overview of application and potential value of AD biomarkers in fluids, including cerebrospinal fluid, blood, and saliva, in diagnosis and treatment. METHODS A comprehensive search of the relevant literature was conducted to summarize potential biomarkers for AD in fluids. The paper further explored the biomarkers' utility in disease diagnosis and drug target development. RESULTS Research on biomarkers mainly focused on amyloid-β (Aβ) plaques, Tau protein abnormal phosphorylation, axon damage, synaptic dysfunction, inflammation, and related hypotheses associated with AD mechanisms. Aβ42 , total Tau (t-Tau), and phosphorylated Tau (p-Tau), have been endorsed for their diagnostic and predictive capability. However, other biomarkers remain controversial. Drugs targeting Aβ have shown some efficacy and those that target BACE1 and Tau are still undergoing development. CONCLUSION Fluid biomarkers hold considerable potential in the diagnosis and drug development of AD. However, improvements in sensitivity and specificity, and approaches for managing sample impurities, need to be addressed for better diagnosis.
Collapse
Affiliation(s)
- Yanan Xu
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- School of Pharmacy, Capital Medical University, Beijing, China
| | - Hailun Jiang
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bin Zhu
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mingnan Cao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Feng
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhongshi Sun
- Department of Pharmacy, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhigang Zhao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- School of Pharmacy, Capital Medical University, Beijing, China
| |
Collapse
|
23
|
Lehmann S, Schraen-Maschke S, Vidal JS, Delaby C, Blanc F, Paquet C, Allinquant B, Bombois S, Gabelle A, Hanon O. Plasma phosphorylated tau 181 predicts amyloid status and conversion to dementia stage dependent on renal function. J Neurol Neurosurg Psychiatry 2023; 94:411-419. [PMID: 37012068 DOI: 10.1136/jnnp-2022-330540] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/15/2022] [Indexed: 04/05/2023]
Abstract
OBJECTIVES Plasma P-tau181 is an increasingly established diagnostic marker for Alzheimer's disease (AD). Further validation in prospective cohorts is still needed, as well as the study of confounding factors that could influence its blood level. METHODS This study is ancillary to the prospective multicentre Biomarker of AmyLoid pepTide and AlZheimer's diseAse Risk cohort that enrolled participants with mild cognitive impairment (MCI) who were examined for conversion to dementia for up to 3 years. Plasma Ptau-181 was measured using the ultrasensitive Quanterix HD-X assay. RESULTS Among 476 MCI participants, 67% were amyloid positive (Aβ+) at baseline and 30% developed dementia. Plasma P-tau181 was higher in the Aβ+ population (3.9 (SD 1.4) vs 2.6 (SD 1.4) pg/mL) and in MCI that converted to dementia (3.8 (SD 1.5) vs 2.9 (SD 1.4) pg/mL). The addition of plasma P-tau181 to a logistic regression model combining age, sex, APOEε4 status and Mini Mental State Examination improved predictive performance (areas under the curve 0.691-0.744 for conversion and 0.786-0.849 for Aβ+). The Kaplan-Meier curve of conversion to dementia, according to the tertiles of plasma P-tau181, revealed a significant predictive value (Log rank p<0.0001) with an HR of 3.8 (95% CI 2.5 to 5.8). In addition, patients with plasma P-Tau(181) ≤2.32 pg/mL had a conversion rate of less than 20% over a 3-year period. Using a linear regression approach, chronic kidney disease, creatinine and estimated glomerular filtration rate were independently associated with plasma P-tau181 concentrations. CONCLUSIONS Plasma P-tau181 effectively detects Aβ+ status and conversion to dementia, confirming the value of this blood biomarker for the management of AD. However, renal function significantly modifies its levels and may thus induce diagnostic errors if not taken into account.
Collapse
Affiliation(s)
- Sylvain Lehmann
- LBPC-PPC, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, Montpellier, France
| | | | - Jean-Sébastien Vidal
- Université Paris Cité, EA 4468, Hopital Broca, Geriatric department, Memory Resource and Research Centre of Paris-Broca-Ile de France, APHP, Paris, France
| | - Constance Delaby
- LBPC-PPC, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, Montpellier, France
- Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Frédéric Blanc
- Neuropsychology Unit and Geriatric Day Hospital, University Hospital of Strasbourg, Strasbourg, France
| | - Claire Paquet
- Université Paris Cité, GHU APHP Nord Lariboisière Fernand Widal, Centre de Neurologie Cognitive, Paris, France
| | - Bernadette Allinquant
- UMR-S 1266, Université Paris Cité, Institute of Psychiatry and Neurosciences, Inserm, Paris, France
| | - Stéphanie Bombois
- Université de Lille, Inserm UMRS1172, CHU Lille, Lille, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neurologie, Centre des Maladies Cognitives et Comportementales, GH Pitié-Salpêtrière, Paris, France
| | - Audrey Gabelle
- Université de Montpellier, CHRU Montpellier, Memory Research and Resources center, department of Neurology, Inserm INM, Montpellier, France
| | - Olivier Hanon
- Université Paris Cité, EA 4468, Hopital Broca, Geriatric department, Memory Resource and Research Centre of Paris-Broca-Ile de France, APHP, Paris, France
| |
Collapse
|
24
|
Delaby C, Hirtz C, Lehmann S. Overview of the blood biomarkers in Alzheimer's disease: Promises and challenges. Rev Neurol (Paris) 2023; 179:161-172. [PMID: 36371265 DOI: 10.1016/j.neurol.2022.09.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022]
Abstract
The increasing number of people with advanced Alzheimer's disease (AD) represents a significant psychological and financial cost to the world population. Accurate detection of the earliest phase of preclinical AD is of major importance for the success of preventive and therapeutic strategies (Cullen et al., 2021). Advances in analytical techniques have been essential for the development of sensitive, specific and reliable diagnostic tests for AD biomarkers in biological fluids (cerebrospinal fluid and blood). Blood biomarkers hold promising potential for early and minimally invasive detection of AD, but also for differential diagnosis of dementia and for monitoring the course of the disease. The aim of this review is to provide an overview of current blood biomarkers of AD, from tau proteins and amyloid peptides to biomarkers of neuronal degeneration and inflammation, reactive and metabolic factors. We thus discuss the informative value of currently candidate blood biomarkers and their potential to be integrated into clinical practice for the management of AD in the near future.
Collapse
Affiliation(s)
- C Delaby
- LBPC-PPC, Université Montpellier, CHU Montpellier, INM Inserm, Montpellier, France; Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
| | - C Hirtz
- LBPC-PPC, Université Montpellier, CHU Montpellier, INM Inserm, Montpellier, France
| | - S Lehmann
- LBPC-PPC, Université Montpellier, CHU Montpellier, INM Inserm, Montpellier, France.
| |
Collapse
|
25
|
Constantinides VC, Paraskevas GP, Boufidou F, Bourbouli M, Pyrgelis ES, Stefanis L, Kapaki E. CSF Aβ42 and Aβ42/Aβ40 Ratio in Alzheimer's Disease and Frontotemporal Dementias. Diagnostics (Basel) 2023; 13:diagnostics13040783. [PMID: 36832271 PMCID: PMC9955886 DOI: 10.3390/diagnostics13040783] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Alzheimer's disease dementia (ADD) may manifest with atypical phenotypes, resembling behavioral variant frontotemporal dementia (bvFTD) and corticobasal syndrome (CBS), phenotypes which typically have an underlying frontotemporal lobar degeneration with tau proteinopathy (FTLD-tau), such as Pick's disease, corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), or FTLD with TDP-43 proteinopathy (FTLD-TDP). CSF biomarkers total and phosphorylated tau (τT and τP-181), and amyloid beta with 42 and 40 amino acids (Aβ42 and Aβ40) are biomarkers of AD pathology. The primary aim of this study was to compare the diagnostic accuracy of Aβ42 to Aβ42/Aβ40 ratio in: (a) differentiating ADD vs. frontotemporal dementias; (b) patients with AD pathology vs. non-AD pathologies; (c) compare biomarker ratios and composite markers to single CSF biomarkers in the differentiation of AD from FTD; Methods: In total, 263 subjects were included (ADD: n = 98; bvFTD: n = 49; PSP: n = 50; CBD: n = 45; controls: n = 21). CSF biomarkers were measured by commercially available ELISAs (EUROIMMUN). Multiple biomarker ratios (Aβ42/Aβ40; τT/τP-181; τT/Aβ42; τP-181/Aβ42) and composite markers (t-tau: τT/(Aβ42/Aβ40); p-tau: τP-181/(Aβ42/Aβ40) were calculated. ROC curve analysis was performed to compare AUCs of Aβ42 and Aβ42/Aβ40 ratio and relevant composite markers between ADD and FTD, as defined clinically. BIOMARKAPD/ABSI criteria (abnormal τT, τP-181 Aβ42, and Aβ42/Aβ40 ratio) were used to re-classify all patients into AD pathology vs. non-AD pathologies, and ROC curve analysis was repeated to compare Aβ42 and Aβ42/Aβ40; Results: Aβ42 did not differ from Aβ42/Aβ40 ratio in the differentiation of ADD from FTD (AUCs 0.752 and 0.788 respectively; p = 0.212). The τT/Aβ42 ratio provided maximal discrimination between ADD and FTD (AUC:0.893; sensitivity 88.8%, specificity 80%). BIOMARKAPD/ABSI criteria classified 60 patients as having AD pathology and 211 as non-AD. A total of 22 had discrepant results and were excluded. Aβ42/Aβ40 ratio was superior to Aβ42 in the differentiation of AD pathology from non-AD pathology (AUCs: 0.939 and 0.831, respectively; p < 0.001). In general, biomarker ratios and composite markers were superior to single CSF biomarkers in both analyses. CONCLUSIONS Aβ42/Aβ40 ratio is superior to Aβ42 in identifying AD pathology, irrespective of the clinical phenotype. CSF biomarker ratios and composite markers provide higher diagnostic accuracy compared to single CSF biomarkers.
