1
|
Soni AG, Verma A, Joshi R, Shah K, Soni D, Kaur CD, Saraf S, Chauhan NS. Phytoactive drugs used in the treatment of Alzheimer's disease and dementia. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8633-8649. [PMID: 38940847 DOI: 10.1007/s00210-024-03243-z] [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: 01/22/2024] [Accepted: 06/14/2024] [Indexed: 06/29/2024]
Abstract
The prevalence of Alzheimer's disease and other forms of dementia is increasing worldwide, and finding effective treatments for these conditions is a major public health challenge. Natural bioactive drugs have been identified as a promising source of potential treatments, due to their ability to target multiple pathways and their low toxicity. This paper reviews the current state of research on natural bioactive drugs used in the treatment of Alzheimer's disease and other dementias. The paper summarizes the findings of studies on various natural compounds, including curcumin, resveratrol, caffeine, genistein, quercetin, GinkoBiloba, Withaniasomnifera, Ginseng Brahmi, Giloy, and huperzine, and their effects on cognitive function, neuroinflammation, and amyloid-beta accumulation. In this review, we discuss the mechanism of action involved in the treatment of Alzheimer's disease. The paper also discusses the challenges associated with developing natural bioactive drugs for dementia treatment, including issues related to bioavailability and standardization. Finally, the paper suggests directions for future research in this area, including the need for more rigorous clinical trials and the development of novel delivery systems to improve the efficacy of natural bioactive drugs. Overall, this review highlights the potential of natural bioactive drugs as a promising avenue for the development of safe and effective treatments for Alzheimer's disease and other dementias.
Collapse
Affiliation(s)
- Anshita Gupta Soni
- Rungta College of Pharmaceutical Sciences and Research, Raipur, Chhattisgarh, India
| | - Astha Verma
- ShriRawatpuraSarkar Institute of Pharmacy, Durg, Chhattisgarh, India
| | - Renjil Joshi
- Rungta College of Pharmaceutical Sciences and Research, Bhilai, Chhattisgarh, India
| | - Kamal Shah
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, (U.P.), India
| | - Deependra Soni
- Faculty of Pharmacy, MATS University Campus, Aarang, Raipur, Chhattisgarh, India
| | - Chanchal Deep Kaur
- Rungta College of Pharmaceutical Sciences and Research, Raipur, Chhattisgarh, India
| | - Swarnlata Saraf
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh, India
| | | |
Collapse
|
2
|
Lin X, Dong X, Sun Y. Dual-Carbon Dots Composite: A Multifunctional Photo-Propelled Nanomotor Against Alzheimer's β-Amyloid. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2407154. [PMID: 39392092 DOI: 10.1002/smll.202407154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/16/2024] [Indexed: 10/12/2024]
Abstract
The abnormal accumulation of β-amyloid protein (Aβ) is considered as the main pathological hallmark of Alzheimer's disease (AD). The design of potent multifunctional theranostic agents targeting Aβ is one of the effective strategies for AD prevention and treatment. Nanomotors as intelligent, advanced, and multifunctional nanoplatforms can perform many complex tasks, but their application in AD theranostics is rare. Herein, sub-10nm multifunctional dual-carbon dots composites (ERCD) with photo-propelled nanomotor behavior are fabricated by conjugating near-infrared (NIR) carbon dots (RCD) of thermogenic and photodynamic capability with the previously reported epigallocatechin gallate-derived carbonized polymer dots (ECD). ERCD-1 (ECD:RCD = 1:2.5) showed potent inhibitory capability similar to ECD in the absence of NIR light, and exhibited photooxygenation activity and nanomotor behavior powered by "self-thermophoretic force" under NIR irradiation, significantly enhancing the inhibition, disaggregation, and photooxygenation capabilities. The nanomotor suppressed Aβ fibrillization and rapidly disaggregated mature Aβ fibrils at very low concentrations (0.5 µg mL-1). Moreover, the NIR-activated ERCD-1 imaged Aβ plaques in vivo and prolonged nematode lifespan by 6 d at 2 µg mL-1. As a proof-of-concept, this work opened a new avenue to the design of multifunctional sub-10nm nanomotors targeting Aβ for AD theranostics.
Collapse
Affiliation(s)
- Xiaoding Lin
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, China
| | - Xiaoyan Dong
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, China
| | - Yan Sun
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, China
| |
Collapse
|
3
|
Zhao K, Wang D, Wang D, Chen P, Wei Y, Tu L, Chen Y, Tang Y, Yao H, Zhou B, Lu J, Wang P, Liao Z, Chen Y, Han Y, Zhang X, Liu Y. Macroscale connectome topographical structure reveals the biomechanisms of brain dysfunction in Alzheimer's disease. SCIENCE ADVANCES 2024; 10:eado8837. [PMID: 39392880 DOI: 10.1126/sciadv.ado8837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 09/11/2024] [Indexed: 10/13/2024]
Abstract
The intricate spatial configurations of brain networks offer essential insights into understanding the specific patterns of brain abnormalities and the underlying biological mechanisms associated with Alzheimer's disease (AD), normal aging, and other neurodegenerative disorders. This study investigated alterations in the topographical structure of the brain related to aging and neurodegenerative diseases by analyzing brain gradients derived from structural MRI data across multiple cohorts (n = 7323). The analysis identified distinct gradient patterns in AD, aging, and other neurodegenerative conditions. Gene enrichment analysis indicated that inorganic ion transmembrane transport was the most significant term in normal aging, while chemical synaptic transmission is a common enrichment term across various neurodegenerative diseases. Moreover, the findings show that each disorder exhibits unique dysfunctional neurophysiological characteristics. These insights are pivotal for elucidating the distinct biological mechanisms underlying AD, thereby enhancing our understanding of its unique clinical phenotypes in contrast to normal aging and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Kun Zhao
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Dawei Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
- Research Institute of Shandong University: Magnetic Field-free Medicine & Functional Imaging, Jinan, China
- Shandong Key Laboratory: Magnetic Field-free Medicine & Functional Imaging (MF), Jinan, China
| | - Dong Wang
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Pindong Chen
- School of Artificial Intelligence, University of Chinese Academy of Sciences & Brainnetome Center, Chinese Academy of Sciences, Beijing, China
| | - Yongbin Wei
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Liyun Tu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Yuqi Chen
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Yi Tang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Hongxiang Yao
- Department of Radiology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Bo Zhou
- Department of Neurology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Jie Lu
- Department of Radiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Pan Wang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhengluan Liao
- Department of Psychiatry, People's Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Yan Chen
- Department of Psychiatry, People's Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Xi Zhang
- Department of Neurology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Yong Liu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences & Brainnetome Center, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
4
|
Diddi SL, Lohidasan S, S A, Dhapte-Pawar V, Mahadik KR. In-situ polyherbal gel as biomedicine in the management of Alzheimer's disease: Understanding ameliorative potential in Trimethyltin induced neurodegeneration. J Pharmacol Toxicol Methods 2024; 130:107567. [PMID: 39393715 DOI: 10.1016/j.vascn.2024.107567] [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: 06/08/2024] [Revised: 08/21/2024] [Accepted: 10/05/2024] [Indexed: 10/13/2024]
Abstract
Alzheimer's disease (AD), classified as neurodegenerative disorder that progresses over a period of time, is characterized by intracellular neurofibrillary tangles and extracellular amyloid plaques. This present research work was designed to develop a polyherbal gel for the treatment of AD. This research study is aimed to confirm the impact and validation of polyherbal gel on tauopathy and neurodegeneration that had been induced by intraperitoneal trimethyltin (TMT) injection to rats. Polyherbal loaded gel was prepared by cold method, and characterized for gel strength, viscosity, permeation and pH. Subsequently, 5 marker based standardized plant materials of Kalyanka ghrita were incorporated in gellan gum and xanthan gum. Finally, an in-vivo investigation employing rats with TMT-induced neurological disease were used to assess the efficacy of the optimized gel. On day 7, the Wistar rats received intraperitoneal injections of TMT. From day 14 to day 35, the corresponding groups received intranasal administration of polyherbal gel. In addition to the molecular parameters such as brain acetyl cholinesterase activity, BDNF (Rat brain derived neurotropic factor), protein phosphatase 2 A, antioxidant parameters, and oxidative stress markers, the behavioral parameters were also determined. Studies were conducted on the brain's monoamine levels and histology. RESULTS: Higher permeation over the nasal mucosa was demonstrated by the optimized In-situ polyherbal gel. Significant improvement in cognition was observed from the reduced escape latency, longer paths, and increased social or novel object recognition tests post polyherbal gel treatment. A documented HPLC technique helped in optimization and standardization of the polyherbal gel. The polyherbal treatment groups exhibited a considerable rise in the levels of monoamines, including norepinephrine, dopamine, and 5-hydroxy tryptamine. CONCLUSION: According to the current study, treating Alzheimer's disease (AD) with a polyherbal gel formulation may be a viable option for successful therapy.
Collapse
Affiliation(s)
- Sneha Latha Diddi
- Department of Pharmacology, Bharati Vidyapeeth (Deemed to be University), Poona College of Pharmacy, Pune 411038, India
| | - Sathiyanarayanan Lohidasan
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth (Deemed to be University), Poona College of Pharmacy, Pune 411038, India
| | - Arulmozhi S
- Department of Pharmacology, Bharati Vidyapeeth (Deemed to be University), Poona College of Pharmacy, Pune 411038, India.
| | - Vividha Dhapte-Pawar
- Department of Pharmaceutics, Bharati Vidyapeeth (Deemed to be University), Poona College of Pharmacy, Pune 411038, India
| | - Kakasaheb R Mahadik
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth (Deemed to be University), Poona College of Pharmacy, Pune 411038, India
| |
Collapse
|
5
|
Song Y, Kim H, Lee J, Kim K. Oxygen-enriching triphase platform for reliable sensing of femtomolar Alzheimer's neurofilament lights. Biosens Bioelectron 2024; 260:116431. [PMID: 38815462 DOI: 10.1016/j.bios.2024.116431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
Accurate quantification of neurofilament lights (NfLs), a prognostic blood biomarker, is highly required to predict neurodegeneration in the presymptomatic stages of Alzheimer's disease. Here, we report self-oxygen-enriching coral structures with triphase interfaces for the label-free photocathodic detection of NfLs in blood plasma with femtomolar sensitivities and high reliability. In conventional photocathodic immunoassays, the poor solubility and sluggish diffusion rate of the dissolved oxygen serving as electron acceptors have necessitated the incorporation of additional electron acceptors or aeration procedures. To address the challenge, we designed the coral-like copper bismuth oxides (CBO) with robust solid-liquid-air contact boundaries that enrich the interfacial oxygen levels without an external aeration source. By optimally assembling the perfluorododecyltrichlorosilane (FTCS) and platinum (Pt) co-catalysts into the silver-doped CBO (Ag:CBO), the stable solid-liquid-air contact boundaries were formed within the sensor interfaces, which allowed for the abundant supply of air phase oxygen through an air pocket connected to the atmosphere. The Pt/FTCS-Ag:CBO exhibited the stable background signals independent of the dissolved oxygen fluctuations and amplified photocurrent signals by 1.76-fold, which were attributed to the elevated interfacial oxygen levels and 11.15 times-lowered mass transport resistance. Under the illumination of white light-emitting diode, the oxygen-enriching photocathodic sensor composed of Pt/FTCS-Ag:CBO conjugated with NfLs-specific antibodies precisely quantified the NfLs in plasma with a low coefficient of variation (≤2.97%), a high degree of recovery (>97.0%), and a limit of detection of 40.38 fg/mL, which was 140 times lower than the typical photocathodic sensor with diphase interfaces.
Collapse
Affiliation(s)
- Yunji Song
- Department of Fiber Convergence Material Engineering, Dankook University, Gyeonggi-Do, 16890, Republic of Korea
| | - Hayeon Kim
- Department of Fiber Convergence Material Engineering, Dankook University, Gyeonggi-Do, 16890, Republic of Korea
| | - Joonseok Lee
- Department of Chemistry, Hanyang University, Seoul, 04763, Republic of Korea.
| | - Kayoung Kim
- Department of Fiber Convergence Material Engineering, Dankook University, Gyeonggi-Do, 16890, Republic of Korea.
| |
Collapse
|
6
|
Swann P, Mirza-Davies A, O'Brien J. Associations Between Neuropsychiatric Symptoms and Inflammation in Neurodegenerative Dementia: A Systematic Review. J Inflamm Res 2024; 17:6113-6141. [PMID: 39262651 PMCID: PMC11389708 DOI: 10.2147/jir.s385825] [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: 04/15/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
Background Neuropsychiatric symptoms are common in dementia and linked to adverse outcomes. Inflammation is increasingly recognized as playing a role as a driver of early disease progression in Alzheimer's disease (AD) and related dementias. Inflammation has also been linked to primary psychiatric disorders, however its association with neuropsychiatric symptoms in neurodegenerative dementias remains uncertain. Methods We conducted a systematic literature review investigating associations between inflammation and neuropsychiatric symptoms in neurodegenerative dementias, including AD, Lewy body, Frontotemporal, Parkinson's (PD) and Huntington's disease dementias. Results Ninety-nine studies met our inclusion criteria, and the majority (n = 59) investigated AD and/or mild cognitive impairment (MCI). Thirty-five studies included PD, and only 6 investigated non-AD dementias. Inflammation was measured in blood, CSF, by genotype, brain tissue and PET imaging. Overall, studies exhibited considerable heterogeneity and evidence for specific inflammatory markers was inconsistent, with lack of replication and few longitudinal studies with repeat biomarkers. Depression was the most frequently investigated symptom. In AD, some studies reported increases in peripheral IL-6, TNF-a associated with depressive symptoms. Preliminary investigations using PET measures of microglial activation found an association with agitation. In PD, studies reported positive associations between TNF-a, IL-6, CRP, MCP-1, IL-10 and depression. Conclusion Central and peripheral inflammation may play a role in neuropsychiatric symptoms in neurodegenerative dementias; however, the evidence is inconsistent. There is a need for multi-site longitudinal studies with detailed assessments of neuropsychiatric symptoms combined with replicable peripheral and central markers of inflammation.