Collapse
Affiliation(s)
- Vasilios C. Constantinides
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Correspondence: ; Tel.: +30-21-0728-9285
| | - George P. Paraskevas
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Second Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, Rimini 1, 12462 Athens, Greece
| | - Fotini Boufidou
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Mara Bourbouli
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Efstratios-Stylianos Pyrgelis
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Leonidas Stefanis
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Elisabeth Kapaki
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| |
Collapse
|
26
|
Hirtz C, Busto GU, Bennys K, Kindermans J, Navucet S, Tiers L, Lista S, Vialaret J, Gutierrez LA, Dauvilliers Y, Berr C, Lehmann S, Gabelle A. Comparison of ultrasensitive and mass spectrometry quantification of blood-based amyloid biomarkers for Alzheimer's disease diagnosis in a memory clinic cohort. Alzheimers Res Ther 2023; 15:34. [PMID: 36800984 PMCID: PMC9938625 DOI: 10.1186/s13195-023-01188-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/09/2023] [Indexed: 02/20/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex neurodegenerative disorder with β-amyloid pathology as a key underlying process. The relevance of cerebrospinal fluid (CSF) and brain imaging biomarkers is validated in clinical practice for early diagnosis. Yet, their cost and perceived invasiveness are a limitation for large-scale implementation. Based on positive amyloid profiles, blood-based biomarkers should allow to detect people at risk for AD and to monitor patients under therapeutics strategies. Thanks to the recent development of innovative proteomic tools, the sensibility and specificity of blood biomarkers have been considerably improved. However, their diagnosis and prognosis relevance for daily clinical practice is still incomplete. METHODS The Plasmaboost study included 184 participants from the Montpellier's hospital NeuroCognition Biobank with AD (n = 73), mild cognitive impairments (MCI) (n = 32), subjective cognitive impairments (SCI) (n = 12), other neurodegenerative diseases (NDD) (n = 31), and other neurological disorders (OND) (n = 36). Dosage of β-amyloid biomarkers was performed on plasma samples using immunoprecipitation-mass spectrometry (IPMS) developed by Shimadzu (IPMS-Shim Aβ42, Aβ40, APP669-711) and Simoa Human Neurology 3-PLEX A assay (Aβ42, Aβ40, t-tau). Links between those biomarkers and demographical and clinical data and CSF AD biomarkers were investigated. Performances of the two technologies to discriminate clinically or biologically based (using the AT(N) framework) diagnosis of AD were compared using receiver operating characteristic (ROC) analyses. RESULTS The amyloid IPMS-Shim composite biomarker (combining APP669-711/Aβ42 and Aβ40/Aβ42 ratios) discriminated AD from SCI (AUC: 0.91), OND (0.89), and NDD (0.81). The IPMS-Shim Aβ42/40 ratio also discriminated AD from MCI (0.78). IPMS-Shim biomarkers have similar relevance to discriminate between amyloid-positive and amyloid-negative individuals (0.73 and 0.76 respectively) and A-T-N-/A+T+N+ profiles (0.83 and 0.85). Performances of the Simoa 3-PLEX Aβ42/40 ratio were more modest. Pilot longitudinal analysis on the progression of plasma biomarkers indicates that IPMS-Shim can detect the decrease in plasma Aβ42 that is specific to AD patients. CONCLUSIONS Our study confirms the potential usefulness of amyloid plasma biomarkers, especially the IPMS-Shim technology, as a screening tool for early AD patients.
Collapse
Affiliation(s)
- Christophe Hirtz
- grid.157868.50000 0000 9961 060XUniversity of Montpellier, IRMB-PPC, INM, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Germain U. Busto
- grid.157868.50000 0000 9961 060XResource and Research Memory Center (CMRR), Department of Neurology, Montpellier University Hospital, 80 avenue Augustin Fliche, 34000 Montpellier, France ,grid.121334.60000 0001 2097 0141Institute for Neurosciences of Montpellier (INM), Univ Montpellier, INSERM, Montpellier, France
| | - Karim Bennys
- grid.157868.50000 0000 9961 060XResource and Research Memory Center (CMRR), Department of Neurology, Montpellier University Hospital, 80 avenue Augustin Fliche, 34000 Montpellier, France
| | - Jana Kindermans
- grid.157868.50000 0000 9961 060XUniversity of Montpellier, IRMB-PPC, INM, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Sophie Navucet
- grid.157868.50000 0000 9961 060XResource and Research Memory Center (CMRR), Department of Neurology, Montpellier University Hospital, 80 avenue Augustin Fliche, 34000 Montpellier, France
| | - Laurent Tiers
- grid.157868.50000 0000 9961 060XUniversity of Montpellier, IRMB-PPC, INM, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Simone Lista
- grid.157868.50000 0000 9961 060XResource and Research Memory Center (CMRR), Department of Neurology, Montpellier University Hospital, 80 avenue Augustin Fliche, 34000 Montpellier, France
| | - Jérôme Vialaret
- grid.157868.50000 0000 9961 060XUniversity of Montpellier, IRMB-PPC, INM, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Laure-Anne Gutierrez
- grid.121334.60000 0001 2097 0141Institute for Neurosciences of Montpellier (INM), Univ Montpellier, INSERM, Montpellier, France
| | - Yves Dauvilliers
- grid.121334.60000 0001 2097 0141Institute for Neurosciences of Montpellier (INM), Univ Montpellier, INSERM, Montpellier, France ,grid.121334.60000 0001 2097 0141Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, University of Montpellier, Montpellier, France
| | - Claudine Berr
- grid.121334.60000 0001 2097 0141Institute for Neurosciences of Montpellier (INM), Univ Montpellier, INSERM, Montpellier, France
| | - Sylvain Lehmann
- University of Montpellier, IRMB-PPC, INM, CHU Montpellier, INSERM CNRS, Montpellier, France.
| | - Audrey Gabelle
- grid.157868.50000 0000 9961 060XResource and Research Memory Center (CMRR), Department of Neurology, Montpellier University Hospital, 80 avenue Augustin Fliche, 34000 Montpellier, France ,grid.121334.60000 0001 2097 0141Institute for Neurosciences of Montpellier (INM), Univ Montpellier, INSERM, Montpellier, France
| |
Collapse
|
27
|
Czerminski JT, King OD, Lawrence JB. Large-scale organoid study suggests effects of trisomy 21 on early fetal neurodevelopment are more subtle than variability between isogenic lines and experiments. Front Neurosci 2023; 16:972201. [PMID: 36817096 PMCID: PMC9935940 DOI: 10.3389/fnins.2022.972201] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 12/08/2022] [Indexed: 02/05/2023] Open
Abstract
This study examines cortical organoids generated from a panel of isogenic trisomic and disomic iPSC lines (subclones) as a model of early fetal brain development in Down syndrome (DS). An initial experiment comparing organoids from one trisomic and one disomic line showed many genome-wide transcriptomic differences and modest differences in cell-type proportions, suggesting there may be a neurodevelopmental phenotype that is due to trisomy of chr21. To better control for multiple sources of variation, we undertook a highly robust study of ∼1,200 organoids using an expanded panel of six all-isogenic lines, three disomic, and three trisomic. The power of this experimental design was indicated by strong detection of the ∼1.5-fold difference in chr21 genes. However, the numerous expression differences in non-chr21 genes seen in the smaller experiment fell away, and the differences in cell-type representation between lines did not correlate with trisomy 21. Results suggest that the initial smaller experiment picked up differences between small organoid samples and individual isogenic lines, which "averaged out" in the larger panel of isogenic lines. Our results indicate that even when organoid and batch variability are better controlled for, variation between isogenic cell lines (even subclones) may obscure, or be conflated with, subtle neurodevelopmental phenotypes that may be present in ∼2nd trimester DS brain development. Interestingly, despite this variability between organoid batches and lines, and the "fetal stage" of these organoids, an increase in secreted Aβ40 peptide levels-an Alzheimer-related cellular phenotype-was more strongly associated with trisomy 21 status than were neurodevelopmental shifts in cell-type composition.
Collapse
Affiliation(s)
- Jan T. Czerminski
- Medical Scientist Training Program, Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Oliver D. King
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Jeanne B. Lawrence
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, United States,Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, United States,*Correspondence: Jeanne B. Lawrence,
| |
Collapse
|
28
|
Utz J, Olm P, Jablonowski J, Siegmann EM, Spitzer P, Lewczuk P, Kornhuber J, Maler JM, Oberstein TJ. Reconceptualization of the Erlangen Score for the Assessment of Dementia Risk: The ERlangen Score. J Alzheimers Dis 2023; 96:265-275. [PMID: 37742651 PMCID: PMC10657695 DOI: 10.3233/jad-230524] [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] [Accepted: 08/17/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND The established Erlangen Score (ES) for the interpretation of cerebrospinal fluid (CSF) biomarkers in the diagnostics of Alzheimer's disease (AD) uses markers of amyloidopathy and tauopathy, equally weighted to form an easy-interpretable ordinal scale. However, these biomarkers are not equally predictive for AD. OBJECTIVE The higher weighting of the Aβ42/Aβ40 ratio, as a reconceptualized ERlangen Score (ERS), was tested for advantages in diagnostic performance. METHODS Non-demented subjects (N = 154) with a mean follow up of 5 years were assigned to a group ranging from 0 to 4 in ES or ERS. Psychometric trajectories and dementia risk were assessed. RESULTS The distribution of subjects between ES and ERS among the groups differed considerably, as grouping allocated 32 subjects to ES group 2, but only 2 to ERS group 2. The discriminative accuracy between the ES (AUC 73.2%, 95% CI [64.2, 82.2]) and ERS (AUC 72.0%, 95% CI [63.1, 81.0]) for dementia risk showed no significant difference. Without consideration of the Aβ42/Aβ40 ratio in ES grouping, the optimal cut-off of the ES shifted to ≥2. CONCLUSIONS The ERS showed advantages over the ES in test interpretation with comparable overall test performance, as fewer cases were allocated to the intermediate risk group. The established cut-off of ≥2 can be maintained for the ERS, whereas it must be adjusted for the ES when determining the Aβ42/Aβ40 ratio.
Collapse
Affiliation(s)
- Janine Utz
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Pauline Olm
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Johannes Jablonowski
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Eva-Maria Siegmann
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Philipp Spitzer
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, and Department of Biochemical Diagnostics, University Hospital of Bialystok, Białystok, Poland
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Juan Manuel Maler
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Timo Jan Oberstein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| |
Collapse
|
29
|
Hanon O, Vidal JS, Lehmann S, Bombois S, Allinquant B, Baret-Rose C, Tréluyer JM, Abdoul H, Gelé P, Delmaire C, Blanc F, Mangin JF, Buée L, Touchon J, Hugon J, Vellas B, Galbrun E, Benetos A, Berrut G, Paillaud E, Wallon D, Castelnovo G, Volpe-Gillot L, Paccalin M, Robert P, Godefroy O, Camus V, Belmin J, Vandel P, Novella JL, Duron E, Rigaud AS, Schraen-Maschke S, Gabelle A. Plasma amyloid beta predicts conversion to dementia in subjects with mild cognitive impairment: The BALTAZAR study. Alzheimers Dement 2022; 18:2537-2550. [PMID: 35187794 DOI: 10.1002/alz.12613] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/20/2021] [Accepted: 12/10/2021] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Blood-based biomarkers are the next challenge for Alzheimer's disease (AD) diagnosis and prognosis. METHODS Mild cognitive impairment (MCI) participants (N = 485) of the BALTAZAR study, a large-scale longitudinal multicenter cohort, were followed-up for 3 years. A total of 165 of them converted to dementia (95% AD). Associations of conversion and plasma amyloid beta (Aβ)1-42 , Aβ1-40 , Aβ1-42 /Aβ1-40 ratio were analyzed with logistic and Cox models. RESULTS Converters to dementia had lower level of plasma Aβ1-42 (37.1 pg/mL [12.5] vs. 39.2 [11.1] , P value = .03) and lower Aβ1-42 /Aβ1-40 ratio than non-converters (0.148 [0.125] vs. 0.154 [0.076], P value = .02). MCI participants in the highest quartile of Aβ1-42 /Aβ1-40 ratio (>0.169) had a significant lower risk of conversion (hazard ratio adjusted for age, sex, education, apolipoprotein E ε4, hippocampus atrophy = 0.52 (95% confidence interval [0.31-0.86], P value = .01). DISCUSSION In this large cohort of MCI subjects we identified a threshold for plasma Aβ1-42 /Aβ1-40 ratio that may detect patients with a low risk of conversion to dementia within 3 years.
Collapse
Affiliation(s)
- Olivier Hanon
- Memory Resource and Research Centre of de Paris-Broca-Ile de France, Université de Paris, EA 4468, APHP, Hopital Broca, Paris, France
| | - Jean-Sébastien Vidal
- Memory Resource and Research Centre of de Paris-Broca-Ile de France, Université de Paris, EA 4468, APHP, Hopital Broca, Paris, France
| | - Sylvain Lehmann
- CHU Montpellier, LBPC, Inserm, Université de Montpellier, Montpellier, France
| | - Stéphanie Bombois
- CHU Lille, U1172-LilNCog, LiCEND, LabEx DISTALZ, Université de Lille, Inserm, Lille, France
| | - Bernadette Allinquant
- UMR-S 1266, Université de Paris, Institute of Psychiatric and Neurosciences, Inserm, Paris, France
| | - Christiane Baret-Rose
- UMR-S 1266, Université de Paris, Institute of Psychiatric and Neurosciences, Inserm, Paris, France
| | - Jean-Marc Tréluyer
- Clinical Research Unit, Université de Paris, APHP, Hôpital Necker, Paris, France
| | - Hendy Abdoul
- Clinical Research Unit, Université de Paris, APHP, Hôpital Necker, Paris, France
| | - Patrick Gelé
- CHU Lille, CRB/CIC1403, Université de Lille, Inserm, Lille, France
| | - Christine Delmaire
- CHU Lille, U1172-LilNCog, LiCEND, LabEx DISTALZ, Université de Lille, Inserm, Lille, France
| | - Fredéric Blanc
- CM2R, pôle de Gériatrie, Laboratoire ICube, FMTS, CNRS, équipe IMIS, Université de Strasbourg, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Jean-François Mangin
- Neurospin, CEA, CNRS, cati-neuroimaging.com, CATI Multicenter Neuroimaging Platform, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Luc Buée
- CHU Lille, U1172-LilNCog, LiCEND, LabEx DISTALZ, Université de Lille, Inserm, Lille, France
| | - Jacques Touchon
- Department of Neurology, Memory Research and Resources Center of Montpellier, Inserm INM NeuroPEPs Team, Excellence Center of Neurodegenerative Disorders, Université de Montpellier, CHU Montpellier, Montpellier, France
| | - Jacques Hugon
- APHP, Groupe Hospitalier Saint Louis-Lariboisière Fernand Widal, Center of Cognitive Neurology, Université de Paris, Paris, France
| | - Bruno Vellas
- Memory Resource and Research Centre of Midi-Pyrénées, Université de Toulouse III, CHU La Grave-Casselardit, Toulouse, France
| | - Evelyne Galbrun
- Department of Gérontology 2, Sorbonne Université, APHP, Centre Hospitalier Dupuytren, Draveil, France
| | - Athanase Benetos
- Memory Resource and Research Centre of Lorraine, Université de Lorraine, CHRU de Nancy, Vandoeuvre-lès-Nancy, France
| | - Gilles Berrut
- Department of Clinical Gerontology, Memory Research Resource Center of Nantes, Université de Nantes, EA 4334 Movement-Interactions-Performance, CHU Nantes, Nantes, France
| | - Elena Paillaud
- Service de Gériatrie, Université de Paris, APHP, Hôpital Europeen Georges Pompidou, Paris, France
| | - David Wallon
- CHU de Rouen, Department of Neurology and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, CIC-CRB1404, Normandie Univ, UNIROUEN, Inserm U1245, Rouen, France
| | | | - Lisette Volpe-Gillot
- Service de Neuro-Psycho-Gériatrie, Memory Clinic, Hôpital Léopold Bellan, Paris, France
| | - Marc Paccalin
- Memory Resource and Research Centre of Poitiers, CHU de Poitiers, Poitiers, France
| | - Philippe Robert
- Memory Research Resource Center of Nice, CoBTek lab, Université Côte d'Azur, CHU de Nice, Nice, France
| | - Olivier Godefroy
- Memory Resource and Research Centre of Amiens Picardie, CHU d'Amiens-Picardie, Amiens, France
| | - Vincent Camus
- CHRU de Tours, UMR Inserm U1253, Université François-Rabelais de Tours, Tours, France
| | - Joël Belmin
- Service de Gériatrie Ambulatoire, Sorbonne Université, APHP, Hôpitaux Universitaires Pitie-Salpêtrière-Charles Foix, Paris, France
| | - Pierre Vandel
- Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, CHU de Besançon, Memory Resource and Research Centre of Besançon Franche-Comté, Université Bourgogne Franche-Comté, Besançon, France
| | - Jean-Luc Novella
- Memory Resource and Research Centre of Champagne-Ardenne, Université de Reims Champagne-Ardenne, EA 3797, CHU de Reims, Reims, France
| | - Emmanuelle Duron
- Département de gériatrie, Équipe MOODS, Inserm 1178, Université Paris-Saclay, APHP, Hôpital Paul Brousse, Villejuif, France
| | - Anne-Sophie Rigaud
- Memory Resource and Research Centre of de Paris-Broca-Ile de France, Université de Paris, EA 4468, APHP, Hopital Broca, Paris, France
| | | | - Audrey Gabelle
- Department of Neurology, Memory Research and Resources Center of Montpellier, Inserm INM NeuroPEPs Team, Excellence Center of Neurodegenerative Disorders, Université de Montpellier, CHU Montpellier, Montpellier, France
| | | |
Collapse
|
30
|
Whelan R, Barbey FM, Cominetti MR, Gillan CM, Rosická AM. Developments in scalable strategies for detecting early markers of cognitive decline. Transl Psychiatry 2022; 12:473. [PMID: 36351888 PMCID: PMC9645320 DOI: 10.1038/s41398-022-02237-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 11/10/2022] Open
Abstract
Effective strategies for early detection of cognitive decline, if deployed on a large scale, would have individual and societal benefits. However, current detection methods are invasive or time-consuming and therefore not suitable for longitudinal monitoring of asymptomatic individuals. For example, biological markers of neuropathology associated with cognitive decline are typically collected via cerebral spinal fluid, cognitive functioning is evaluated from face-to-face assessments by experts and brain measures are obtained using expensive, non-portable equipment. Here, we describe scalable, repeatable, relatively non-invasive and comparatively inexpensive strategies for detecting the earliest markers of cognitive decline. These approaches are characterized by simple data collection protocols conducted in locations outside the laboratory: measurements are collected passively, by the participants themselves or by non-experts. The analysis of these data is, in contrast, often performed in a centralized location using sophisticated techniques. Recent developments allow neuropathology associated with potential cognitive decline to be accurately detected from peripheral blood samples. Advances in smartphone technology facilitate unobtrusive passive measurements of speech, fine motor movement and gait, that can be used to predict cognitive decline. Specific cognitive processes can be assayed using 'gamified' versions of standard laboratory cognitive tasks, which keep users engaged across multiple test sessions. High quality brain data can be regularly obtained, collected at-home by users themselves, using portable electroencephalography. Although these methods have great potential for addressing an important health challenge, there are barriers to be overcome. Technical obstacles include the need for standardization and interoperability across hardware and software. Societal challenges involve ensuring equity in access to new technologies, the cost of implementation and of any follow-up care, plus ethical issues.
Collapse
Affiliation(s)
- Robert Whelan
- School of Psychology, Trinity College Dublin, Dublin, Ireland.