Collapse
Affiliation(s)
- Peter Swann
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - Anastasia Mirza-Davies
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - John O'Brien
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
7
|
Sokouti B. The identification of biomarkers for Alzheimer's disease using a systems biology approach based on lncRNA-circRNA-miRNA-mRNA ceRNA networks. Comput Biol Med 2024; 179:108860. [PMID: 38996555 DOI: 10.1016/j.compbiomed.2024.108860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/16/2024] [Accepted: 07/06/2024] [Indexed: 07/14/2024]
Abstract
In addition to being the most prevalent form of neurodegeneration among the elderly, AD is a devastating multifactorial disease. Currently, treatments address only its symptoms. Several clinical studies have shown that the disease begins to manifest decades before the first symptoms appear, indicating that studying early changes is crucial to improving early diagnosis and discovering novel treatments. Our study used bioinformatics and systems biology to identify biomarkers in AD that could be used for diagnosis and prognosis. The procedure was performed on data from the GEO database, and GO and KEGG enrichment analysis were performed. Then, we set up a network of interactions between proteins. Several miRNA prediction tools including miRDB, miRWalk, and TargetScan were used. The ceRNA network led to the identification of eight mRNAs, four circRNAs, seven miRNAs, and seven lncRNAs. Multiple mechanisms, including the cell cycle and DNA replication, have been linked to the promotion of AD development by the ceRNA network. By using the ceRNA network, it should be possible to extract prospective biomarkers and therapeutic targets for the treatment of AD. It is possible that the processes involved in DNA cell cycle and the replication of DNA contribute to the development of Alzheimer's disease.
Collapse
Affiliation(s)
- Babak Sokouti
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
8
|
Boudries R, Williams H, Paquereau-Gaboreau S, Bashir S, Hojjat Jodaylami M, Chisanga M, Trudeau LÉ, Masson JF. Surface-Enhanced Raman Scattering Nanosensing and Imaging in Neuroscience. ACS NANO 2024; 18:22620-22647. [PMID: 39088751 DOI: 10.1021/acsnano.4c05200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
Monitoring neurochemicals and imaging the molecular content of brain tissues in vitro, ex vivo, and in vivo is essential for enhancing our understanding of neurochemistry and the causes of brain disorders. This review explores the potential applications of surface-enhanced Raman scattering (SERS) nanosensors in neurosciences, where their adoption could lead to significant progress in the field. These applications encompass detecting neurotransmitters or brain disorders biomarkers in biofluids with SERS nanosensors, and imaging normal and pathological brain tissues with SERS labeling. Specific studies highlighting in vitro, ex vivo, and in vivo analysis of brain disorders using fit-for-purpose SERS nanosensors will be detailed, with an emphasis on the ability of SERS to detect clinically pertinent levels of neurochemicals. Recent advancements in designing SERS-active nanomaterials, improving experimentation in biofluids, and increasing the usage of machine learning for interpreting SERS spectra will also be discussed. Furthermore, we will address the tagging of tissues presenting pathologies with nanoparticles for SERS imaging, a burgeoning domain of neuroscience that has been demonstrated to be effective in guiding tumor removal during brain surgery. The review also explores future research applications for SERS nanosensors in neuroscience, including monitoring neurochemistry in vivo with greater penetration using surface-enhanced spatially offset Raman scattering (SESORS), near-infrared lasers, and 2-photon techniques. The article concludes by discussing the potential of SERS for investigating the effectiveness of therapies for brain disorders and for integrating conventional neurochemistry techniques with SERS sensing.
Collapse
Affiliation(s)
- Ryma Boudries
- Department of Chemistry, Institut Courtois, Quebec Center for Advanced Materials (QCAM), and Regroupement Québécois sur les Matériaux de Pointe (RQMP), Université de Montréal, C.P. 6128 Succ. Centre-Ville, Montréal, Quebec H3C 3J7, Canada
| | - Hannah Williams
- Department of Chemistry, Institut Courtois, Quebec Center for Advanced Materials (QCAM), and Regroupement Québécois sur les Matériaux de Pointe (RQMP), Université de Montréal, C.P. 6128 Succ. Centre-Ville, Montréal, Quebec H3C 3J7, Canada
| | - Soraya Paquereau-Gaboreau
- Department of Chemistry, Institut Courtois, Quebec Center for Advanced Materials (QCAM), and Regroupement Québécois sur les Matériaux de Pointe (RQMP), Université de Montréal, C.P. 6128 Succ. Centre-Ville, Montréal, Quebec H3C 3J7, Canada
- Department of Pharmacology and Physiology, Department of Neurosciences, Faculty of Medicine, Université de Montréal, C.P. 6128 Succ. Centre-ville, Montréal, Quebec H3C 3J7, Canada
- Neural Signalling and Circuitry Research Group (SNC), Center for Interdisciplinary Research on the Brain and Learning (CIRCA), Université de Montréal, C.P. 6128 Succ. Centre-ville, Montréal, Quebec H3C 3J7, Canada
| | - Saba Bashir
- Department of Chemistry, Institut Courtois, Quebec Center for Advanced Materials (QCAM), and Regroupement Québécois sur les Matériaux de Pointe (RQMP), Université de Montréal, C.P. 6128 Succ. Centre-Ville, Montréal, Quebec H3C 3J7, Canada
| | - Maryam Hojjat Jodaylami
- Department of Chemistry, Institut Courtois, Quebec Center for Advanced Materials (QCAM), and Regroupement Québécois sur les Matériaux de Pointe (RQMP), Université de Montréal, C.P. 6128 Succ. Centre-Ville, Montréal, Quebec H3C 3J7, Canada
| | - Malama Chisanga
- Department of Chemistry, Institut Courtois, Quebec Center for Advanced Materials (QCAM), and Regroupement Québécois sur les Matériaux de Pointe (RQMP), Université de Montréal, C.P. 6128 Succ. Centre-Ville, Montréal, Quebec H3C 3J7, Canada
| | - Louis-Éric Trudeau
- Department of Pharmacology and Physiology, Department of Neurosciences, Faculty of Medicine, Université de Montréal, C.P. 6128 Succ. Centre-ville, Montréal, Quebec H3C 3J7, Canada
- Neural Signalling and Circuitry Research Group (SNC), Center for Interdisciplinary Research on the Brain and Learning (CIRCA), Université de Montréal, C.P. 6128 Succ. Centre-ville, Montréal, Quebec H3C 3J7, Canada
| | - Jean-Francois Masson
- Department of Chemistry, Institut Courtois, Quebec Center for Advanced Materials (QCAM), and Regroupement Québécois sur les Matériaux de Pointe (RQMP), Université de Montréal, C.P. 6128 Succ. Centre-Ville, Montréal, Quebec H3C 3J7, Canada
- Neural Signalling and Circuitry Research Group (SNC), Center for Interdisciplinary Research on the Brain and Learning (CIRCA), Université de Montréal, C.P. 6128 Succ. Centre-ville, Montréal, Quebec H3C 3J7, Canada
| |
Collapse
|
9
|
Ma Z, Zhang Z, Lv X, Zhang H, Lu K, Su G, Huang B, Chen H. Dual sensitivity-enhanced microring resonance-based integrated microfluidic biosensor for Aβ 42 detection. Talanta 2024; 275:126111. [PMID: 38657362 DOI: 10.1016/j.talanta.2024.126111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 04/07/2024] [Accepted: 04/13/2024] [Indexed: 04/26/2024]
Abstract
Sensitive, accurate, and straightforward biosensors are pivotal in the battle against Alzheimer's disease, particularly in light of the escalating patient population. These biosensors enable early adjunctive diagnosis, thereby facilitating prompt intervention, alleviating socioeconomic burdens, and preserving individual well-being. In this study, we introduce the development of a highly sensitive add-drop dual-microring resonant microfluidic sensing chip boasting a sensitivity of 188.11 nm/RIU, marking a significant 20.7% enhancement over single microring systems. Leveraging ultra-thin Parylene C for streamlined antibody immobilization and non-destructive removal, this platform facilitates the precise quantification of the Alzheimer's disease biomarker Aβ42. Employing an immune sensing strategy that amplifies and captures antigen signals using Au-labeled antibodies, we achieve an exceptional limit of detection of 9.02 pg/mL. The designed microring-based microfluidic biosensor chip exhibits outstanding specificity and sensitivity for Aβ42 in serum samples, offering a promising avenue for the early adjunctive diagnosis of Alzheimer's disease.
Collapse
Affiliation(s)
- Zhengtai Ma
- Key Laboratory of Optoelectronic Materials and Devices, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, China; College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Zan Zhang
- School of Electronic and Control Engineering, Chang'an University, Xi'an, China
| | - Xiaoqing Lv
- Key Laboratory of Optoelectronic Materials and Devices, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, China; College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Huan Zhang
- Key Laboratory of Optoelectronics Technology, Ministry of Education, Beijing University of Technology, Beijing, China
| | - Kaiwei Lu
- Key Laboratory of Optoelectronic Materials and Devices, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, China; College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Guoshuai Su
- Suzhou Institute of Microelectronics and Optoelectronics Integration, Suzhou, China; Suzhou Jiwei Photoelectric Co., Ltd, Suzhou, China
| | - Beiju Huang
- Key Laboratory of Optoelectronic Materials and Devices, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, China; College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, China.
| | - Hongda Chen
- Key Laboratory of Optoelectronic Materials and Devices, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, China; College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
10
|
Krasny S, Yan C, Hartley SL, Handen BL, Wisch JK, Boehrwinkle AH, Ances BM, Rafii MS. Assessing amyloid PET positivity and cognitive function in Down syndrome to guide clinical trials targeting amyloid. Alzheimers Dement 2024; 20:5570-5577. [PMID: 38940611 PMCID: PMC11350128 DOI: 10.1002/alz.14068] [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: 03/01/2024] [Revised: 04/26/2024] [Accepted: 05/21/2024] [Indexed: 06/29/2024]
Abstract
INTRODUCTION Trisomy 21, or Down syndrome (DS), predisposes individuals to early-onset Alzheimer's disease (AD). While monoclonal antibodies (mAbs) targeting amyloid are approved for older AD patients, their efficacy in DS remains unexplored. This study examines amyloid positron emission tomography (PET) positivity (A+), memory function, and clinical status across ages in DS to guide mAb trial designs. METHODS Cross-sectional data from the Alzheimer Biomarker Consortium-Down Syndrome (ABC-DS) was analyzed. PET amyloid beta in Centiloids classified amyloid status using various cutoffs. Episodic memory was assessed using the modified Cued Recall Test, and clinical status was determined through consensus processes. RESULTS Four hundred nine DS adults (mean age = 44.83 years) were evaluated. A+ rates increased with age, with mean amyloid load rising significantly. Memory decline and cognitive impairment are also correlated with age. DISCUSSION These findings emphasize the necessity of tailoring mAb trials for DS, considering age-related AD characteristics. HIGHLIGHTS There is rapid increase in prevalence of amyloid beta (Aβ) positron emission tomography (PET) positivity in Down syndrome (DS) after the age of 40 years. Aβ PET positivity thresholds have significant impact on prevalence rates in DS. There is a significant lag between Aβ PET positivity and clinical symptom onset in DS.
Collapse
Affiliation(s)
| | - Cynthia Yan
- Department of NeurologyWashington UniversitySaint LouisMissouriUSA
| | | | - Ben L. Handen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Julie K. Wisch
- Department of NeurologyWashington UniversitySaint LouisMissouriUSA
| | | | - Beau M. Ances
- Department of NeurologyWashington UniversitySaint LouisMissouriUSA
| | - Michael S. Rafii
- Alzheimer's Therapeutic Research InstituteKeck School of Medicine of University of Southern CaliforniaSan DiegoCaliforniaUSA
| | | |
Collapse
|
11
|
Halder D, Das S, Joseph A. An insight into structure-activity relationship of naturally derived biological macromolecules for the treatment of Alzheimer's disease: a review. J Biomol Struct Dyn 2024; 42:6455-6471. [PMID: 37378526 DOI: 10.1080/07391102.2023.2230279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/21/2023] [Indexed: 06/29/2023]
Abstract
Alzheimer's disease (AD) is a neurological disorder that affects millions of people worldwide. There are currently no cures for AD, although various drugs are used to manage the symptoms and reduce the disease's progression. AChE inhibitors such as rivastigmine, donepezil, galantamine, and the NMDA glutamate receptor antagonist memantine are currently FDA-approved drugs used in the treatment of AD. Recently, naturally derived biological macromolecules have shown promising results in the treatment of AD. Several biological macromolecules derived from natural sources are in various stages of preclinical and clinical trials. During the literature search, it was observed that there is a lack of a comprehensive review that particularly focuses on the role of naturally derived biological macromolecules (protein, carbohydrates, lipids, and nucleic acids) in the treatment of AD and the structure-activity relationship (SAR) approach for understanding the medicinal chemistry perspective. This review focuses on the SAR and probable mechanisms of action of biological macromolecules derived from natural sources for the treatment of AD, including peptides, proteins, enzymes, and polysaccharides. The paper further addresses the therapeutic possibilities of monoclonal antibodies, enzymes, and vaccines for the treatment of AD. Overall, the review provides insight into the SAR of naturally derived biological macromolecules in the treatment of AD. The ongoing research in this field holds great promise for the future development of AD treatment and provides hope for individuals affected by this devastating disease.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Debojyoti Halder
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Subham Das
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Alex Joseph
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
12
|
Kim S, Wang SM, Kang DW, Um YH, Han EJ, Park SY, Ha S, Choe YS, Kim HW, Kim REY, Kim D, Lee CU, Lim HK. A Comparative Analysis of Two Automated Quantification Methods for Regional Cerebral Amyloid Retention: PET-Only and PET-and-MRI-Based Methods. Int J Mol Sci 2024; 25:7649. [PMID: 39062892 PMCID: PMC11276670 DOI: 10.3390/ijms25147649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/06/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Accurate quantification of amyloid positron emission tomography (PET) is essential for early detection of and intervention in Alzheimer's disease (AD) but there is still a lack of studies comparing the performance of various automated methods. This study compared the PET-only method and PET-and-MRI-based method with a pre-trained deep learning segmentation model. A large sample of 1180 participants in the Catholic Aging Brain Imaging (CABI) database was analyzed to calculate the regional standardized uptake value ratio (SUVR) using both methods. The logistic regression models were employed to assess the discriminability of amyloid-positive and negative groups through 10-fold cross-validation and area under the receiver operating characteristics (AUROC) metrics. The two methods showed a high correlation in calculating SUVRs but the PET-MRI method, incorporating MRI data for anatomical accuracy, demonstrated superior performance in predicting amyloid-positivity. The parietal, frontal, and cingulate importantly contributed to the prediction. The PET-MRI method with a pre-trained deep learning model approach provides an efficient and precise method for earlier diagnosis and intervention in the AD continuum.
Collapse
Affiliation(s)
- Sunghwan Kim
- Department of Psychiatry, College of Medicine, Yeouido St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Sheng-Min Wang
- Department of Psychiatry, College of Medicine, Yeouido St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Dong Woo Kang
- Department of Psychiatry, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Yoo Hyun Um
- Department of Psychiatry, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Eun Ji Han
- Division of Nuclear Medicine, Department of Radiology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Sonya Youngju Park
- Division of Nuclear Medicine, Department of Radiology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Yeong Sim Choe
- Research Institute, Neurophet Inc., Seoul 06234, Republic of Korea (R.E.K.)
| | - Hye Weon Kim
- Research Institute, Neurophet Inc., Seoul 06234, Republic of Korea (R.E.K.)
| | - Regina EY Kim
- Research Institute, Neurophet Inc., Seoul 06234, Republic of Korea (R.E.K.)
| | - Donghyeon Kim
- Research Institute, Neurophet Inc., Seoul 06234, Republic of Korea (R.E.K.)
| | - Chang Uk Lee
- Department of Psychiatry, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Hyun Kook Lim
- Department of Psychiatry, College of Medicine, Yeouido St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
- CMC Institute for Basic Medical Science, The Catholic Medical Center of The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| |
Collapse
|
13
|
Shou Q, Cen S, Chen NK, Ringman JM, Wen J, Kim H, Wang DJJ. Diffusion model enables quantitative CBF analysis of Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.01.24309791. [PMID: 39006427 PMCID: PMC11245071 DOI: 10.1101/2024.07.01.24309791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Objectives Cerebral blood flow (CBF) measured by arterial spin labeling (ASL) is a promising biomarker for Alzheimer's Disease (AD). ASL data from multiple vendors were included in the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset. However, the M0 images were missing in Siemens ASL data, prohibiting CBF quantification. Here, we utilized a generative diffusion model to impute the missing M0 and validated generated CBF data with acquired data from GE. Methods A conditional latent diffusion model was trained to generate the M0 image and validate it on an in-house dataset (N=55) based on image similarity metrics, accuracy of CBF quantification, and consistency with the physical model. This model was then applied to the ADNI dataset (Siemens: N=211) to impute the missing M0 for CBF calculation. We further compared the imputed data (Siemens) and acquired data (GE) regarding regional CBF differences by AD stages, their classification accuracy for AD prediction, and CBF trajectory slopes estimated by a mixed effect model. Results The trained diffusion model generated the M0 image with high fidelity (Structural similarity index, SSIM=0.924±0.019; peak signal-to-noise ratio, PSNR=33.348±1.831) and caused minimal bias in CBF values (mean difference in whole brain is 1.07±2.12ml/100g/min). Both generated and acquired CBF data showed similar differentiation patterns by AD stages, similar classification performance, and decreasing slopes with AD progression in specific AD-related regions. Generated CBF data also improved accuracy in classifying AD stages compared to qualitative perfusion data. Interpretation/Conclusion This study shows the potential of diffusion models for imputing missing modalities for large-scale studies of CBF variation with AD.
Collapse
Affiliation(s)
- Qinyang Shou
- Laboratory of Functional MRI Technology (LOFT), Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States
| | - Steven Cen
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Nan-kuei Chen
- Department of Biomedical Engineering, University of Arizona
| | - John M Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Junhao Wen
- Laboratory of AI & Biomedical Science (LABS), Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States
| | - Hosung Kim
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States
| | - Danny JJ Wang
- Laboratory of Functional MRI Technology (LOFT), Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States
| | | |
Collapse
|
14
|
Ritchie C, Blennow K, Gispert JD, Johnson S, van Maurik I, Vermunt L, Suárez-Calvet M, McHugh CP, Clement MHS, Anastasiu A, Rosenfeld E, Cosma O, Logan CA, Quevenco FC, Dias MC, Carboni M. NeuroToolKit Data Hackathon: advancing data collaboration in Alzheimer's disease. Front Neurosci 2024; 18:1339742. [PMID: 38994272 PMCID: PMC11238719 DOI: 10.3389/fnins.2024.1339742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 05/01/2024] [Indexed: 07/13/2024] Open
Affiliation(s)
- Craig Ritchie
- Scottish Brain Sciences, Edinburgh, United Kingdom
- Edinburgh Dementia Prevention and Centre for Clinical Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Sterling Johnson
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Ingrid van Maurik
- Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Neurology, Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
| | - Lisa Vermunt
- Neurology, Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
- Neurochemisty Laboratory, Clinical Chemistry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
- Servei de Neurología, Hospital del Mar, Barcelona, Spain
| | | | | | | | | | | | - Chad A. Logan
- Roche Diagnostics GmbH, PHCS Biostatistics & Data Management, Penzberg, Germany
| | | | | | | |
Collapse
|
15
|
Estarellas M, Oxtoby NP, Schott JM, Alexander DC, Young AL. Multimodal subtypes identified in Alzheimer's Disease Neuroimaging Initiative participants by missing-data-enabled subtype and stage inference. Brain Commun 2024; 6:fcae219. [PMID: 39035417 PMCID: PMC11259979 DOI: 10.1093/braincomms/fcae219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 03/14/2024] [Accepted: 06/22/2024] [Indexed: 07/23/2024] Open
Abstract
Alzheimer's disease is a highly heterogeneous disease in which different biomarkers are dynamic over different windows of the decades-long pathophysiological processes, and potentially have distinct involvement in different subgroups. Subtype and Stage Inference is an unsupervised learning algorithm that disentangles the phenotypic heterogeneity and temporal progression of disease biomarkers, providing disease insight and quantitative estimates of individual subtype and stage. However, a key limitation of Subtype and Stage Inference is that it requires a complete set of biomarkers for each subject, reducing the number of datapoints available for model fitting and limiting applications of Subtype and Stage Inference to modalities that are widely collected, e.g. volumetric biomarkers derived from structural MRI. In this study, we adapted the Subtype and Stage Inference algorithm to handle missing data, enabling the application of Subtype and Stage Inference to multimodal data (magnetic resonance imaging, positron emission tomography, cerebrospinal fluid and cognitive tests) from 789 participants in the Alzheimer's Disease Neuroimaging Initiative. Missing-data Subtype and Stage Inference identified five subtypes having distinct progression patterns, which we describe by the earliest unique abnormality as 'Typical AD with Early Tau', 'Typical AD with Late Tau', 'Cortical', 'Cognitive' and 'Subcortical'. These new multimodal subtypes were differentially associated with age, years of education, Apolipoprotein E (APOE4) status, white matter hyperintensity burden and the rate of conversion from mild cognitive impairment to Alzheimer's disease, with the 'Cognitive' subtype showing the fastest clinical progression, and the 'Subcortical' subtype the slowest. Overall, we demonstrate that missing-data Subtype and Stage Inference reveals a finer landscape of Alzheimer's disease subtypes, each of which are associated with different risk factors. Missing-data Subtype and Stage Inference has broad utility, enabling the prediction of progression in a much wider set of individuals, rather than being restricted to those with complete data.
Collapse
Affiliation(s)
- Mar Estarellas
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Neil P Oxtoby
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - Jonathan M Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Daniel C Alexander
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - Alexandra L Young
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| |
Collapse
|
16
|
Koppelmans V, Ruitenberg MF, Schaefer SY, King JB, Jacobo JM, Silvester BP, Mejia AF, van der Geest J, Hoffman JM, Tasdizen T, Duff K. Classification of Mild Cognitive Impairment and Alzheimer's Disease Using Manual Motor Measures. NEURODEGENER DIS 2024; 24:54-70. [PMID: 38865972 PMCID: PMC11381162 DOI: 10.1159/000539800] [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: 03/18/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024] Open
Abstract
INTRODUCTION Manual motor problems have been reported in mild cognitive impairment (MCI) and Alzheimer's disease (AD), but the specific aspects that are affected, their neuropathology, and potential value for classification modeling is unknown. The current study examined if multiple measures of motor strength, dexterity, and speed are affected in MCI and AD, related to AD biomarkers, and are able to classify MCI or AD. METHODS Fifty-three cognitively normal (CN), 33 amnestic MCI, and 28 AD subjects completed five manual motor measures: grip force, Trail Making Test A, spiral tracing, finger tapping, and a simulated feeding task. Analyses included (1) group differences in manual performance; (2) associations between manual function and AD biomarkers (PET amyloid β, hippocampal volume, and APOE ε4 alleles); and (3) group classification accuracy of manual motor function using machine learning. RESULTS Amnestic MCI and AD subjects exhibited slower psychomotor speed and AD subjects had weaker dominant hand grip strength than CN subjects. Performance on these measures was related to amyloid β deposition (both) and hippocampal volume (psychomotor speed only). Support vector classification well-discriminated control and AD subjects (area under the curve of 0.73 and 0.77, respectively) but poorly discriminated MCI from controls or AD. CONCLUSION Grip strength and spiral tracing appear preserved, while psychomotor speed is affected in amnestic MCI and AD. The association of motor performance with amyloid β deposition and atrophy could indicate that this is due to amyloid deposition in and atrophy of motor brain regions, which generally occurs later in the disease process. The promising discriminatory abilities of manual motor measures for AD emphasize their value alongside other cognitive and motor assessment outcomes in classification and prediction models, as well as potential enrichment of outcome variables in AD clinical trials.
Collapse
Affiliation(s)
- Vincent Koppelmans
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
- Huntsman Mental Health Institute, University of Utah, Salt Lake City, UT, USA
| | - Marit F.L. Ruitenberg
- Department of Health, Medical and Neuropsychology, Leiden University, Leiden, The Netherlands
- Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| | - Sydney Y. Schaefer
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Jace B. King
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| | - Jasmine M. Jacobo
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
- Huntsman Mental Health Institute, University of Utah, Salt Lake City, UT, USA
| | - Benjamin P. Silvester
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
- Huntsman Mental Health Institute, University of Utah, Salt Lake City, UT, USA
| | - Amanda F. Mejia
- Department of Statistics, University of Indiana, Bloomington, IN, USA
| | | | - John M. Hoffman
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
- Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Tolga Tasdizen
- Electrical and Computer Engineering, University of Utah, Salt Lake City, UT, USA
| | - Kevin Duff
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Department of Neurology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
17
|
Jácome D, Cotrufo T, Andrés-Benito P, Lidón L, Martí E, Ferrer I, Del Río JA, Gavín R. miR-519a-3p, found to regulate cellular prion protein during Alzheimer's disease pathogenesis, as a biomarker of asymptomatic stages. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167187. [PMID: 38653354 DOI: 10.1016/j.bbadis.2024.167187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
Clinical relevance of miRNAs as biomarkers is growing due to their stability and detection in biofluids. In this, diagnosis at asymptomatic stages of Alzheimer's disease (AD) remains a challenge since it can only be made at autopsy according to Braak NFT staging. Achieving the objective of detecting AD at early stages would allow possible therapies to be addressed before the onset of cognitive impairment. Many studies have determined that the expression pattern of some miRNAs is dysregulated in AD patients, but to date, none has been correlated with downregulated expression of cellular prion protein (PrPC) during disease progression. That is why, by means of cross studies of miRNAs up-regulated in AD with in silico identification of potential miRNAs-binding to 3'UTR of human PRNP gene, we selected miR-519a-3p for our study. Then, in vitro experiments were carried out in two ways. First, we validated miR-519a-3p target on 3'UTR-PRNP, and second, we analyzed the levels of PrPC expression after using of mimic technology on cell culture. In addition, RT-qPCR was performed to analyzed miR-519a-3p expression in human cerebral samples of AD at different stages of disease evolution. Additionally, samples of other neurodegenerative diseases such as other non-AD tauopathies and several synucleinopathies were included in the study. Our results showed that miR-519a-3p overlaps with PRNP 3'UTR in vitro and promotes downregulation of PrPC. Moreover, miR-519a-3p was found to be up-regulated exclusively in AD samples from stage I to VI, suggesting its potential use as a novel label of preclinical stages of the disease.