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.
| | - Florentine M Barbey
- School of Psychology, Trinity College Dublin, Dublin, Ireland
- Cumulus Neuroscience Ltd, Dublin, Ireland
| | - Marcia R Cominetti
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
- Department of Gerontology, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Claire M Gillan
- School of Psychology, Trinity College Dublin, Dublin, Ireland
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Anna M Rosická
- School of Psychology, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
31
|
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
|
32
|
Delaby C, Teunissen CE, Blennow K, Alcolea D, Arisi I, Amar EB, Beaume A, Bedel A, Bellomo G, Bigot‐Corbel E, Bjerke M, Blanc‐Quintin M, Boada M, Bousiges O, Chapman MD, DeMarco ML, D'Onofrio M, Dumurgier J, Dufour‐Rainfray D, Engelborghs S, Esselmann H, Fogli A, Gabelle A, Galloni E, Gondolf C, Grandhomme F, Grau‐Rivera O, Hart M, Ikeuchi T, Jeromin A, Kasuga K, Keshavan A, Khalil M, Körtvelyessy P, Kulczynska‐Przybik A, Laplanche J, Lewczuk P, Li Q, Lleó A, Malaplate C, Marquié M, Masters CL, Mroczko B, Nogueira L, Orellana A, Otto M, Oudart J, Paquet C, Paoletti FP, Parnetti L, Perret‐Liaudet A, Peoc'h K, Poesen K, Puig‐Pijoan A, Quadrio I, Quillard‐Muraine M, Rucheton B, Schraen S, Schott JM, Shaw LM, Suárez‐Calvet M, Tsolaki M, Tumani H, Udeh‐Momoh CT, Vaudran L, Verbeek MM, Verde F, Vermunt L, Vogelgsang J, Wiltfang J, Zetterberg H, Lehmann S. Clinical reporting following the quantification of cerebrospinal fluid biomarkers in Alzheimer's disease: An international overview. Alzheimers Dement 2022; 18:1868-1879. [PMID: 34936194 PMCID: PMC9787404 DOI: 10.1002/alz.12545] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 10/11/2021] [Accepted: 10/25/2021] [Indexed: 01/28/2023]
Abstract
INTRODUCTION The current practice of quantifying cerebrospinal fluid (CSF) biomarkers as an aid in the diagnosis of Alzheimer's disease (AD) varies from center to center. For a same biochemical profile, interpretation and reporting of results may differ, which can lead to misunderstandings and raises questions about the commutability of tests. METHODS We obtained a description of (pre-)analytical protocols and sample reports from 40 centers worldwide. A consensus approach allowed us to propose harmonized comments corresponding to the different CSF biomarker profiles observed in patients. RESULTS The (pre-)analytical procedures were similar between centers. There was considerable heterogeneity in cutoff definitions and report comments. We therefore identified and selected by consensus the most accurate and informative comments regarding the interpretation of CSF biomarkers in the context of AD diagnosis. DISCUSSION This is the first time that harmonized reports are proposed across worldwide specialized laboratories involved in the biochemical diagnosis of AD.
Collapse
Affiliation(s)
- Constance Delaby
- LBPC‐PPCUniv MontpellierCHU MontpellierINSERMMontpellierFrance,Hospital de la Santa Creu i Sant Pau ‐ Biomedical Research Institute Sant Pau ‐ Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Charlotte E. Teunissen
- Neurochemistry LabDepartment of Clinical ChemistryAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamNetherlands
| | - Kaj Blennow
- Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| | - Daniel Alcolea
- Hospital de la Santa Creu i Sant Pau ‐ Biomedical Research Institute Sant Pau ‐ Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Ivan Arisi
- European Brain Research Institute (EBRI) “Rita Levi‐Montalcini”RomaItaly
| | - Elodie Bouaziz Amar
- Université de ParisCognitive Neurology CenterGHU APHP Nord Lariboisière Fernand‐Widal HospitalParisFrance
| | | | | | - Giovanni Bellomo
- Lab of Clinical NeurochemistrySection of NeurologyDept. of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | | | - Maria Bjerke
- Vrije Universiteit BrusselCenter for Neurosciences and Department of Clinical BiologyClinical Neurochemistry LaboratoryUniversitair Ziekenhuis BrusselBrusselsBelgium,Department of Biomedical Sciences, Institute Born‐BungeUniversity of AntwerpAntwerpBelgium
| | | | - Mercè Boada
- Research Center and Memory ClinicFundació ACEInstitut Català de Neurociències Aplicades and Universitat Internacional de Catalunya (UIC)BarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Olivier Bousiges
- Laboratoire de Biochimie et Biologie Moléculaire, et CNRSICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg)Team IMISHôpitaux Universitaires de StrasbourgStrasbourgFrance
| | - Miles D Chapman
- Department of NeuroimmunologyNational Hospital for Neurology and Neurosurgery, UCL Queen SquareLondonUK
| | - Mari L. DeMarco
- Department of Pathology and Laboratory MedicineSt. Paul's Hospital, Providence Health Care, Vancouver, Canada & Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverCanada
| | - Mara D'Onofrio
- European Brain Research Institute (EBRI) “Rita Levi‐Montalcini”RomaItaly
| | - Julien Dumurgier
- Université de ParisCognitive Neurology CenterGHU APHP Nord Lariboisière Fernand‐Widal HospitalParisFrance
| | | | - Sebastiaan Engelborghs
- Department of Biomedical Sciences, Institute Born‐BungeUniversity of AntwerpAntwerpBelgium,Vrije Universiteit BrusselUniversitair Ziekenhuis BrusselCenter for Neurosciences and Department of NeurologyBrusselsBelgium
| | - Hermann Esselmann
- Department of Psychiatry and PsychotherapyUniversity Medical Center Goettingen (UMGGoettingenGermany
| | - Anne Fogli
- CHU Clermont‐FerrandClermont‐FerrandFrance
| | - Audrey Gabelle
- LBPC‐PPCUniv MontpellierCHU MontpellierINSERMMontpellierFrance
| | | | | | | | - Oriol Grau‐Rivera
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain,Servei de NeurologiaHospital del MarUnitat de deteriorament cognitiu i transtorns del movimentBarcelonaSpain,IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Melanie Hart
- Department of NeuroimmunologyNational Hospital for Neurology and Neurosurgery, UCL Queen SquareLondonUK
| | - Takeshi Ikeuchi
- Dept. of Molecular GeneticsCenter for BioresourcesBrain Research InstituteNiigata UniversityNiigataJapan
| | | | - Kensaku Kasuga
- Dept. of Molecular GeneticsCenter for BioresourcesBrain Research InstituteNiigata UniversityNiigataJapan
| | - Ashvini Keshavan
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | | | - Peter Körtvelyessy
- Freie Universität Berlin and Humboldt‐Universität zu BerlinDepartment of NeurologyGerman Center for Neurodegenerative Diseases, Magdeburg, Germany and Charité‐Universitäts medizin BerlinBerlinGermany
| | | | - Jean‐Louis Laplanche
- Université de ParisCognitive Neurology CenterGHU APHP Nord Lariboisière Fernand‐Widal HospitalParisFrance
| | - Piotr Lewczuk
- Department of Neurodegeneration DiagnosticsMedical University of BialystokBialystokPoland,Lab for Clinical Neurochemistry and Neurochemical Dementia DiagnosticsUniversitätsklinikum Erlangen and Friedrich‐Alexander Universität Erlangen‐NürnbergErlangenGermany
| | - Qiao‐Xin Li
- Florey Institute and The University of MelbourneMelbourneVictoriaAustralia
| | - Alberto Lleó
- Hospital de la Santa Creu i Sant Pau ‐ Biomedical Research Institute Sant Pau ‐ Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Catherine Malaplate
- CHRU de NancyLaboratoire de BiochimieBiologie Moléculaire et Nutrition/ Université de LorraineNancyFrance
| | - Marta Marquié
- Research Center and Memory ClinicFundació ACEInstitut Català de Neurociències Aplicades and Universitat Internacional de Catalunya (UIC)BarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Colin L. Masters
- Florey Institute and The University of MelbourneMelbourneVictoriaAustralia
| | - Barbara Mroczko
- Department of Neurodegeneration DiagnosticsMedical University of BialystokBialystokPoland
| | - Léonor Nogueira
- Laboratoire de Biologie Cellulaire et CytologieCHU PURPANToulouseFrance
| | - Adelina Orellana
- Research Center and Memory ClinicFundació ACEInstitut Català de Neurociències Aplicades and Universitat Internacional de Catalunya (UIC)BarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Markus Otto
- Department of Neurology and CSF LaboratoryUniversity of UlmUlmGermany
| | | | - Claire Paquet
- Université de ParisCognitive Neurology CenterGHU APHP Nord Lariboisière Fernand‐Widal HospitalParisFrance
| | - Federico Paolini Paoletti
- Lab of Clinical NeurochemistrySection of NeurologyDept. of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Lucilla Parnetti
- Lab of Clinical NeurochemistrySection of NeurologyDept. of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Armand Perret‐Liaudet
- Lyon Neuroscience Research Center BIORAN Team ‐ CNRS UMR 5292INSERM U1028Lyon University HospitalLyonFrance
| | - Katell Peoc'h
- Université de Paris GHU APHP Nord Beaujon HospitalParisFrance
| | - Koen Poesen
- Laboratory for Molecular Neurobiomarker Research (LaMoN)Department of NeurosciencesKU LeuvenLeuven Brain InstituteLeuvenBelgium
| | - Albert Puig‐Pijoan
- Servei de NeurologiaHospital del MarUnitat de deteriorament cognitiu i transtorns del movimentBarcelonaSpain,IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
| | - Isabelle Quadrio
- Lyon Neuroscience Research Center BIORAN Team ‐ CNRS UMR 5292INSERM U1028Lyon University HospitalLyonFrance
| | - Muriel Quillard‐Muraine
- UNIROUENRouen University HospitalDepartment of Clinical biologyBiochemistry laboratoryNormandie UnivRouenFrance
| | | | - Susanna Schraen
- InsermCHU LilleU1172‐LilNCogLICENDLabEx DISTALZUniversité de LilleLilleFrance
| | | | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine HospitalUniversity of PennsylvaniaPennsylvaniaUSA
| | - Marc Suárez‐Calvet
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain,Servei de NeurologiaHospital del MarUnitat de deteriorament cognitiu i transtorns del movimentBarcelonaSpain,IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Magda Tsolaki
- 1st Department of NeurologySchool of MedicineFaculty of Health of SciencesAristotle University of ThessalonikiThessalonikiGreece
| | - Hayrettin Tumani
- Department of Neurology and CSF LaboratoryUniversity of UlmUlmGermany
| | | | | | - Marcel M Verbeek
- Donders Institute for Brain, Cognition and BehaviourRadboud Alzheimer CentreDepartments of Neurology and Laboratory MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Federico Verde
- Department of Neurology ‐ Stroke Unit and Laboratory of NeuroscienceIRCCS Istituto Auxologico ItalianoMilanItaly,Department of Pathophysiology and Transplantation“Dino Ferrari” Center, Università degli Studi di MilanoMilanItaly
| | - Lisa Vermunt
- Neurochemistry LabDepartment of Clinical ChemistryAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamNetherlands
| | - Jonathan Vogelgsang
- Department of Psychiatry and PsychotherapyUniversity Medical Center Goettingen (UMGGoettingenGermany,McLean HospitalTranslational Neuroscience LaboratoryHarvard Medical SchoolBelmontMassachusettsUSA
| | - Jens Wiltfang
- Department of Psychiatry and PsychotherapyUniversity Medical Center Goettingen (UMGGoettingenGermany,German Center for Neurodegenerative Diseases (DZNE)GoettingenGermany,Neurosciences and Signaling GroupInstitute of Biomedicine (iBiMED)Department of Medical SciencesUniversity of AveiroAveiroPortugal
| | - Henrik Zetterberg
- Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden,Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden,UK Dementia Research Institute at UCLLondonUK,Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
| | - Sylvain Lehmann
- LBPC‐PPCUniv MontpellierCHU MontpellierINSERMMontpellierFrance
| |
Collapse
|
33
|
Xu C, Zhao L, Dong C. A Review of Application of Aβ42/40 Ratio in Diagnosis and Prognosis of Alzheimer’s Disease. J Alzheimers Dis 2022; 90:495-512. [DOI: 10.3233/jad-220673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The number of patients with Alzheimer’s disease (AD) and non-Alzheimer’s disease (non-AD) has drastically increased over recent decades. The amyloid cascade hypothesis attributes a vital role to amyloid-β protein (Aβ) in the pathogenesis of AD. As the main pathological hallmark of AD, amyloid plaques consist of merely the 42 and 40 amino acid variants of Aβ (Aβ 42 and Aβ 40). The cerebrospinal fluid (CSF) biomarker Aβ 42/40 has been extensively investigated and eventually integrated into important diagnostic tools to support the clinical diagnosis of AD. With the development of highly sensitive assays and technologies, blood-based Aβ 42/40, which was obtained using a minimally invasive and cost-effective method, has been proven to be abnormal in synchrony with CSF biomarker values. This paper presents the recent progress of the CSF Aβ 42/40 ratio and plasma Aβ 42/40 for AD as well as their potential clinical application as diagnostic markers or screening tools for dementia.
Collapse
Affiliation(s)
- Chang Xu
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Li Zhao
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Chunbo Dong
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
34
|
Zhou J, Benoit M, Sharoar MG. Recent advances in pre-clinical diagnosis of Alzheimer's disease. Metab Brain Dis 2022; 37:1703-1725. [PMID: 33900524 DOI: 10.1007/s11011-021-00733-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/05/2021] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is the most common dementia with currently no known cures or disease modifying treatments (DMTs), despite much time and effort from the field. Diagnosis and intervention of AD during the early pre-symptomatic phase of the disease is thought to be a more effective strategy. Therefore, the detection of biomarkers has emerged as a critical tool for monitoring the effect of new AD therapies, as well as identifying patients most likely to respond to treatment. The establishment of the amyloid/tau/neurodegeneration (A/T/N) framework in 2018 has codified the contexts of use of AD biomarkers in neuroimaging and bodily fluids for research and diagnostic purposes. Furthermore, a renewed drive for novel AD biomarkers and innovative methods of detection has emerged with the goals of adding additional insight to disease progression and discovery of new therapeutic targets. The use of biomarkers has accelerated the development of AD drugs and will bring new therapies to patients in need. This review highlights recent methods utilized to diagnose antemortem AD.
Collapse
Affiliation(s)
- John Zhou
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA
- Molecular Medicine Program, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Marc Benoit
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Md Golam Sharoar
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA.
| |
Collapse
|
35
|
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
|
36
|
Alzheimer’s Disease CSF Biomarker Profiles in Idiopathic Normal Pressure Hydrocephalus. J Pers Med 2022; 12:jpm12060935. [PMID: 35743720 PMCID: PMC9225293 DOI: 10.3390/jpm12060935] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/25/2022] [Accepted: 05/31/2022] [Indexed: 12/02/2022] Open
Abstract
Patients with idiopathic normal pressure hydrocephalus (iNPH) frequently show pathologic CSF Aβ42 levels, comparable with Alzheimer’s Disease (AD). Nevertheless, the clinical meaning of these findings has not been fully explained. We aimed to assess the role of AD CSF biomarkers (Aβ42, Aβ42/Aβ40, p-tau, t-tau) in iNPH. To this purpose, we enrolled 44 patients diagnosed with iNPH and 101 with AD. All the patients underwent CSF sampling. We compared CSF biomarker levels in iNPH and AD: Aβ42 levels were not different between iNPH and AD, while Aβ42/Aβ40, p-tau, and t-tau were significantly different and showed excellent accuracy in distinguishing iNPH and AD. A multiple logistic regression analysis showed that Aβ42/Aβ40 was the variable that most contributed to differentiating the two groups. Furthermore, iNPH patients with positive Aβ42/Aβ40 had higher p-tau and t-tau than iNPH patients with negative Aβ42/Aβ40. Those iNPH patients who showed cognitive impairment had lower Aβ42/Aβ40 and higher p-tau than patients without cognitive impairment. We concluded that positive CSF Aβ42 with negative Aβ42/Aβ40, p-tau, and t-tau is a typical CSF profile of iNPH. On the contrary, positive Aβ42/Aβ40 in iNPH patients, especially when associated with positive p-tau, may lead to suspicion of a coexistent AD pathology.
Collapse
|
37
|
Temviriyanukul P, Kittibunchakul S, Trisonthi P, Kunkeaw T, Inthachat W, Siriwan D, Suttisansanee U. Mangifera indica ‘Namdokmai’ Prevents Neuronal Cells from Amyloid Peptide Toxicity and Inhibits BACE-1 Activities in a Drosophila Model of Alzheimer’s Amyloidosis. Pharmaceuticals (Basel) 2022; 15:ph15050591. [PMID: 35631418 PMCID: PMC9146065 DOI: 10.3390/ph15050591] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurological illness with few effective treatments. Thus, ameliorating the effects of AD using natural products has attracted global attention with promising efficacy and safety. In this study, ten tropical fruits including Ananas comosus ‘Phulae’, Ananas comosus ‘Pattavia’, Carica papaya ‘Khaekdum’, Carica papaya ‘Khaeknuan’, Durio zibethinus ‘Monthong’, Durio zibethinus ‘Chanee’, Psidium guajava ‘Kimju’, Psidium guajava ‘Keenok’, Mangifera indica ‘Kaew’ and Mangifera indica ‘Namdokmai’ were screened for their inhibitory activities against the key enzymes, cholinesterases and β-secretase (BACE-1), involved in AD pathogenesis. The top three fruit extracts with promising in vitro anti-AD activities were further investigated using rat pheochromocytoma PC-12 neuronal cell line and Drosophila AD model. Data showed that M. indica ‘Kaew’, M. indica ‘Namdokmai’ and P. guajava ‘Kimju’ reduced Aβ1–42-mediated neurotoxicity by promoting glutathione-dependent enzymes, while M. indica ‘Namdokmai’ limited Aβ1–42 peptide formation via BACE-1 inhibition and amended locomotory behavior of the Drosophila AD model. Results indicated the potential anti-AD properties of tropical fruits, particularly M. indica ‘Namdokmai’ in the prevention of Aβ1–42-mediated neurotoxicity and as a BACE-1 blocker.
Collapse
Affiliation(s)
- Piya Temviriyanukul
- Food and Nutrition Academic and Research Cluster, Institute of Nutrition, Mahidol University, Salaya, Phuttamonthon, Nakhon Pathom 73170, Thailand; (P.T.); (S.K.); (T.K.); (W.I.)
| | - Suwapat Kittibunchakul
- Food and Nutrition Academic and Research Cluster, Institute of Nutrition, Mahidol University, Salaya, Phuttamonthon, Nakhon Pathom 73170, Thailand; (P.T.); (S.K.); (T.K.); (W.I.)
| | - Piyapat Trisonthi
- Institute of Food Research and Product Development, Kasetsart University, Bangkok 10900, Thailand;
| | - Thanit Kunkeaw
- Food and Nutrition Academic and Research Cluster, Institute of Nutrition, Mahidol University, Salaya, Phuttamonthon, Nakhon Pathom 73170, Thailand; (P.T.); (S.K.); (T.K.); (W.I.)
| | - Woorawee Inthachat
- Food and Nutrition Academic and Research Cluster, Institute of Nutrition, Mahidol University, Salaya, Phuttamonthon, Nakhon Pathom 73170, Thailand; (P.T.); (S.K.); (T.K.); (W.I.)
| | - Dalad Siriwan
- Institute of Food Research and Product Development, Kasetsart University, Bangkok 10900, Thailand;
- Correspondence: (D.S.); (U.S.)
| | - Uthaiwan Suttisansanee
- Food and Nutrition Academic and Research Cluster, Institute of Nutrition, Mahidol University, Salaya, Phuttamonthon, Nakhon Pathom 73170, Thailand; (P.T.); (S.K.); (T.K.); (W.I.)