Collapse
Affiliation(s)
- Dayaneth Jácome
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain.
| | - Tiziana Cotrufo
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain; Institute of Neuroscience, University of Barcelona, Barcelona, Spain.
| | - Pol Andrés-Benito
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), Barcelona, Madrid, Spain; Neurologic Diseases and Neurogenetics Group, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.
| | - Laia Lidón
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain; Institute of Neuroscience, University of Barcelona, Barcelona, Spain; Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), Barcelona, Madrid, Spain.
| | - Eulàlia Martí
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain; Functional Genomics of Neurodegenerative Diseases, Department of Biomedical Sciences, University of Barcelona, Barcelona, Spain; CIBERESP (Centro en Red de Epidemiología y Salud Pública), Spain.
| | - Isidre Ferrer
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain; Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), Barcelona, Madrid, Spain; Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain; Senior Consultant Neuropathology, Service of Pathology, Bellvitge University Hospital, Hospitalet de Llobregat, Spain.
| | - José Antonio Del Río
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain; Institute of Neuroscience, University of Barcelona, Barcelona, Spain; Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), Barcelona, Madrid, Spain.
| | - Rosalina Gavín
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain; Institute of Neuroscience, University of Barcelona, Barcelona, Spain; Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), Barcelona, Madrid, Spain.
| |
Collapse
|
18
|
Huang Y, Wang YF, Miao J, Zheng RF, Li JY. Short-chain fatty acids: Important components of the gut-brain axis against AD. Biomed Pharmacother 2024; 175:116601. [PMID: 38749177 DOI: 10.1016/j.biopha.2024.116601] [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: 01/20/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 06/03/2024] Open
Abstract
Alzheimer's disease (AD) comprises a group of neurodegenerative disorders with some changes in the brain, which could lead to the deposition of certain proteins and result in the degeneration and death of brain cells. Patients with AD manifest primarily as cognitive decline, psychiatric symptoms, and behavioural disorders. Short-chain fatty acids (SCFAs) are a class of saturated fatty acids (SFAs) produced by gut microorganisms through the fermentation of dietary fibre ingested. SCFAs, as a significant mediator of signalling, can have diverse physiological and pathological roles in the brain through the gut-brain axis, and play a positive effect on AD via multiple pathways. Firstly, differences in SCFAs and microbial changes have been stated in AD cases of humans and mice in this paper. And then, mechanisms of three main SCFAs in treating with AD have been summarized, as well as differences of gut bacteria. Finally, functions of SCFAs played in regulating intestinal flora homeostasis, modulating the immune system, and the metabolic system, which were considered to be beneficial for the treatment of AD, have been elucidated, and the key roles of gut bacteria and SCFAs were pointed out. All in all, this paper provides an overview of SCFAs and gut bacteria in AD, and can help people to understand the importance of gut-brain axis in AD.
Collapse
Affiliation(s)
- Yan Huang
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China
| | - Yi Feng Wang
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China
| | - Jing Miao
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi 830004, China.
| | - Rui Fang Zheng
- Xinjiang Key Laboratory of Uygur Medical Research, Xinjiang Institute of Materia Medica, Urumqi 830004, China.
| | - Jin Yao Li
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi 830004, China.
| |
Collapse
|
19
|
Liu H, Yuan X, Liu T, Zhang W, Dong H, Chu Z. Freestanding Nanofiber-Assembled Aptasensor for Precisely and Ultrafast Electrochemical Detection of Alzheimer's Disease Biomarkers. Adv Healthc Mater 2024; 13:e2304355. [PMID: 38387159 PMCID: PMC11468682 DOI: 10.1002/adhm.202304355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/07/2024] [Indexed: 02/24/2024]
Abstract
Amyloid beta-protein (AβAβ) is a main hallmark of Alzheimer's disease (AD), and a low amount of Aβ protein accumulation appears to be a potential marker for AD. Here, an electrochemical DNA biosensor based on polyamide/polyaniline carbon nanotubes (PA/PANI-CNTs) is developed with the aim of diagnosing AD early using a simple, low-cost, and accessible method to rapidly detect Aβ42 in human blood. Electrospun PA nanofibers served as the skeleton for the successive in situ deposition of PANI and CNTs, which contribute both high conductivity and abundant binding sites for the Aβ42 aptamers. After the aptamers are immobilized, this aptasensor exhibits precise and specific detection of Aβ42 in human blood within only 4 min with an extremely fast response rate, lower detection limit, and excellent linear detection range. These findings make a significant contribution to advancing the development of serum-based detection techniques for Aβ42, thereby paving the way for improved diagnostic capabilities in the field of AD.
Collapse
Affiliation(s)
- Hui Liu
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolResearch Institute of StomatologyNanjing University30 Zhongyang RoadNanjingJiangsu210008China
| | - Xueli Yuan
- State Key Laboratory of Materials‐Oriented Chemical EngineeringCollege of Chemical EngineeringNanjing Tech UniversityNanjing211816China
| | - Tao Liu
- State Key Laboratory of Materials‐Oriented Chemical EngineeringCollege of Chemical EngineeringNanjing Tech UniversityNanjing211816China
| | - Wei Zhang
- Nanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing University321 Zhongshan RoadNanjing210008China
| | - Heng Dong
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolResearch Institute of StomatologyNanjing University30 Zhongyang RoadNanjingJiangsu210008China
| | - Zhenyu Chu
- State Key Laboratory of Materials‐Oriented Chemical EngineeringCollege of Chemical EngineeringNanjing Tech UniversityNanjing211816China
| |
Collapse
|
20
|
Kerl HU, Baazaoui H, Herrmann K, Adlung A, Ludwig NK, Hausner L, Frölich L, Schad L, Groden C, Mohamed SA. Sodium signal intensity of CSF using 1H-guided 23Na-MRI as a potential noninvasive biomarker in Alzheimer's disease. J Neuroimaging 2024. [PMID: 38807265 DOI: 10.1111/jon.13216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND AND PURPOSE Alzheimer's disease (AD) is characterized by cognitive decline and mnestic deficits. The pathophysiology of AD is not fully understood, which renders the development of accurate tools for early diagnosis and effective therapies exceedingly difficult. In this study, we investigated the use of 23Na-MRI to measure the relative sodium signal intensities (rSSIs) in CSF in patients with AD and healthy controls. METHODS We prospectively recruited 11 patients with biomarker-diagnosed early-stage AD, as well as 12 cognitively healthy age-matched controls. All participants underwent 23Na-MRI to measure rSSI. Statistical analyses were performed to compare CSF sodium signal intensities between groups and to evaluate the specificity and sensitivity of the rSSI in the diagnosis of AD. RESULTS RSSIs in CSF were significantly higher in AD patients (mean = 68.6% ± 7.7%) compared to healthy controls (mean = 56.9% ± 5.5%) (p < .001). There was also a significant negative correlation between rSSI in CSF and hippocampus and amygdala volumes (r = -.54 and -.49, p < .05) as well as a positive correlation with total CSF volumes (r = .81, p < .05). Receiver operating characteristic analysis showed high diagnostic accuracy for rSSI in discriminating between AD patients and healthy controls (area under the curve = .94). CONCLUSION Our study provides evidence that rSSI in CSF is increased in AD patients in comparison to healthy controls. rSSI may serve as a potential marker for early detection and monitoring of disease progression. Larger, longitudinal studies are needed to confirm our findings and to investigate the association between rSSI in CSF and the severity of cognitive impairment.
Collapse
Affiliation(s)
- Hans-Ulrich Kerl
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hakim Baazaoui
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Katrin Herrmann
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anne Adlung
- Department of Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Nadia K Ludwig
- Department of Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Löwenstein Medical Technology, Karlsruhe, Germany
| | - Lucrezia Hausner
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Lothar Schad
- Department of Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Groden
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sherif A Mohamed
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
21
|
Tao X, Zhu Z, Wang L, Li C, Sun L, Wang W, Gong W. Biomarkers of Aging and Relevant Evaluation Techniques: A Comprehensive Review. Aging Dis 2024; 15:977-1005. [PMID: 37611906 PMCID: PMC11081160 DOI: 10.14336/ad.2023.00808-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023] Open
Abstract
The risk of developing chronic illnesses and disabilities is increasing with age. To predict and prevent aging, biomarkers relevant to the aging process must be identified. This paper reviews the known molecular, cellular, and physiological biomarkers of aging. Moreover, we discuss the currently available technologies for identifying these biomarkers, and their applications and potential in aging research. We hope that this review will stimulate further research and innovation in this emerging and fast-growing field.
Collapse
Affiliation(s)
- Xue Tao
- Department of Research, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China.
| | - Ziman Zhu
- Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing, China.
| | - Liguo Wang
- Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
| | - Chunlin Li
- School of Biomedical Engineering, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing, China.
| | - Liwei Sun
- School of Biomedical Engineering, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing, China.
| | - Wei Wang
- Department of Rehabilitation Radiology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China.
| | - Weijun Gong
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
22
|
Leckey CA, Coulton JB, Giovannucci TA, He Y, Aslanyan A, Laban R, Heslegrave A, Doykov I, Ammoscato F, Chataway J, De Angelis F, Gnanapavan S, Byrne LM, Schott JM, Wild EJ, Barthelémy NR, Zetterberg H, Wray S, Bateman RJ, Mills K, Paterson RW. CSF neurofilament light chain profiling and quantitation in neurological diseases. Brain Commun 2024; 6:fcae132. [PMID: 38707707 PMCID: PMC11069115 DOI: 10.1093/braincomms/fcae132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 05/07/2024] Open
Abstract
Neurofilament light chain is an established marker of neuroaxonal injury that is elevated in CSF and blood across various neurological diseases. It is increasingly used in clinical practice to aid diagnosis and monitor progression and as an outcome measure to assess safety and efficacy of disease-modifying therapies across the clinical translational neuroscience field. Quantitative methods for neurofilament light chain in human biofluids have relied on immunoassays, which have limited capacity to describe the structure of the protein in CSF and how this might vary in different neurodegenerative diseases. In this study, we characterized and quantified neurofilament light chain species in CSF across neurodegenerative and neuroinflammatory diseases and healthy controls using targeted mass spectrometry. We show that the quantitative immunoprecipitation-tandem mass spectrometry method developed in this study strongly correlates to single-molecule array measurements in CSF across the broad spectrum of neurodegenerative diseases and was replicable across mass spectrometry methods and centres. In summary, we have created an accurate and cost-effective assay for measuring a key biomarker in translational neuroscience research and clinical practice, which can be easily multiplexed and translated into clinical laboratories for the screening and monitoring of neurodegenerative disease or acute brain injury.