- Correspondence: (D.S.); (U.S.)
| |
Collapse
|
38
|
Li DD, Zheng CQ, Zhang F, Shi JS. Potential neuroprotection by Dendrobium nobile Lindl alkaloid in Alzheimer's disease models. Neural Regen Res 2022; 17:972-977. [PMID: 34558510 PMCID: PMC8552836 DOI: 10.4103/1673-5374.324824] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 01/20/2021] [Accepted: 02/20/2021] [Indexed: 01/05/2023] Open
Abstract
At present, treatments for Alzheimer's disease can temporarily relieve symptoms but cannot prevent the decline of cognitive ability and other neurodegenerative changes. Dendrobium nobile Lindl alkaloid is the main active component of Dendrobium nobile Lindl. Dendrobium nobile Lindl alkaloid has been shown to resist aging, prolong life span, and exhibit immunomodulatory effects in animals. This review summarizes the mechanisms behind the neuroprotective effects reported in Alzheimer's disease animal models. The neuroprotective effects of Dendrobium nobile Lindl alkaloid have not been studied in patients. The mechanisms by which Dendrobium nobile Lindl alkaloid has been reported to improve cognitive dysfunction in Alzheimer's disease animal models may be associated with extracellular amyloid plaque production, regulation of tau protein hyperphosphorylation, inhibition of neuroinflammation and neuronal apoptosis, activation of autophagy, and enhanced synaptic connections.
Collapse
Affiliation(s)
- Dai-Di Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Lab of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chang-Qing Zheng
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Lab of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Lab of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Jing-Shan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Lab of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou Province, China
| |
Collapse
|
39
|
Palotai M, Schregel K, Nazari N, Merchant JP, Taylor WM, Guttmann CRG, Sinkus R, Young-Pearse TL, Patz S. Magnetic resonance elastography to study the effect of amyloid plaque accumulation in a mouse model. J Neuroimaging 2022; 32:617-628. [PMID: 35384128 DOI: 10.1111/jon.12996] [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: 01/18/2022] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Biomechanical changes in the brain have not been fully elucidated in Alzheimer's disease (AD). We aimed to investigate the effect of β-amyloid accumulation on mouse brain viscoelasticity. METHODS Magnetic resonance elastography was used to calculate magnitude of the viscoelastic modulus (|G*|), elasticity (Gd ), and viscosity (Gl ) in the whole brain parenchyma (WB) and bilateral hippocampi of 9 transgenic J20 (AD) mice (5 males/4 females) and 10 wild-type (WT) C57BL/6 mice (5 males/5 females) at 11 and 14 months of age. RESULTS Cross-sectional analyses showed no significant difference between AD and WT mice at either timepoints. No sex-specific differences were observed at 11 months of age, but AD females showed significantly higher hippocampal |G*| and Gl and WB |G*|, Gd , and Gl compared to both AD and WT males at 14 months of age. Similar trending differences were found between female AD and female WT animals but did not reach significance. Longitudinal analyses showed significant increases in hippocampal |G*|, Gd , and Gl , and significant decreases in WB |G*|, Gd , and Gl between 11 and 14 months in both AD and WT mice. Each subgroup showed significant increases in all hippocampal and significant decreases in all WB measures, with the exception of AD females, which showed no significant changes in WB |G*|, Gd , or Gl . CONCLUSION Aging had region-specific effects on cerebral viscoelasticity, namely, WB softening and hippocampal stiffening. Amyloid plaque deposition may have sex-specific effects, which require further scrutiny.
Collapse
Affiliation(s)
- Miklos Palotai
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Katharina Schregel
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Institute of Neuroradiology, University Medical Center Göttingen, Göttingen, Germany.,Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Navid Nazari
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| | - Julie P Merchant
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Walter M Taylor
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Charles R G Guttmann
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Ralph Sinkus
- School of Biomedical Imaging and Imaging Sciences, King's College London, London, UK.,INSERM U1148, Laboratory for Vascular Translational Science, University Paris Diderot, University Paris 13, Paris, France
| | - Tracy L Young-Pearse
- Harvard Medical School, Boston, Massachusetts, USA.,Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Samuel Patz
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
40
|
Manual and automated analysis of atrophy patterns in dementia with Lewy bodies on MRI. BMC Neurol 2022; 22:114. [PMID: 35331168 PMCID: PMC8943955 DOI: 10.1186/s12883-022-02642-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 03/14/2022] [Indexed: 11/10/2022] Open
Abstract
Background Dementia with Lewy bodies (DLB) is the second most common dementia type in patients older than 65 years. Its atrophy patterns remain unknown. Its similarities to Parkinson's disease and differences from Alzheimer's disease are subjects of current research. Methods The aim of our study was (i) to form a group of patients with DLB (and a control group) and create a 3D MRI data set (ii) to volumetrically analyze the entire brain in these groups, (iii) to evaluate visual and manual metric measurements of the innominate substance for real-time diagnosis, and (iv) to compare our groups and results with the latest literature. We identified 102 patients with diagnosed DLB in our psychiatric and neurophysiological archives. After exclusion, 63 patients with valid 3D data sets remained. We compared them with a control group of 25 patients of equal age and sex distribution. We evaluated the atrophy patterns in both (1) manually and (2) via Fast Surfers segmentation and volumetric calculations. Subgroup analyses were done of the CSF data and quality of 3D T1 data sets. Results Concordant with the literature, we detected moderate, symmetric atrophy of the hippocampus, entorhinal cortex and amygdala, as well as asymmetric atrophy of the right parahippocampal gyrus in DLB. The caudate nucleus was unaffected in patients with DLB, while all the other measured territories were slightly too moderately atrophied. The area under the curve analysis of the left hippocampus volume ratio (< 3646mm3) revealed optimal 76% sensitivity and 100% specificity (followed by the right hippocampus and left amygdala). The substantia innominata’s visual score attained a 51% optimal sensitivity and 84% specificity, and the measured distance 51% optimal sensitivity and 68% specificity in differentiating DLB from our control group. Conclusions In contrast to other studies, we observed a caudate nucleus sparing atrophy of the whole brain in patients with DLB. As the caudate nucleus is known to be the last survivor in dopamine-uptake, this could be the result of an overstimulation or compensation mechanism deserving further investigation. Its relative hypertrophy compared to all other brain regions could enable an imaging based identification of patients with DLB via automated segmentation and combined volumetric analysis of the hippocampus and amygdala. Supplementary Information The online version contains supplementary material available at 10.1186/s12883-022-02642-0.
Collapse
|
41
|
Delaby C, Alcolea D, Hirtz C, Vialaret J, Kindermans J, Morichon L, Fortea J, Belbin O, Gabelle A, Blennow K, Zetterberg H, Lleó A, Lehmann S. Blood amyloid and tau biomarkers as predictors of cerebrospinal fluid profiles. J Neural Transm (Vienna) 2022; 129:231-237. [PMID: 35169889 PMCID: PMC8866346 DOI: 10.1007/s00702-022-02474-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/05/2022] [Indexed: 11/25/2022]
Abstract
Introduction Blood biomarkers represent a major advance for improving the management, diagnosis, and monitoring of Alzheimer's disease (AD). However, their context of use in relation to routine cerebrospinal fluid (CSF) analysis for the quantification of amyloid peptides and tau proteins remains to be determined. Methods We studied in two independent cohorts, the performance of blood biomarkers in detecting “nonpathological” (A−/T−/N−), amyloid (A+) or neurodegenerative (T+ /N+) CSF profiles. Results Plasma Aβ1–42/Aβ1–40 ratio and phosphorylated tau (p-tau(181)) were independent and complementary predictors of the different CSF profile and in particular of the nonpathological (A−/T−/N−) profile with a sensitivity and specificity close to 85%. These performances and the corresponding biomarker thresholds were significantly different from those related to AD detection. Conclusion The use of blood biomarkers to identify patients who may benefit from secondary CSF testing represents an attractive stratification strategy in the clinical management of patients visiting memory clinics. This could reduce the need for lumbar puncture and foreshadow the use of blood testing on larger populations. Supplementary Information The online version contains supplementary material available at 10.1007/s00702-022-02474-9.
Collapse
Affiliation(s)
- Constance Delaby
- Laboratoire de Biochimie Protéomique Clinique (LBPC-PPC), Univ Montpellier, CHU Montpellier, INM INSERM, Hôpital St Eloi, IRMB 80 av A Fliche, 34295, Montpellier, France
- Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Daniel Alcolea
- Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Christophe Hirtz
- Laboratoire de Biochimie Protéomique Clinique (LBPC-PPC), Univ Montpellier, CHU Montpellier, INM INSERM, Hôpital St Eloi, IRMB 80 av A Fliche, 34295, Montpellier, France
| | - Jérôme Vialaret
- Laboratoire de Biochimie Protéomique Clinique (LBPC-PPC), Univ Montpellier, CHU Montpellier, INM INSERM, Hôpital St Eloi, IRMB 80 av A Fliche, 34295, Montpellier, France
| | - Jana Kindermans
- Laboratoire de Biochimie Protéomique Clinique (LBPC-PPC), Univ Montpellier, CHU Montpellier, INM INSERM, Hôpital St Eloi, IRMB 80 av A Fliche, 34295, Montpellier, France
| | - Lisa Morichon
- Laboratoire de Biochimie Protéomique Clinique (LBPC-PPC), Univ Montpellier, CHU Montpellier, INM INSERM, Hôpital St Eloi, IRMB 80 av A Fliche, 34295, Montpellier, France
| | - Juan Fortea
- Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Olivia Belbin
- Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Audrey Gabelle
- CMRR, Univ Montpellier, CHU Montpellier, INM INSERM, Montpellier, France
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the 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, Hong Kong, China
| | - Alberto Lleó
- Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sylvain Lehmann
- Laboratoire de Biochimie Protéomique Clinique (LBPC-PPC), Univ Montpellier, CHU Montpellier, INM INSERM, Hôpital St Eloi, IRMB 80 av A Fliche, 34295, Montpellier, France.