Collapse
Affiliation(s)
- Claire A Leckey
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- Translational Mass Spectrometry Research Group, UCL Great Ormond Street Hospital Institute of Child Health, University College London, London, WC1N 1EH, UK
- UK Dementia Research Institute at UCL, University College London, London, WC1E 6BT, UK
| | - John B Coulton
- Department of Neurology, Washington University School of Medicine, Washington University in St Louis, St Louis, MO 63110, USA
- Tracy Family SILQ Center, Washington University School of Medicine, Washington University in St Louis, St Louis, MO 63110, USA
| | - Tatiana A Giovannucci
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, University College London, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Yingxin He
- Department of Neurology, Washington University School of Medicine, Washington University in St Louis, St Louis, MO 63110, USA
- Tracy Family SILQ Center, Washington University School of Medicine, Washington University in St Louis, St Louis, MO 63110, USA
| | - Aram Aslanyan
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Rhiannon Laban
- UK Dementia Research Institute at UCL, University College London, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Amanda Heslegrave
- UK Dementia Research Institute at UCL, University College London, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Ivan Doykov
- Translational Mass Spectrometry Research Group, UCL Great Ormond Street Hospital Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Francesca Ammoscato
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Blizard Institute, Centre for Neuroscience, London, E1 2AT, UK
| | - Jeremy Chataway
- Department of Neuroinflammation, Faculty of Brain Sciences, Queen Square Multiple Sclerosis Centre, UCL Queen Square Institute of Neurology, University College London, London, WC1B 5EH, UK
- National Institute for Health and Care Research, University College London Hospitals, Biomedical Research Centre, London, W1T 7DN, UK
| | - Floriana De Angelis
- Department of Neuroinflammation, Faculty of Brain Sciences, Queen Square Multiple Sclerosis Centre, UCL Queen Square Institute of Neurology, University College London, London, WC1B 5EH, UK
- National Institute for Health and Care Research, University College London Hospitals, Biomedical Research Centre, London, W1T 7DN, UK
| | | | - Lauren M Byrne
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Jonathan M Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Edward J Wild
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Nicolas R Barthelémy
- Department of Neurology, Washington University School of Medicine, Washington University in St Louis, St Louis, MO 63110, USA
- Tracy Family SILQ Center, Washington University School of Medicine, Washington University in St Louis, St Louis, MO 63110, USA
| | - Henrik Zetterberg
- UK Dementia Research Institute at UCL, University College London, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 43180, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 43180, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI53792, USA
| | - Selina Wray
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, Washington University in St Louis, St Louis, MO 63110, USA
- Tracy Family SILQ Center, Washington University School of Medicine, Washington University in St Louis, St Louis, MO 63110, USA
| | - Kevin Mills
- Translational Mass Spectrometry Research Group, UCL Great Ormond Street Hospital Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Ross W Paterson
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, University College London, London, WC1E 6BT, UK
- Department of Neurology, Darent Valley Hospital, Dartford, Kent, DA2 8DA, UK
| |
Collapse
|
23
|
Lu Y, Pike JR, Hoogeveen R, Walker K, Raffield L, Selvin E, Avery C, Engel S, Mielke MM, Garcia T, Heiss G, Palta P. Nonalcoholic Fatty Liver Disease and Longitudinal Change in Imaging and Plasma Biomarkers of Alzheimer Disease and Vascular Pathology. Neurology 2024; 102:e209203. [PMID: 38471046 PMCID: PMC11033987 DOI: 10.1212/wnl.0000000000209203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/23/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Prospective measures of plasma and cerebral MRI biomarkers of Alzheimer disease (AD) and vascular neuropathology provide an opportunity to investigate possible mechanisms linking liver disease and dementia. We aimed to quantify the association of midlife nonalcoholic fatty liver disease (NAFLD) with change in plasma and brain MRI biomarkers of AD and vascular neuropathology. METHODS We included participants from the Atherosclerosis Risk in Communities Study with brain MRI measurements of white matter hyperintensity (WMH) volume and temporal-parietal lobe cortical thickness meta region of interest (ROI) at up to 2 different visits, in 2011-13 and 2016-19, and plasma biomarkers of β-amyloid (Aβ)42:40, phosphorylated tau at threonine 181, and neurofilament light (NfL) were measured up to 3 times in 1993-95, 2011-13, and 2016-19. NAFLD was categorized using the fatty liver index in 1990-92. Multivariate linear regression was performed for associations between midlife NAFLD and change in plasma and brain MRI biomarkers of AD and vascular neuropathology. The primary models adjusted for demographics, Apolipoprotein E, alcohol use, and kidney function. RESULTS Among 1,706 participants (mean age 56 years, 62% female, 28% Black), midlife NAFLD vs no NAFLD was associated with greater late-life WMH volume (difference per SD 0.19, 95% CI 0.06-0.31) and faster late-life WMH increase over 6 years (difference in annual change, SD 0.28, 95% CI 0.05-0.51), suggesting accumulating vascular pathology. Midlife NAFLD vs no NAFLD was also associated with AD biomarkers in midlife (lower Aβ42:40 [SD -0.21, 95% CI -0.39 to -0.04] measured in 1993-95) and late life (lower Aβ42:40 [SD -0.13, 95% CI -0.23 to -0.03] and lower temporal-parietal lobe cortical thickness meta ROI [SD -0.16, 95% CI -0.28 to -0.05] measured in 2011-13). Although midlife NfL was lower in individuals with vs without midlife NAFLD, those with NAFLD exhibited a faster rate of NfL increase that accelerated over time. DISCUSSION Midlife NAFLD shows associations with AD and accumulating vascular pathology, revealing potential pathways linking liver function to dementia. Plasma biomarkers of neuropathology and neuronal injury may serve as easily measurable and dynamic indicators for monitoring the impacts of impaired liver function on brain health.
Collapse
Affiliation(s)
- Yifei Lu
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - James R Pike
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Ron Hoogeveen
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Keenan Walker
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Laura Raffield
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Elizabeth Selvin
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Christy Avery
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Stephanie Engel
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Michelle M Mielke
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Tanya Garcia
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Gerardo Heiss
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Priya Palta
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| |
Collapse
|
24
|
Shue F, White LJ, Hendrix R, Ulrich J, Henson RL, Knight W, Martens YA, Wang N, Roy B, Starling SC, Ren Y, Xiong C, Asmann YW, Syrjanen JA, Vassilaki M, Mielke MM, Timsina J, Sung YJ, Cruchaga C, Holtzman DM, Bu G, Petersen RC, Heckman MG, Kanekiyo T. CSF biomarkers of immune activation and Alzheimer's disease for predicting cognitive impairment risk in the elderly. SCIENCE ADVANCES 2024; 10:eadk3674. [PMID: 38569027 PMCID: PMC10990276 DOI: 10.1126/sciadv.adk3674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 02/23/2024] [Indexed: 04/05/2024]
Abstract
The immune system substantially influences age-related cognitive decline and Alzheimer's disease (AD) progression, affected by genetic and environmental factors. In a Mayo Clinic Study of Aging cohort, we examined how risk factors like APOE genotype, age, and sex affect inflammatory molecules and AD biomarkers in cerebrospinal fluid (CSF). Among cognitively unimpaired individuals over 65 (N = 298), we measured 365 CSF inflammatory molecules, finding age, sex, and diabetes status predominantly influencing their levels. We observed age-related correlations with AD biomarkers such as total tau, phosphorylated tau-181, neurofilament light chain (NfL), and YKL40. APOE4 was associated with lower Aβ42 and higher SNAP25 in CSF. We explored baseline variables predicting cognitive decline risk, finding age, CSF Aβ42, NfL, and REG4 to be independently correlated. Subjects with older age, lower Aβ42, higher NfL, and higher REG4 at baseline had increased cognitive impairment risk during follow-up. This suggests that assessing CSF inflammatory molecules and AD biomarkers could predict cognitive impairment risk in the elderly.
Collapse
Affiliation(s)
- Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Launia J. White
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Rachel Hendrix
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jason Ulrich
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rachel L. Henson
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - William Knight
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yuka A. Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ni Wang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Bhaskar Roy
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Yingxue Ren
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 93110, USA
| | - Yan W. Asmann
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Jeremy A. Syrjanen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester MN 55905, USA
| | - Maria Vassilaki
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester MN 55905, USA
| | - Michelle M. Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester MN 55905, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 93110, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 93110, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 93110, USA
| | - David M. Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Michael G. Heckman
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
25
|
Zhang Y, Lv Q, Yin Y, Wang H, Bueber MA, Phillips MR, Li T. Research in China about the biological mechanisms that potentially link socioenvironmental changes and mental health: a scoping review. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2024; 45:100610. [PMID: 38699292 PMCID: PMC11064722 DOI: 10.1016/j.lanwpc.2022.100610] [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] [Indexed: 05/05/2024]
Abstract
China's rapid socioeconomic development since 1990 makes it a fitting location to summarise research about how biological changes associated with socioenvironmental changes affect population mental health and, thus, lay the groundwork for subsequent, more focused studies. An initial search identified 308 review articles in the international literature about biomarkers associated with 12 common mental health disorders. We then searched for studies conducted in China that assessed the association of the identified mental health related-biomarkers with socioenvironmental factors in English-language and Chinese-language databases. We located 1330 articles published between 1 January 1990 and 1 August 2021 that reported a total of 3567 associations between 56 specific biomarkers and 11 socioenvironmental factors: 3156 (88·5%) about six types of environmental pollution, 381 (10·7%) about four health-related behaviours (diet, physical inactivity, internet misuse, and other lifestyle factors), and 30 (0·8%) about socioeconomic inequity. Only 245 (18·4%) of the papers simultaneously considered the possible effect of the biomarkers on mental health conditions; moreover, most of these studies assessed biomarkers in animal models of mental disorders, not human subjects. Among the 245 papers, mental health conditions were linked with biomarkers of environmental pollution in 188 (76·7%), with biomarkers of health-related behaviours in 48 (19·6%), and with biomarkers of socioeconomic inequality in 9 (3·7%). The 604 biomarker-mental health condition associations reported (107 in human subjects and 497 in animal models) included 379 (62·7%) about cognitive functioning, 117 (19·4%) about anxiety, 56 (9·3%) about depression, 21 (3·5%) about neurodevelopmental conditions, and 31 (5·1%) about neurobehavioural symptoms. Improved understanding of the biological mechanisms linking socioenvironmental changes to community mental health will require expanding the range of socioenvironmental factors considered, including mental health outcomes in more of the studies about the association of biomarkers with socioenvironmental factors, and increasing the proportion of studies that assess mental health outcomes in humans.
Collapse
Affiliation(s)
- Yamin Zhang
- Department of Neurobiology and Affiliated Mental Health Center, Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiuyue Lv
- Mental Health Center and Psychiatric Laboratory, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yubing Yin
- Mental Health Center and Psychiatric Laboratory, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Han Wang
- West China School of Medicine, Chengdu, Sichuan, China
| | - Marlys Ann Bueber
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Michael Robert Phillips
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Departments of Psychiatry and Epidemiology, Columbia University, New York, NY, USA
| | - Tao Li
- Department of Neurobiology and Affiliated Mental Health Center, Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
26
|
Shim KH, Kim D, Kang MJ, Pyun J, Park YH, Youn YC, Park KW, Suk K, Lee H, Gomes BF, Zetterberg H, An SSA, Kim S. Subsequent correlated changes in complement component 3 and amyloid beta oligomers in the blood of patients with Alzheimer's disease. Alzheimers Dement 2024; 20:2731-2741. [PMID: 38411315 PMCID: PMC11032549 DOI: 10.1002/alz.13734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 12/05/2023] [Accepted: 01/19/2024] [Indexed: 02/28/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) involves the complement cascade, with complement component 3 (C3) playing a key role. However, the relationship between C3 and amyloid beta (Aβ) in blood is limited. METHODS Plasma C3 and Aβ oligomerization tendency (AβOt) were measured in 35 AD patients and 62 healthy controls. Correlations with cerebrospinal fluid (CSF) biomarkers, cognitive impairment, and amyloid positron emission tomography (PET) were analyzed. Differences between biomarkers were compared in groups classified by concordances of biomarkers. RESULTS Plasma C3 and AβOt were elevated in AD patients and in CSF or amyloid PET-positive groups. Weak positive correlation was found between C3 and AβOt, while both had strong negative correlations with CSF Aβ42 and cognitive performance. Abnormalities were observed for AβOt and CSF Aβ42 followed by C3 changes. DISCUSSION Increased plasma C3 in AD are associated with amyloid pathology, possibly reflecting a defense response for Aβ clearance. Further studies on Aβ-binding proteins will enhance understanding of Aβ mechanisms in blood.
Collapse
Affiliation(s)
- Kyu Hwan Shim
- Department of Bionano TechnologyGachon UniversitySeongnamRepublic of Korea
| | - Danyeong Kim
- Department of Bionano TechnologyGachon UniversitySeongnamRepublic of Korea
| | - Min Ju Kang
- Department of NeurologyVeterans Medical Research InstituteVeterans Health Service Medical CenterSeoulRepublic of Korea
| | - Jung‐Min Pyun
- Department of NeurologySoonchunhyang University Seoul HospitalSoonchunhyang University College of MedicineSeoulRepublic of Korea
| | - Young Ho Park
- Department of NeurologySeoul National University College of Medicine and Clinical Neuroscience CenterSeoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Young Chul Youn
- Department of NeurologyChung‐Ang University College of MedicineSeoulRepublic of Korea
| | - Kyung Won Park
- Department of NeurologyDong‐A University College of Medicine and Institute of Convergence Bio‐HealthBusanRepublic of Korea
| | - Kyoungho Suk
- Department of PharmacologyKyungpook National University School of MedicineDaeguRepublic of Korea
| | - Ho‐Won Lee
- Department of NeurologyKyungpook National University School of MedicineDaeguRepublic of Korea
| | - Bárbara Fernandes Gomes
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of Neurology, Queen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water BayHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Seong Soo A. An
- Department of Bionano TechnologyGachon UniversitySeongnamRepublic of Korea
| | - SangYun Kim
- Department of NeurologySeoul National University College of Medicine and Clinical Neuroscience CenterSeoul National University Bundang HospitalSeongnamRepublic of Korea
| | | |
Collapse
|
27
|
Cheslow L, Snook AE, Waldman SA. Biomarkers for Managing Neurodegenerative Diseases. Biomolecules 2024; 14:398. [PMID: 38672416 PMCID: PMC11048498 DOI: 10.3390/biom14040398] [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: 03/03/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Neurological disorders are the leading cause of cognitive and physical disability worldwide, affecting 15% of the global population. Due to the demographics of aging, the prevalence of neurological disorders, including neurodegenerative diseases, will double over the next two decades. Unfortunately, while available therapies provide symptomatic relief for cognitive and motor impairment, there is an urgent unmet need to develop disease-modifying therapies that slow the rate of pathological progression. In that context, biomarkers could identify at-risk and prodromal patients, monitor disease progression, track responses to therapy, and parse the causality of molecular events to identify novel targets for further clinical investigation. Thus, identifying biomarkers that discriminate between diseases and reflect specific stages of pathology would catalyze the discovery and development of therapeutic targets. This review will describe the prevalence, known mechanisms, ongoing or recently concluded therapeutic clinical trials, and biomarkers of three of the most prevalent neurodegenerative diseases, including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Parkinson's disease (PD).
Collapse
Affiliation(s)
- Lara Cheslow
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (L.C.); (A.E.S.)
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Adam E. Snook
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (L.C.); (A.E.S.)
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Scott A. Waldman
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (L.C.); (A.E.S.)
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
28
|
Jiang H, Tang M, Xu Z, Wang Y, Li M, Zheng S, Zhu J, Lin Z, Zhang M. CRISPR/Cas9 system and its applications in nervous system diseases. Genes Dis 2024; 11:675-686. [PMID: 37692518 PMCID: PMC10491921 DOI: 10.1016/j.gendis.2023.03.017] [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: 11/28/2022] [Accepted: 03/05/2023] [Indexed: 09/12/2023] Open
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system is an acquired immune system of many bacteria and archaea, comprising CRISPR loci, Cas genes, and its associated proteins. This system can recognize exogenous DNA and utilize the Cas9 protein's nuclease activity to break DNA double-strand and to achieve base insertion or deletion by subsequent DNA repair. In recent years, multiple laboratory and clinical studies have revealed the therapeutic role of the CRISPR/Cas9 system in neurological diseases. This article reviews the CRISPR/Cas9-mediated gene editing technology and its potential for clinical application against neurological diseases.
Collapse
Affiliation(s)
- Haibin Jiang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Mengyan Tang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zidi Xu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yanan Wang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Mopu Li
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shuyin Zheng
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jianghu Zhu
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang 325027, China
| | - Zhenlang Lin
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang 325027, China
| | - Min Zhang
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
29
|
Manippa V, Palmisano A, Nitsche MA, Filardi M, Vilella D, Logroscino G, Rivolta D. Cognitive and Neuropathophysiological Outcomes of Gamma-tACS in Dementia: A Systematic Review. Neuropsychol Rev 2024; 34:338-361. [PMID: 36877327 PMCID: PMC10920470 DOI: 10.1007/s11065-023-09589-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 01/23/2023] [Indexed: 03/07/2023]
Abstract
Despite the numerous pharmacological interventions targeting dementia, no disease-modifying therapy is available, and the prognosis remains unfavorable. A promising perspective involves tackling high-frequency gamma-band (> 30 Hz) oscillations involved in hippocampal-mediated memory processes, which are impaired from the early stages of typical Alzheimer's Disease (AD). Particularly, the positive effects of gamma-band entrainment on mouse models of AD have prompted researchers to translate such findings into humans using transcranial alternating current stimulation (tACS), a methodology that allows the entrainment of endogenous cortical oscillations in a frequency-specific manner. This systematic review examines the state-of-the-art on the use of gamma-tACS in Mild Cognitive Impairment (MCI) and dementia patients to shed light on its feasibility, therapeutic impact, and clinical effectiveness. A systematic search from two databases yielded 499 records resulting in 10 included studies and a total of 273 patients. The results were arranged in single-session and multi-session protocols. Most of the studies demonstrated cognitive improvement following gamma-tACS, and some studies showed promising effects of gamma-tACS on neuropathological markers, suggesting the feasibility of gamma-tACS in these patients anyhow far from the strong evidence available for mouse models. Nonetheless, the small number of studies and their wide variability in terms of aims, parameters, and measures, make it difficult to draw firm conclusions. We discuss results and methodological limitations of the studies, proposing possible solutions and future avenues to improve research on the effects of gamma-tACS on dementia.
Collapse
Affiliation(s)
- Valerio Manippa
- Department of Education, Psychology and Communication, University of Bari "Aldo Moro", Bari, Italy.
| | - Annalisa Palmisano
- Department of Education, Psychology and Communication, University of Bari "Aldo Moro", Bari, Italy
| | - Michael A Nitsche
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
- Department of Neurology, University Medical Hospital Bergmannsheil, Bochum, Germany
| | - Marco Filardi
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro" at Pia Fondazione "Cardinale G. Panico", Tricase, Lecce, Italy
- Department of Basic Medicine, Neuroscience and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Davide Vilella
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro" at Pia Fondazione "Cardinale G. Panico", Tricase, Lecce, Italy
| | - Giancarlo Logroscino
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro" at Pia Fondazione "Cardinale G. Panico", Tricase, Lecce, Italy
- Department of Basic Medicine, Neuroscience and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Davide Rivolta
- Department of Education, Psychology and Communication, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
30
|
van der Gaag BL, Deshayes NAC, Breve JJP, Bol JGJM, Jonker AJ, Hoozemans JJM, Courade JP, van de Berg WDJ. Distinct tau and alpha-synuclein molecular signatures in Alzheimer's disease with and without Lewy bodies and Parkinson's disease with dementia. Acta Neuropathol 2024; 147:14. [PMID: 38198008 PMCID: PMC10781859 DOI: 10.1007/s00401-023-02657-y] [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: 07/30/2023] [Revised: 11/10/2023] [Accepted: 11/22/2023] [Indexed: 01/11/2024]
Abstract
Alpha-synuclein (aSyn) pathology is present in approximately 50% of Alzheimer's disease (AD) cases at autopsy and might impact the age-of-onset and disease progression in AD. Here, we aimed to determine whether tau and aSyn profiles differ between AD cases with Lewy bodies (AD-LB), pure AD and Parkinson's disease with dementia (PDD) cases using epitope-, post-translational modification- (PTM) and isoform-specific tau and aSyn antibody panels spanning from the N- to C-terminus. We included the middle temporal gyrus (MTG) and amygdala (AMY) of clinically diagnosed and pathologically confirmed cases and performed dot blotting, western blotting and immunohistochemistry combined with quantitative and morphological analyses. All investigated phospho-tau (pTau) species, except pT181, were upregulated in AD-LB and AD cases compared to PDD and control cases, but no significant differences were observed between AD-LB and AD subjects. In addition, tau antibodies targeting the proline-rich regions and C-terminus showed preferential binding to AD-LB and AD brain homogenates. Antibodies targeting C-terminal aSyn epitopes and pS129 aSyn showed stronger binding to AD-LB and PDD cases compared to AD and control cases. Two pTau species (pS198 and pS396) were specifically detected in the soluble protein fractions of AD-LB and AD subjects, indicative of early involvement of these PTMs in the multimerization process of tau. Other phospho-variants for both tau (pT212/S214, pT231 and pS422) and aSyn (pS129) were only detected in the insoluble protein fraction of AD-LB/AD and AD-LB/PDD cases, respectively. aSyn load was higher in the AMY of AD-LB cases compared to PDD cases, suggesting aggravated aSyn pathology under the presence of AD pathology, while tau load was similar between AD-LB and AD cases. Co-localization of pTau and aSyn could be observed within astrocytes of AD-LB cases within the MTG. These findings highlight a unique pathological signature for AD-LB cases compared to pure AD and PDD cases.
Collapse
Affiliation(s)
- Bram L van der Gaag
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Natasja A C Deshayes
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - John J P Breve
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - John G J M Bol
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Allert J Jonker
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Jeroen J M Hoozemans
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | | | - Wilma D J van de Berg
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands.
| |
Collapse
|
31
|
Xiong C, Schindler S, Luo J, Morris J, Bateman R, Holtzman D, Cruchaga C, Babulal G, Henson R, Benzinger T, Bui Q, Agboola F, Grant E, Emily G, Moulder K, Geldmacher D, Clay O, Roberson E, Murchison C, Wolk D, Shaw L. Baseline levels and longitudinal rates of change in plasma Aβ42/40 among self-identified Black/African American and White individuals. RESEARCH SQUARE 2024:rs.3.rs-3783571. [PMID: 38260384 PMCID: PMC10802715 DOI: 10.21203/rs.3.rs-3783571/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Objective The use of blood-based biomarkers of Alzheimer disease (AD) may facilitate access to biomarker testing of groups that have been historically under-represented in research. We evaluated whether plasma Aβ42/40 has similar or different baseline levels and longitudinal rates of change in participants racialized as Black or White. Methods The Study of Race to Understand Alzheimer Biomarkers (SORTOUT-AB) is a multi-center longitudinal study to evaluate for potential differences in AD biomarkers between individuals racialized as Black or White. Plasma samples collected at three AD Research Centers (Washington University, University of Pennsylvania, and University of Alabama-Birmingham) underwent analysis with C2N Diagnostics' PrecivityAD™ blood test for Aβ42 and Aβ40. General linear mixed effects models were used to estimate the baseline levels and rates of longitudinal change for plasma Aβ measures in both racial groups. Analyses also examined whether dementia status, age, sex, education, APOE ε4 carrier status, medical comorbidities, or fasting status modified potential racial differences. Results Of the 324 Black and 1,547 White participants, there were 158 Black and 759 White participants with plasma Aβ measures from at least two longitudinal samples over a mean interval of 6.62 years. At baseline, the group of Black participants had lower levels of plasma Aβ40 but similar levels of plasma Aβ42 as compared to the group of White participants. As a result, baseline plasma Aβ42/40 levels were higher in the Black group than the White group, consistent with the Black group having lower levels of amyloid pathology. Racial differences in plasma Aβ42/40 were not modified by age, sex, education, APOE ε4 carrier status, medical conditions (hypertension and diabetes), or fasting status. Despite differences in baseline levels, the Black and White groups had a similar longitudinal rate of change in plasma Aβ42/40. Interpretation Black individuals participating in AD research studies had a higher mean level of plasma Aβ42/40, consistent with a lower level of amyloid pathology, which, if confirmed, may imply a lower proportion of Black individuals being eligible for AD clinical trials in which the presence of amyloid is a prerequisite. However, there was no significant racial difference in the rate of change in plasma Aβ42/40, suggesting that amyloid pathology accumulates similarly across racialized groups.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Quoc Bui
- Washington University School of Medicine
| | | | | | | | | | | | | | | | | | - David Wolk
- Department of Neurology, University of Pennsylvania
| | - Leslie Shaw
- Perelman School of Medicine, University of Pennsylvania
| |
Collapse
|
32
|
Klee M, Aho VTE, May P, Heintz-Buschart A, Landoulsi Z, Jónsdóttir SR, Pauly C, Pavelka L, Delacour L, Kaysen A, Krüger R, Wilmes P, Leist AK. Education as Risk Factor of Mild Cognitive Impairment: The Link to the Gut Microbiome. J Prev Alzheimers Dis 2024; 11:759-768. [PMID: 38706292 PMCID: PMC11060993 DOI: 10.14283/jpad.2024.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 12/03/2023] [Indexed: 05/07/2024]
Abstract
BACKGROUND With differences apparent in the gut microbiome in mild cognitive impairment (MCI) and dementia, and risk factors of dementia linked to alterations of the gut microbiome, the question remains if gut microbiome characteristics may mediate associations of education with MCI. OBJECTIVES We sought to examine potential mediation of the association of education and MCI by gut microbiome diversity or composition. DESIGN Cross-sectional study. SETTING Luxembourg, the Greater Region (surrounding areas in Belgium, France, Germany). PARTICIPANTS Control participants of the Luxembourg Parkinson's Study. MEASUREMENTS Gut microbiome composition, ascertained with 16S rRNA gene amplicon sequencing. Differential abundance, assessed across education groups (0-10, 11-16, 16+ years of education). Alpha diversity (Chao1, Shannon and inverse Simpson indices). Mediation analysis with effect decomposition was conducted with education as exposure, MCI as outcome and gut microbiome metrics as mediators. RESULTS After exclusion of participants below 50, or with missing data, n=258 participants (n=58 MCI) were included (M [SD] Age=64.6 [8.3] years). Higher education (16+ years) was associated with MCI (Odds ratio natural direct effect=0.35 [95% CI 0.15-0.81]. Streptococcus and Lachnospiraceae-UCG-001 genera were more abundant in higher education. CONCLUSIONS Education is associated with gut microbiome composition and MCI risk without clear evidence for mediation. However, our results suggest signatures of the gut microbiome that have been identified previously in AD and MCI to be reflected in lower education and suggest education as important covariate in microbiome studies.
Collapse
Affiliation(s)
- M Klee
- Matthias Klee, University of Luxembourg, Institute for Research on Socio-Economic Inequality, Department of Social Sciences, 11, Porte des Sciences, L-4366, Esch-sur-Alzett, Luxembourg, Mail: , Phone: +352 46 66 44 5161
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Halder A, Drummond E. Strategies for translating proteomics discoveries into drug discovery for dementia. Neural Regen Res 2024; 19:132-139. [PMID: 37488854 PMCID: PMC10479849 DOI: 10.4103/1673-5374.373681] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/25/2023] [Accepted: 04/06/2023] [Indexed: 07/26/2023] Open
Abstract
Tauopathies, diseases characterized by neuropathological aggregates of tau including Alzheimer's disease and subtypes of frontotemporal dementia, make up the vast majority of dementia cases. Although there have been recent developments in tauopathy biomarkers and disease-modifying treatments, ongoing progress is required to ensure these are effective, economical, and accessible for the globally ageing population. As such, continued identification of new potential drug targets and biomarkers is critical. "Big data" studies, such as proteomics, can generate information on thousands of possible new targets for dementia diagnostics and therapeutics, but currently remain underutilized due to the lack of a clear process by which targets are selected for future drug development. In this review, we discuss current tauopathy biomarkers and therapeutics, and highlight areas in need of improvement, particularly when addressing the needs of frail, comorbid and cognitively impaired populations. We highlight biomarkers which have been developed from proteomic data, and outline possible future directions in this field. We propose new criteria by which potential targets in proteomics studies can be objectively ranked as favorable for drug development, and demonstrate its application to our group's recent tau interactome dataset as an example.
Collapse
Affiliation(s)
- Aditi Halder
- School of Medical Sciences and Brain & Mind Center, University of Sydney, NSW, Sydney, Australia
- Department of Aged Care, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Eleanor Drummond
- School of Medical Sciences and Brain & Mind Center, University of Sydney, NSW, Sydney, Australia
| |
Collapse
|
34
|
Tripathi S, Sharma Y, Rane R, Kumar D. CRISPR/Cas9 Gene Editing: A Novel Approach Towards Alzheimer's Disease Treatment. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1405-1424. [PMID: 38716549 DOI: 10.2174/0118715273283786240408034408] [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: 11/15/2023] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 10/22/2024]
Abstract
In defiance of the vast amount of information regarding Alzheimer's disease (AD) that has been learned over the past thirty years, progress toward developing an effective therapy has been difficult. A neurological ailment that progresses and cannot be reversed is Alzheimer's disease, which shows neurofibrillary tangles, beta-amyloid plaque, and a lack of cognitive processes that is created by tau protein clumps with hyperphosphorylation that finally advances to neuronal damage without a recognized treatment, which has stimulated research into new therapeutic strategies. The protein CAS9 is linked to CRISPR, which is a clustered Regularly Interspaced Short Palindromic Repeat that inactivates or corrects a gene by recognizing a gene sequence that produces a doublestranded break has enchanted a whole amount of interest towards its potency to cure gene sequences in AD. The novel CRISPR-Cas9 applications for developing in vitro and in vivo models to the benefit of AD investigation and therapies are thoroughly analyzed in this work. The discussion will also touch on the creation of delivery methods, which is a significant obstacle to the therapeutic use of CRISPR/Cas9 technology. By concentrating on specific genes, such as those that are significant early- onset AD risk factors and late-onset AD risk factors, like the apolipoprotein E4 (APOE4) gene, this study aims to evaluate the potential application of CRISPR/Cas9 as a possible treatment for AD.