| |
Collapse
|
42
|
Delaby C, Estellés T, Zhu N, Arranz J, Barroeta I, Carmona-Iragui M, Illán-Gala I, Santos-Santos MÁ, Altuna M, Sala I, Sánchez-Saudinós MB, Videla L, Valldeneu S, Subirana A, Tondo M, Blanco-Vaca F, Lehmann S, Belbin O, Blesa R, Fortea J, Lleó A, Alcolea D. The Aβ1-42/Aβ1-40 ratio in CSF is more strongly associated to tau markers and clinical progression than Aβ1-42 alone. Alzheimers Res Ther 2022; 14:20. [PMID: 35105351 PMCID: PMC8809019 DOI: 10.1186/s13195-022-00967-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/20/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) Aβ1-42 levels and the Aβ1-42/Aβ1-40 ratio are markers of amyloid pathology, but previous studies suggest that their levels might be influenced by additional pathophysiological processes. AIMS To compare Aβ1-42 and the Aβ1-42/Aβ1-40 ratio in CSF in different neurodegenerative disorders and study their association with other biomarkers (tTau, pTau181, and NfL) and with cognitive and functional progression. METHODS We included all participants from the Sant Pau Initiative on Neurodegeneration (SPIN) with CSF Aβ1-42 and Aβ1-42/Aβ1-40. Participants had diagnoses of Alzheimer's disease (AD), dementia with Lewy bodies, frontotemporal lobar degeneration-related syndromes, non-neurodegenerative conditions, or were cognitively normal. We classified participants as "positive" or "negative" according to each marker. We compared CSF levels of tTau, pTau181, and NfL between concordant and discordant groups through ANCOVA and assessed differences in cognitive (MMSE, FCSRT) and functional (GDS, CDR-SOB) progression using Cox regression and linear-mixed models. RESULTS In the 1791 participants, the agreement between Aβ1-42 and Aβ1-42/Aβ1-40 was 78.3%. The Aβ1-42/Aβ1-40 ratio showed a stronger correlation with tTau and pTau181 than Aβ1-42 and an agreement with tTau and pTau181 of 73.1% and 77.1%, respectively. Participants with a low Aβ1-42/Aβ1-40 ratio showed higher tTau and pTau181 and worse cognitive and functional prognosis, regardless of whether they were positive or negative for Aβ1-42. The results were consistent across stages, diagnostic categories, and use of different cutoffs. CONCLUSION Although Aβ1-42 and Aβ1-42/Aβ1-40 are considered markers of the same pathophysiological pathway, our findings provide evidence favoring the use of the Aβ1-42/Aβ1-40 ratio in clinical laboratories in the context of AD.
Collapse
Affiliation(s)
- Constance Delaby
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- IRMB, INM, Université de Montpellier, INSERM, CHU de Montpellier, Laboratoire de Biochimie-Protéomique clinique, Montpellier, France
| | - Teresa Estellés
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Nuole Zhu
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Javier Arranz
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Isabel Barroeta
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - María Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Ignacio Illán-Gala
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Miguel Ángel Santos-Santos
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Miren Altuna
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Isabel Sala
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - M Belén Sánchez-Saudinós
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Laura Videla
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
- Fundació Catalana Síndrome de Down, Centre Mèdic Down, Barcelona, Spain
| | - Sílvia Valldeneu
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Andrea Subirana
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Mireia Tondo
- Servei de Bioquímica, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Servei de Bioquímica i Biologia Molecular, Unversitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas (CIBERDEM), Madrid, Spain
| | - Francisco Blanco-Vaca
- Servei de Bioquímica, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Servei de Bioquímica i Biologia Molecular, Unversitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas (CIBERDEM), Madrid, Spain
| | - Sylvain Lehmann
- IRMB, INM, Université de Montpellier, INSERM, CHU de Montpellier, Laboratoire de Biochimie-Protéomique clinique, Montpellier, France
| | - Olivia Belbin
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain.
| |
Collapse
|
43
|
Park S, Kim Y. Bias-generating factors in biofluid amyloid-β measurements for Alzheimer's disease diagnosis. Biomed Eng Lett 2021; 11:287-295. [PMID: 34616582 DOI: 10.1007/s13534-021-00201-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 01/03/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia worldwide, yet the dearth of readily accessible diagnostic biomarkers is a substantial hindrance towards progressing to effective preventive and therapeutic approaches. Due to a long delay between cerebral amyloid-β (Aβ) accumulation and the onset of cognitive impairments, biomarkers that reflect Aβ pathology and enable routine screening for disease progression are of urgent need for application in the clinical diagnosis of AD. According to accumulating evidences, cerebrospinal fluid (CSF) and plasma offer windows to the brain as they allow monitoring of biochemical changes in the brain. Considering the high availability and accuracy in depicting Aβ deposition in the brain, Aβ levels in CSF and plasma are regarded as promising fluid biomarkers for the diagnosis of AD patients at an early stage. However, clinical data with intra- and interindividual variations in the concentrations of CSF and plasma Aβ implicate the need to reevaluate current Aβ detection methods and establish a standardized operating procedure. Therefore, this review introduces three bias-generating factors in biofluid Aβ measurement that may hamper the accurate Aβ quantification and how such complications can be overcome for the widespread implementation of fluid Aβ detection in clinical practice.
Collapse
Affiliation(s)
- Sohui Park
- Department of Pharmacy, Department of Integrative Biotechnology and Translational Medicine, and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983 Republic of Korea
| | - YoungSoo Kim
- Department of Pharmacy, Department of Integrative Biotechnology and Translational Medicine, and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983 Republic of Korea
| |
Collapse
|
44
|
Lombardi G, Pupi A, Bessi V, Polito C, Padiglioni S, Ferrari C, Lucidi G, Berti V, De Cristofaro MT, Piaceri I, Bagnoli S, Nacmias B, Sorbi S. Challenges in Alzheimer's Disease Diagnostic Work-Up: Amyloid Biomarker Incongruences. J Alzheimers Dis 2021; 77:203-217. [PMID: 32716357 DOI: 10.3233/jad-200119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Discordance among amyloid biomarkers is a challenge to overcome in order to increase diagnostic accuracy in dementia. OBJECTIVES 1) To verify that cerebrospinal fluid (CSF) Aβ42/Aβ40 ratio (AβR) better agrees with Amyloid PET (Amy-PET) results compared to CSF Aβ42; 2) to detect differences among concordant positive, concordant negative, and discordant cases, basing the concordance definition on the agreement between CSF AβR and Amy-PET results; 3) to define the suspected underlying pathology of discordant cases using in vivo biomarkers. METHOD We retrospectively enrolled 39 cognitively impaired participants in which neuropsychological tests, apolipoprotein E genotype determination, TC/MRI, FDG-PET, Amy-PET, and CSF analysis had been performed. In all cases, CSF analysis was repeated using the automated Lumipulse method. In discordant cases, FDG-PET scans were evaluated visually and using automated classifiers. RESULTS CSF AβR better agreed with Amy-PET compared to CSF Aβ42 (Cohen's K 0.431 versus 0.05). Comparisons among groups did not show any difference in clinical characteristics except for age at symptoms onset that was higher in the 6 discordant cases with abnormal CSF AβR values and negative Amy-PET (CSF AβR+/AmyPET-). FDG-PET and all CSF markers (Aβ42, AβR, p-Tau, t-Tau) were suggestive of Alzheimer's disease (AD) in 5 of these 6 cases. CONCLUSION 1) CSF AβR is the CSF amyloid marker that shows the better level of agreement with Amy-PET results; 2) The use of FDG-PET and CSF-Tau markers in CSFAβR+/Amy-PET-discordant cases can support AD diagnosis; 3) Disagreement between positive CSF AβR and negative Amy-PET in symptomatic aged AD patients could be due to the variability in plaques conformation and a negative Amy-PET scan cannot be always sufficient to rule out AD.
Collapse
Affiliation(s)
- Gemma Lombardi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy.,Fondazione Filippo Turati, Pistoia, Italy
| | | | | | - Cristina Polito
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", Nuclear Medicine Unit, University of Florence, Florence, Italy
| | - Sonia Padiglioni
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Camilla Ferrari
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | | | - Valentina Berti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", Nuclear Medicine Unit, University of Florence, Florence, Italy
| | | | - Irene Piaceri
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy.,Fondazione IRCCS Don Carlo Gnocchi, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy.,Fondazione IRCCS Don Carlo Gnocchi, Florence, Italy
| |
Collapse
|
45
|
Supraja P, Tripathy S, Singh R, Singh V, Chaudhury G, Singh SG. Towards point-of-care diagnosis of Alzheimer's disease: Multi-analyte based portable chemiresistive platform for simultaneous detection of β-amyloid (1-40) and (1-42) in plasma. Biosens Bioelectron 2021; 186:113294. [PMID: 33971525 DOI: 10.1016/j.bios.2021.113294] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 04/11/2021] [Accepted: 04/28/2021] [Indexed: 01/05/2023]
Abstract
Label-free simultaneous detection of Alzheimer's disease (AD) specific biomarkers Aβ40 and Aβ42 peptides on a single platform using polypyrrole nanoparticle-based chemiresistive biosensors is reported here. The proposed interdigitated-microelectrode based inexpensive multisensor-platform can concurrently detect Aβ40 and Aβ42 in spiked-plasma in the range of 10-14 - 10-6 g/mL (with LoDs being 5.71 and 9.09 fg/mL, respectively), enabling the estimation of diagnostically significant Aβ42/Aβ40 ratio. A detailed study has been undertaken here to record the individual sensor responses against spiked-plasma samples with varying amounts and proportions of the two target peptides, towards enabling disease-progression monitoring using the Aβ-ratio. As compared to the existing cost-ineffective brain-imaging techniques such as PET and MRI, and the high-risk CSF based invasive AD biomarkers detecting procedures, the proposed approach offers a viable alternative for affordable point-of-care AD diagnostics, with possible usage in performance evaluation of therapeutic drugs. Towards point-of-care applications, the portable readout used in this work was conjugated with an android-based mobile app for data-acquisition and analysis.
Collapse
Affiliation(s)
- Patta Supraja
- Department of Electrical Engineering, Indian Institute of Technology Hyderabad, 502285, India.
| | - Suryasnata Tripathy
- Department of Electrical Engineering, Indian Institute of Technology Hyderabad, 502285, India.
| | - Ranjana Singh
- Department of Electrical Engineering, Indian Institute of Technology Hyderabad, 502285, India.
| | - Vikrant Singh
- School of Medicine, University of California Davis, USA.
| | - Gajendranath Chaudhury
- Department of Electrical Engineering, Indian Institute of Technology Hyderabad, 502285, India.
| | - Shiv Govind Singh
- Department of Electrical Engineering, Indian Institute of Technology Hyderabad, 502285, India.
| |
Collapse
|
46
|
Peng Q, Zhang Z. The fluid biomarkers of Alzheimer’s disease. BRAIN SCIENCE ADVANCES 2021. [DOI: 10.26599/bsa.2021.9050001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder. However, it still has no available disease‐modifying therapies. Its pathology cascade begins decades before symptomatic presentation. For these reasons, highly sensitive and highly specific fluid biomarkers should be developed for the early diagnosis of AD. In this study, the well‐established and emerging fluid biomarkers of AD are summarized, and recent advances on their role in early diagnosis and progression monitoring as well as their correlations with AD pathology are highlighted. Future prospects and related research directions are also discussed.
Collapse
Affiliation(s)
- Qinyu Peng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
47
|
Oyarzún MP, Tapia-Arellano A, Cabrera P, Jara-Guajardo P, Kogan MJ. Plasmonic Nanoparticles as Optical Sensing Probes for the Detection of Alzheimer's Disease. SENSORS (BASEL, SWITZERLAND) 2021; 21:2067. [PMID: 33809416 PMCID: PMC7998661 DOI: 10.3390/s21062067] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD), considered a common type of dementia, is mainly characterized by a progressive loss of memory and cognitive functions. Although its cause is multifactorial, it has been associated with the accumulation of toxic aggregates of the amyloid-β peptide (Aβ) and neurofibrillary tangles (NFTs) of tau protein. At present, the development of highly sensitive, high cost-effective, and non-invasive diagnostic tools for AD remains a challenge. In the last decades, nanomaterials have emerged as an interesting and useful tool in nanomedicine for diagnostics and therapy. In particular, plasmonic nanoparticles are well-known to display unique optical properties derived from their localized surface plasmon resonance (LSPR), allowing their use as transducers in various sensing configurations and enhancing detection sensitivity. Herein, this review focuses on current advances in in vitro sensing techniques such as Surface-enhanced Raman scattering (SERS), Surface-enhanced fluorescence (SEF), colorimetric, and LSPR using plasmonic nanoparticles for improving the sensitivity in the detection of main biomarkers related to AD in body fluids. Additionally, we refer to the use of plasmonic nanoparticles for in vivo imaging studies in AD.
Collapse
Affiliation(s)
- María Paz Oyarzún
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| | - Andreas Tapia-Arellano
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| | - Pablo Cabrera
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| | - Pedro Jara-Guajardo
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| | - Marcelo J. Kogan
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| |
Collapse
|
48
|
Phan LMT, Hoang TX, Vo TAT, Pham HL, Le HTN, Chinnadayyala SR, Kim JY, Lee SM, Cho WW, Kim YH, Choi SH, Cho S. Nanomaterial-based Optical and Electrochemical Biosensors for Amyloid beta and Tau: Potential for early diagnosis of Alzheimer's Disease. Expert Rev Mol Diagn 2021; 21:175-193. [PMID: 33560154 DOI: 10.1080/14737159.2021.1887732] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD), a heterogeneous pathological process representing the most common causes of dementia worldwide, has required early and accurate diagnostic tools. Neuropathological hallmarks of AD involve the aberrant accumulation of Amyloid beta (Aβ) into Amyloid plaques and hyperphosphorylated Tau into neurofibrillary tangles, occurring long before the onset of brain dysfunction.Areas covered:Considering the significance of Aβ and Tau in AD pathogenesis, these proteins have been adopted as core biomarkers of AD, and their quantification has provided precise diagnostic information to develop next-generation AD therapeutic approaches. However, conventional diagnostic methods may not suffice to achieve clinical criteria that are acceptable for proper diagnosis and treatment. The advantages of nanomaterial-based biosensors including facile miniaturization, mass fabrication, ultra-sensitivity, make them useful to be promising tools to measure Aβ and Tau simultaneously for accurate validation of low-abundance yet potentially informative biomarkers of AD.. EXPERT OPINION The study has identified the potential application of advanced biosensors as standardized clinical diagnostic tools for AD, evolving the way for new and efficient AD control with minimum economic and social burden. After clinical trial, nanobiosensors for measuring Aβ and Tau simultaneously possess innovative diagnosis of AD to provide significant contributions to primary Alzheimer's care intervention.
Collapse
Affiliation(s)
- Le Minh Tu Phan
- Department of Electronic Engineering, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea.,School of Medicine and Pharmacy, The University of Danang, Danang, Vietnam
| | - Thi Xoan Hoang
- Department of Life Science, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Thuy Anh Thu Vo
- Department of Life Science, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hoang Lan Pham
- Department of Life Science, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hien T Ngoc Le
- Department of Electronic Engineering, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | | | - Jae Young Kim
- Department of Life Science, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | | | - Won Woo Cho
- Cantis Inc., Ansan-si, Gyeonggi-do, Republic of Korea
| | - Young Hyo Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medicine, Inha University, Incheon, Republic of Korea
| | - Seong Hye Choi
- Department of Neurology, School of Medicine, Inha University, Incheon, Republic of Korea
| | - Sungbo Cho
- Department of Electronic Engineering, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea.,Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| |
Collapse
|
49
|
McGrowder DA, Miller F, Vaz K, Nwokocha C, Wilson-Clarke C, Anderson-Cross M, Brown J, Anderson-Jackson L, Williams L, Latore L, Thompson R, Alexander-Lindo R. Cerebrospinal Fluid Biomarkers of Alzheimer's Disease: Current Evidence and Future Perspectives. Brain Sci 2021; 11:215. [PMID: 33578866 PMCID: PMC7916561 DOI: 10.3390/brainsci11020215] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease is a progressive, clinically heterogeneous, and particularly complex neurodegenerative disease characterized by a decline in cognition. Over the last two decades, there has been significant growth in the investigation of cerebrospinal fluid (CSF) biomarkers for Alzheimer's disease. This review presents current evidence from many clinical neurochemical studies, with findings that attest to the efficacy of existing core CSF biomarkers such as total tau, phosphorylated tau, and amyloid-β (Aβ42), which diagnose Alzheimer's disease in the early and dementia stages of the disorder. The heterogeneity of the pathophysiology of the late-onset disease warrants the growth of the Alzheimer's disease CSF biomarker toolbox; more biomarkers showing other aspects of the disease mechanism are needed. This review focuses on new biomarkers that track Alzheimer's disease pathology, such as those that assess neuronal injury (VILIP-1 and neurofilament light), neuroinflammation (sTREM2, YKL-40, osteopontin, GFAP, progranulin, and MCP-1), synaptic dysfunction (SNAP-25 and GAP-43), vascular dysregulation (hFABP), as well as CSF α-synuclein levels and TDP-43 pathology. Some of these biomarkers are promising candidates as they are specific and predict future rates of cognitive decline. Findings from the combinations of subclasses of new Alzheimer's disease biomarkers that improve their diagnostic efficacy in detecting associated pathological changes are also presented.
Collapse
Affiliation(s)
- Donovan A. McGrowder
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Fabian Miller
- Department of Physical Education, Faculty of Education, The Mico University College, 1A Marescaux Road, Kingston 5, Jamaica;
- Department of Biotechnology, Faculty of Science and Technology, The University of the West Indies, Kingston 7, Jamaica;
| | - Kurt Vaz
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Chukwuemeka Nwokocha
- Department of Basic Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (C.N.); (C.W.-C.); (R.A.-L.)
| | - Cameil Wilson-Clarke
- Department of Basic Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (C.N.); (C.W.-C.); (R.A.-L.)
| | - Melisa Anderson-Cross
- School of Allied Health and Wellness, College of Health Sciences, University of Technology, Kingston 7, Jamaica;
| | - Jabari Brown
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Lennox Anderson-Jackson
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Lowen Williams
- Department of Biotechnology, Faculty of Science and Technology, The University of the West Indies, Kingston 7, Jamaica;
| | - Lyndon Latore
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Rory Thompson
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Ruby Alexander-Lindo
- Department of Basic Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (C.N.); (C.W.-C.); (R.A.-L.)
| |
Collapse
|
50
|
Guyon A, Rousseau J, Lamothe G, Tremblay JP. The protective mutation A673T in amyloid precursor protein gene decreases Aβ peptides production for 14 forms of Familial Alzheimer's Disease in SH-SY5Y cells. PLoS One 2020; 15:e0237122. [PMID: 33370284 PMCID: PMC7769289 DOI: 10.1371/journal.pone.0237122] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022] Open
Abstract
The deposition of Aβ plaques in the brain leads to the onset and development of Alzheimer’s disease. The Amyloid precursor protein (APP) is cleaved by α-secretase (non-amyloidogenic processing of APP), however increased cleavage by β-secretase (BACE1) leads to the accumulation of Aβ peptides, which forms plaques. APP mutations mapping to exons 16 and 17 favor plaque accumulation and cause Familial Alzheimer Disease (FAD). However, a variant of the APP gene (A673T) originally found in an Icelandic population reduces BACE1 cleavage by 40%. A series of plasmids containing the APP gene, each with one of 29 different FAD mutations mapping to exon 16 and exon 17 was created. These plasmids were then replicated with the addition of the A673T mutation. Combined these formed the library of plasmids that was used in this study. The plasmids were transfected in neuroblastomas to assess the effect of this mutation on Aβ peptide production. The production of Aβ peptides was decreased for some FAD mutations due to the presence of the co-dominant A673T mutation. The reduction of Aβ peptide concentrations for the London mutation (V717I) even reached the same level as for A673T control in SH-SY5Y cells. These preliminary results suggest that the insertion of A673T in APP genes containing FAD mutations might confer a clinical benefit in preventing or delaying the onset of some FADs.
Collapse
Affiliation(s)
- Antoine Guyon
- Centre de Recherche du CHU, Québec-Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire, l’Université Laval Québec, Québec, Québec, Canada
- * E-mail:
| | - Joël Rousseau
- Centre de Recherche du CHU, Québec-Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire, l’Université Laval Québec, Québec, Québec, Canada
| | - Gabriel Lamothe
- Centre de Recherche du CHU, Québec-Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire, l’Université Laval Québec, Québec, Québec, Canada
| | - Jacques P. Tremblay
- Centre de Recherche du CHU, Québec-Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire, l’Université Laval Québec, Québec, Québec, Canada
| |
Collapse
|