Collapse
Affiliation(s)
- Siddhant Tripathi
- Department of Pharm Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune Maharashtra 411038, India
| | - Yashika Sharma
- Department of Pharm Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune Maharashtra 411038, India
| | - Rajesh Rane
- Department of Pharm Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune Maharashtra 411038, India
| | - Dileep Kumar
- Department of Pharm Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune Maharashtra 411038, India
| |
Collapse
|
35
|
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
|
36
|
Angelidou IA, Stocker H, Beyreuther K, Teichmann B. Validation of the "Perceptions Regarding pRE-Symptomatic Alzheimer's Disease Screening" (PRE-ADS) Questionnaire in the German Population: Attitudes, Motivations, and Barriers to Pre-Symptomatic Dementia Screening. J Alzheimers Dis 2024; 97:309-325. [PMID: 38189757 PMCID: PMC10789340 DOI: 10.3233/jad-230961] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Attitudes, motivations, and barriers to pre-symptomatic screening for Alzheimer's disease (AD) in the general population are unclear, and validated measurement tools are lacking. OBJECTIVE Translation and validation of the German version of the "Perceptions regarding pRE-symptomatic Alzheimer's Disease Screening" (PRE-ADS) questionnaire. METHODS A convenience sample (N = 256) was recruited via an online platform. Validation of the PRE-ADS-D consisted of assessments of reliability, structural validity using Principal Component Analysis (PCA) and Exploratory Factor Analysis (EFA) and construct validity using known-group tests. A subscale "Acceptability of Screening", with 5 PRE-ADS-D items, was extracted to measure acceptance of screening in clinical practice. The STROBE checklist was used for reporting. RESULTS EFA revealed a three-factor model for the PRE-ADS-D. Acceptable to good internal consistency was found for the 25-item scale (α= 0.78), as well as for the three factors "Concerns about Screening" (α= 0.85), "Intention to be Screened" (α= 0.87), and "Preventive Health Behaviors" (α= 0.81). Construct validity was confirmed for both the 25-item PRE-ADS-D and the "Acceptability of Screening" scale (α= 0.91). Overall, 51.2% of the participants showed a preference for screening. Non-parametric tests were conducted to further explore group differences of the sample. CONCLUSIONS The PRE-ADS-D is a reliable and valid tool to measure attitudes, motives, and barriers regarding pre-symptomatic dementia screening in the German-speaking general population. Additionally, the subscale "Acceptability of Screening" demonstrated good construct validity and reliability, suggesting its promising potential as a practical tool in clinical practice.
Collapse
Affiliation(s)
| | - Hannah Stocker
- Network Aging Research, Heidelberg University, Heidelberg, Germany
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | | | - Birgit Teichmann
- Network Aging Research, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
37
|
Zheng HT, Wu Z, Mielke MM, Murray AM, Ryan J. Plasma Biomarkers of Alzheimer's Disease and Neurodegeneration According to Sociodemographic Characteristics and Chronic Health Conditions. J Prev Alzheimers Dis 2024; 11:1189-1197. [PMID: 39350363 PMCID: PMC11436401 DOI: 10.14283/jpad.2024.142] [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: 05/21/2024] [Accepted: 06/23/2024] [Indexed: 10/04/2024]
Abstract
Ultrasensitive assays have been developed which enable biomarkers of Alzheimer's disease pathology and neurodegeneration to be measured in blood. These biomarkers can aid in diagnosis, and have been used to predict risk of cognitive decline and Alzheimer's disease. The ease and cost-effectiveness of blood collections means that these biomarkers could be applied more broadly in population-based screening, however it is critical to first understand what other factors could affect blood biomarker levels. The aim of this review was to determine the extent that sociodemographic, lifestyle and health factors have been associated with blood biomarkers of Alzheimer's disease and neuropathology. Of the 32 studies included in this review, all but one measured biomarker levels in plasma, and age and sex were the most commonly investigated factors. The most consistent significant findings were a positive association between age and neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP), and females had higher GFAP than men. Apolipoprotein ε4 allele carriers had lower Aβ42 and Aβ42/40 ratio. Body mass index was negatively associated with GFAP and NfL, and chronic kidney disease with higher levels of all biomarkers. Too few studies have investigated other chronic health conditions and this requires further investigation. Given the potential for plasma biomarkers to enhance Alzheimer's disease diagnosis in primary care, it is important to understand how to interpret the biomarkers in light of factors that physiologically impact blood biomarker levels. This information will be critical for the establishment of reference ranges and thus the correct interpretation of these biomarkers in clinical screening.
Collapse
Affiliation(s)
- H T Zheng
- Joanne Ryan, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia,
| | | | | | | | | |
Collapse
|
38
|
Li Z, Fan Z, Zhang Q. The Associations of Phosphorylated Tau 181 and Tau 231 Levels in Plasma and Cerebrospinal Fluid with Cognitive Function in Alzheimer's Disease: A Systematic Review and Meta-Analysis. J Alzheimers Dis 2024; 98:13-32. [PMID: 38339929 DOI: 10.3233/jad-230799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Background Cerebrospinal fluid (CSF) or blood biomarkers like phosphorylated tau proteins (p-tau) are used to detect Alzheimer's disease (AD) early. Increasing studies on cognitive function and blood or CSF p-tau levels are controversial. Objective Our study examined the potential of p-tau as a biomarker of cognitive status in normal control (NC), mild cognitive impairment (MCI), and AD patients. Methods We searched PubMed, Cochrane, Embase, and Web of Science for relevant material through 12 January 2023. 5,017 participants from 20 studies-1,033 AD, 2,077 MCI, and 1,907 NC-were evaluated. Quantitative analysis provided continuous outcomes as SMDs with 95% CIs. Begg tested publication bias. Results MCI patients had lower CSF p-tau181 levels than AD patients (SMD =-0.60, 95% CI (-0.85, -0.36)) but higher than healthy controls (SMD = 0.67). AD/MCI patients had greater plasma p-tau181 levels than healthy people (SMD =-0.73, 95% CI (-1.04, -0.43)). MCI patients had significantly lower p-tau231 levels than AD patients in plasma and CSF (SMD =-0.90, 95% CI (-0.82, -0.45)). MCI patients showed greater CSF and plasma p-tau231 than healthy controls (SMD = 1.34, 95% CI (0.89, 1.79) and 0.43, (0.23, 0.64)). Plasma p-tau181/231 levels also distinguished the three categories. MCI patients had higher levels than healthy people, while AD patients had higher levels than MCI patients. Conclusions CSF p-tau181 and p-tau231 biomarkers distinguished AD, MCI, and healthy populations. Plasma-based p-tau181 and p-tau231 biomarkers for AD and MCI need further study.
Collapse
Affiliation(s)
- Zhirui Li
- Department of Disease Control and Prevention, Sichuan Provincial Center for Disease Control and Prevention, Sichuan Chengdu, China
| | - Zixuan Fan
- School of Health Policy and Management, Peking Union Medical College, Beijing, China
| | - Qian Zhang
- Department of Oncology, Xiamen Fifth Hospital, Fujian Xiamen, China
| |
Collapse
|
39
|
Elghanam Y, Purja S, Kim EY. Biomarkers as Endpoints in Clinical Trials for Alzheimer's Disease. J Alzheimers Dis 2024; 99:693-703. [PMID: 38669547 DOI: 10.3233/jad-240008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disease that imposes economic and societal burden. Biomarkers have played a crucial role in the recent approval of aducanumab and lecanemab as disease-modifying therapies which marked a significant milestone for the treatment of AD. The inclusion of biomarkers in AD trials facilitates precise diagnosis, monitors safety, demonstrates target engagement, and supports disease modification. Objective This study analyzed the utilization state and trends of biomarkers as endpoints in AD trials. Methods In this retrospective study, trials were collected by searching clinicaltrials.gov using the term "Alzheimer". Primary and secondary outcomes were analyzed separately for each phase. Results Among the 1,048 analyzed trials, 313 (29.87%) adopted biomarkers as primary endpoints and 364 (34.73%) as secondary endpoints, mainly in phases 1 and 2. The top three biomarkers adopted as primary endpoints in phases 1, 2, and 3 were amyloid-PET, tau-PET, and MRI. The top three biomarkers adopted as secondary endpoints, in phase 1, were cerebrospinal fluid (CSF) amyloid-β (Aβ), blood Aβ and amyloid-PET; in phase 2, they were MRI, CSF Aβ, and CSF phospho-tau; and in phase 3, they were amyloid PET, MRI, and blood Aβ. There was a statistically significant increase in the adoption of biomarkers as primary endpoints in phase 2 trials (p = 0.001) and secondary endpoints in phase 3 trials (p = 0.001). Conclusions The growing recognition of the importance of biomarkers in AD trial' design and drug development is evident by the significant steady increase in biomarkers' utilization in phases 2 and 3.
Collapse
Affiliation(s)
- Yomna Elghanam
- Department of Health, Evidence-Based and Clinical Research Laboratory, Social, and Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Sujata Purja
- Department of Health, Evidence-Based and Clinical Research Laboratory, Social, and Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Eun Young Kim
- Department of Health, Evidence-Based and Clinical Research Laboratory, Social, and Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul, Korea
- The Graduate School for Pharmaceutical Industry Management, College of Pharmacy, Chung-Ang University, Seoul, Korea
- The Department of Pharmaceutical Regulatory Sciences, Chung-Ang University, Seoul, Korea
| |
Collapse
|
40
|
Elliott ML, Nielsen JA, Hanford LC, Hamadeh A, Hilbert T, Kober T, Dickerson BC, Hyman BT, Mair RW, Eldaief MC, Buckner RL. Precision Brain Morphometry Using Cluster Scanning. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.23.23300492. [PMID: 38234845 PMCID: PMC10793507 DOI: 10.1101/2023.12.23.23300492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Measurement error limits the statistical power to detect group differences and longitudinal change in structural MRI morphometric measures (e.g., hippocampal volume, prefrontal thickness). Recent advances in scan acceleration enable extremely fast T1-weighted scans (~1 minute) to achieve morphometric errors that are close to the errors in longer traditional scans. As acceleration allows multiple scans to be acquired in rapid succession, it becomes possible to pool estimates to increase measurement precision, a strategy known as "cluster scanning." Here we explored brain morphometry using cluster scanning in a test-retest study of 40 individuals (12 younger adults, 18 cognitively unimpaired older adults, and 10 adults diagnosed with mild cognitive impairment or Alzheimer's Dementia). Morphometric errors from a single compressed sensing (CS) 1.0mm scan with 6x acceleration (CSx6) were, on average, 12% larger than a traditional scan using the Alzheimer's Disease Neuroimaging Initiative (ADNI) protocol. Pooled estimates from four clustered CSx6 acquisitions led to errors that were 34% smaller than ADNI despite having a shorter total acquisition time. Given a fixed amount of time, a gain in measurement precision can thus be achieved by acquiring multiple rapid scans instead of a single traditional scan. Errors were further reduced when estimates were pooled from eight CSx6 scans (51% smaller than ADNI). Neither pooling across a break nor pooling across multiple scan resolutions boosted this benefit. We discuss the potential of cluster scanning to improve morphometric precision, boost statistical power, and produce more sensitive disease progression biomarkers.
Collapse
Affiliation(s)
- Maxwell L Elliott
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Jared A Nielsen
- Department of Psychology, Neuroscience Center, Brigham Young University, Provo, UT, 84602, USA
| | - Lindsay C Hanford
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Aya Hamadeh
- Baylor College of Medicine, Houston, TX 77030
| | - Tom Hilbert
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Switzerland
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tobias Kober
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Switzerland
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Bradford C Dickerson
- Frontotemporal Disorders Unit
- Alzheimer's Disease Research Center
- Athinoula A. Martinos Center for Biomedical Imaging
- Department of Neurology, Massachusetts General Hospital & Harvard Medical School
- Department of Psychiatry, Massachusetts General Hospital & Harvard Medical School, Charlestown, MA 02129, USA
| | - Bradley T Hyman
- Alzheimer's Disease Research Center
- Department of Neurology, Massachusetts General Hospital & Harvard Medical School
| | - Ross W Mair
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Athinoula A. Martinos Center for Biomedical Imaging
| | - Mark C Eldaief
- Frontotemporal Disorders Unit
- Alzheimer's Disease Research Center
- Department of Neurology, Massachusetts General Hospital & Harvard Medical School
- Department of Psychiatry, Massachusetts General Hospital & Harvard Medical School, Charlestown, MA 02129, USA
| | - Randy L Buckner
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Alzheimer's Disease Research Center
- Athinoula A. Martinos Center for Biomedical Imaging
- Department of Psychiatry, Massachusetts General Hospital & Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
41
|
Rahmani F, Brier MR, Gordon BA, McKay N, Flores S, Keefe S, Hornbeck R, Ances B, Joseph‐Mathurin N, Xiong C, Wang G, Raji CA, Libre‐Guerra JJ, Perrin RJ, McDade E, Daniels A, Karch C, Day GS, Brickman AM, Fulham M, Jack CR, la La Fougère C, Reischl G, Schofield PR, Oh H, Levin J, Vöglein J, Cash DM, Yakushev I, Ikeuchi T, Klunk WE, Morris JC, Bateman RJ, Benzinger TLS. T1 and FLAIR signal intensities are related to tau pathology in dominantly inherited Alzheimer disease. Hum Brain Mapp 2023; 44:6375-6387. [PMID: 37867465 PMCID: PMC10681640 DOI: 10.1002/hbm.26514] [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: 07/24/2023] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 10/24/2023] Open
Abstract
Carriers of mutations responsible for dominantly inherited Alzheimer disease provide a unique opportunity to study potential imaging biomarkers. Biomarkers based on routinely acquired clinical MR images, could supplement the extant invasive or logistically challenging) biomarker studies. We used 1104 longitudinal MR, 324 amyloid beta, and 87 tau positron emission tomography imaging sessions from 525 participants enrolled in the Dominantly Inherited Alzheimer Network Observational Study to extract novel imaging metrics representing the mean (μ) and standard deviation (σ) of standardized image intensities of T1-weighted and Fluid attenuated inversion recovery (FLAIR) MR scans. There was an exponential decrease in FLAIR-μ in mutation carriers and an increase in FLAIR and T1 signal heterogeneity (T1-σ and FLAIR-σ) as participants approached the symptom onset in both supramarginal, the right postcentral and right superior temporal gyri as well as both caudate nuclei, putamina, thalami, and amygdalae. After controlling for the effect of regional atrophy, FLAIR-μ decreased and T1-σ and FLAIR-σ increased with increasing amyloid beta and tau deposition in numerous cortical regions. In symptomatic mutation carriers and independent of the effect of regional atrophy, tau pathology demonstrated a stronger relationship with image intensity metrics, compared with amyloid pathology. We propose novel MR imaging intensity-based metrics using standard clinical T1 and FLAIR images which strongly associates with the progression of pathology in dominantly inherited Alzheimer disease. We suggest that tau pathology may be a key driver of the observed changes in this cohort of patients.
Collapse
Affiliation(s)
| | | | - Brian A. Gordon
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Nicole McKay
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Shaney Flores
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Sarah Keefe
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Russ Hornbeck
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Beau Ances
- Washington University School of MedicineSt. LouisMissouriUSA
| | | | - Chengjie Xiong
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Guoqiao Wang
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Cyrus A. Raji
- Washington University School of MedicineSt. LouisMissouriUSA
| | | | | | - Eric McDade
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Alisha Daniels
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Celeste Karch
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Gregory S. Day
- Mayo Clinic, Department of NeurologyJacksonvilleFloridaUSA
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer's Disease & the Aging Brain, and Department of Neurology College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | | | | | - Christian la La Fougère
- Department of Nuclear Medicine and Clinical Molecular ImagingUniversity Hospital TuebingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE) TuebingenTübingenGermany
- Department of Preclinical Imaging and RadiopharmacyEberhard Karls University TübingenTübingenGermany
| | - Gerald Reischl
- Department of Nuclear Medicine and Clinical Molecular ImagingUniversity Hospital TuebingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE) TuebingenTübingenGermany
- Department of Preclinical Imaging and RadiopharmacyEberhard Karls University TübingenTübingenGermany
| | - Peter R. Schofield
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
- School of Biomedical SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - Hwamee Oh
- Brown UniversityProvidenceRhode IslandUSA
| | - Johannes Levin
- Department of NeurologyLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE), site MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Jonathan Vöglein
- Department of NeurologyLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE), site MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - David M. Cash
- UK Dementia Research Institute at University College LondonLondonUK
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | - Igor Yakushev
- Department of NeurologyLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE), site MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | | | | | - John C. Morris
- Washington University School of MedicineSt. LouisMissouriUSA
| | | | | | | |
Collapse
|
42
|
Sosa S, Bringas G, Urrutia N, Peñalver AI, López D, González E, Fernández A, Hernández ZM, Viña A, Peña Y, Batista JF, Valenzuela C, León K, Crombet T, Rodríguez T, Pérez L. NeuroEPO plus (NeuralCIM ®) in mild-to-moderate Alzheimer's clinical syndrome: the ATHENEA randomized clinical trial. Alzheimers Res Ther 2023; 15:215. [PMID: 38093366 PMCID: PMC10716956 DOI: 10.1186/s13195-023-01356-w] [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: 08/24/2023] [Accepted: 11/19/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND NeuroEPO plus is a recombinant human erythropoietin without erythropoietic activity and shorter plasma half-life due to its low sialic acid content. NeuroEPO plus prevents oxidative damage, neuroinflammation, apoptosis and cognitive deficit in an Alzheimer's disease (AD) models. The aim of this study was to assess efficacy and safety of neuroEPO plus. METHODS This was a double-blind, randomized, placebo-controlled, phase 2-3 trial involving participants ≥ 50 years of age with mild-to-moderate AD clinical syndrome. Participants were randomized in a 1:1:1 ratio to receive 0.5 or 1.0 mg of neuroEPO plus or placebo intranasally 3 times/week for 48 weeks. The primary outcome was change in the 11-item cognitive subscale of the AD Assessment Scale (ADAS-Cog11) score from baseline to 48 weeks (range, 0 to 70; higher scores indicate greater impairment). Secondary outcomes included CIBIC+, GDS, MoCA, NPI, Activities of Daily Living Scales, cerebral perfusion, and hippocampal volume. RESULTS A total of 174 participants were enrolled and 170 were treated (57 in neuroEPO plus 0.5 mg, 56 in neuroEPO plus 1.0 mg and 57 in placebo group). Mean age, 74.0 years; 121 (71.2%) women and 85% completed the trial. The median change in ADAS-Cog11 score at 48 weeks was -3.0 (95% CI, -4.3 to -1.7) in the 0.5 mg neuroEPO plus group, -4.0 (95% CI, -5.9 to -2.1) in the 1.0 mg neuroEPO plus group and 4.0 (95% CI, 1.9 to 6.1) in the placebo group. The difference of neuroEPO plus 0.5 mg vs. placebo was 7.0 points (95% CI, 4.5-9.5) P = 0.000 and between the neuroEPO plus 1.0 mg vs. placebo was 8.0 points (95% CI, 5.2-10.8) P = 0.000. NeuroEPO plus treatment induced a statistically significant improvement in some of clinical secondary outcomes vs. placebo including CIBIC+, GDS, MoCA, NPI, and the brain perfusion. CONCLUSIONS Among participants with mild-moderate Alzheimer's disease clinical syndrome, neuroEPO plus improved the cognitive evaluation at 48 weeks, with a very good safety profile. Larger trials are warranted to determine the efficacy and safety of neuroEPO plus in Alzheimer's disease. TRIAL REGISTRATION https://rpcec.sld.cu Identifier: RPCEC00000232.
Collapse
Affiliation(s)
- Saily Sosa
- Hospital Iván Portuondo, Calle 78 e/ Ave. 33 y 37, San Antonio de los Baños, Artemisa, CP 32 500, Cuba
| | - Giosmany Bringas
- National Institute of Neurology (INN), Calle 29 esquina D, Vedado, Havana, CP 10 400, Cuba
| | - Nelky Urrutia
- Hospital Iván Portuondo, Calle 78 e/ Ave. 33 y 37, San Antonio de los Baños, Artemisa, CP 32 500, Cuba
| | - Ana Ivis Peñalver
- National Institute of Neurology (INN), Calle 29 esquina D, Vedado, Havana, CP 10 400, Cuba
| | - Danay López
- Hospital Iván Portuondo, Calle 78 e/ Ave. 33 y 37, San Antonio de los Baños, Artemisa, CP 32 500, Cuba
| | - Evelio González
- Cuban Neurosciences Center (CNEURO), Avenida 25, No. 15 007, Cubanacán, Havana, CP 11 600, Cuba
| | - Ana Fernández
- Cuban Neurosciences Center (CNEURO), Avenida 25, No. 15 007, Cubanacán, Havana, CP 11 600, Cuba
| | - Zenaida Milagros Hernández
- Center of Neurological Restoration (CIREN), Calle 216 esquina 13, Siboney, Playa, Havana, CP 11 600, Cuba
| | - Ariel Viña
- Cuban Neurosciences Center (CNEURO), Avenida 25, No. 15 007, Cubanacán, Havana, CP 11 600, Cuba
| | - Yamile Peña
- Center for Clinical Investigation, CENTIS, Calle 45 No. 4501, esquina a 34, Reparto Kolhy, Havana, CP 11 300, Cuba
| | - Juan Felipe Batista
- Center for Clinical Investigation, CENTIS, Calle 45 No. 4501, esquina a 34, Reparto Kolhy, Havana, CP 11 300, Cuba
| | - Carmen Valenzuela
- Institute of Cybernetics, Mathematics and Physics (ICIMAF), Calle 15 #551 entre C y D, Plaza de la Revolución, Vedado, Havana, CP 10 400, Cuba
| | - Kalet León
- Center of Molecular Immunology (CIM), Calle 216 esquina 15, Siboney, Playa , Havana, CP 11 600, Cuba
| | - Tania Crombet
- Center of Molecular Immunology (CIM), Calle 216 esquina 15, Siboney, Playa , Havana, CP 11 600, Cuba
| | - Teresita Rodríguez
- Center of Molecular Immunology (CIM), Calle 216 esquina 15, Siboney, Playa , Havana, CP 11 600, Cuba
| | - Leslie Pérez
- Center of Molecular Immunology (CIM), Calle 216 esquina 15, Siboney, Playa , Havana, CP 11 600, Cuba.
| |
Collapse
|
43
|
Frank A, Ismail Z, Wilson M, Gauthier S, Verret L, Hsiung GYR, Borrie M. Health System Change for Alzheimer's Disease-Modifying Therapies in Canada: Beginning the Discussion. Can J Neurol Sci 2023:1-9. [PMID: 38052729 DOI: 10.1017/cjn.2023.322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that accounts for 60%-70% of patients with dementia, and it is estimated that over one million Canadians will be living with dementia by 2030. Disease-modifying therapies (DMTs) targeting the underlying pathophysiology of AD are currently in development. Several models have demonstrated that the potential arrival of Alzheimer's DMTs will most likely overwhelm the already-constrained Canadian healthcare system. Canada does not have a strategy to address the extensive requirements of using DMTs, including providing an early diagnosis of AD, confirming DMT eligibility via amyloid biomarkers, and conducting ongoing treatment monitoring. Thus, a multidisciplinary group of experts involved in AD care in Canada gathered to review (1) the current barriers to diagnosis and management of AD; (2) how existing clinic models, including those used in multiple sclerosis (MS), could be applied to address key barriers in AD; and (3) how to design and implement optimal care pathways in the future. The actions outlined in this review will help clinicians and healthcare systems improve readiness to integrate the use of disease-modifying therapies in Alzheimer's disease, if such therapies are approved in Canada.
Collapse
Affiliation(s)
- Andrew Frank
- Bruyere Research Institute, and University of Ottawa, Ottawa, ON, Canada
| | - Zahinoor Ismail
- Departments of Psychiatry, Clinical Neurosciences, and Community Health Sciences, Hotchkiss Brain Institute and O'Brien Institute for Public Health, University of Calgary, Calgary, AB, Canada
| | | | - Serge Gauthier
- McGill Center for Studies in Aging and Dementia Education Program, McGill University, Montreal, QC, Canada
| | - Louis Verret
- Service de Neurologie, Clinique Interdisciplinaire de Mémoire, CHU de Québec-Université Laval, Hôpital Enfant-Jésus, Québec, QC, Canada
| | - Ging-Yuek Robin Hsiung
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Michael Borrie
- Division of Geriatric Medicine, Lawson Health Research Institute, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| |
Collapse
|
44
|
Bhattarai P, Taha A, Soni B, Thakuri DS, Ritter E, Chand GB. Predicting cognitive dysfunction and regional hubs using Braak staging amyloid-beta biomarkers and machine learning. Brain Inform 2023; 10:33. [PMID: 38043122 PMCID: PMC10694120 DOI: 10.1186/s40708-023-00213-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/21/2023] [Indexed: 12/05/2023] Open
Abstract
Mild cognitive impairment (MCI) is a transitional stage between normal aging and early Alzheimer's disease (AD). The presence of extracellular amyloid-beta (Aβ) in Braak regions suggests a connection with cognitive dysfunction in MCI/AD. Investigating the multivariate predictive relationships between regional Aβ biomarkers and cognitive function can aid in the early detection and prevention of AD. We introduced machine learning approaches to estimate cognitive dysfunction from regional Aβ biomarkers and identify the Aβ-related dominant brain regions involved with cognitive impairment. We employed Aβ biomarkers and cognitive measurements from the same individuals to train support vector regression (SVR) and artificial neural network (ANN) models and predict cognitive performance solely based on Aβ biomarkers on the test set. To identify Aβ-related dominant brain regions involved in cognitive prediction, we built the local interpretable model-agnostic explanations (LIME) model. We found elevated Aβ in MCI compared to controls and a stronger correlation between Aβ and cognition, particularly in Braak stages III-IV and V-VII (p < 0.05) biomarkers. Both SVR and ANN, especially ANN, showed strong predictive relationships between regional Aβ biomarkers and cognitive impairment (p < 0.05). LIME integrated with ANN showed that the parahippocampal gyrus, inferior temporal gyrus, and hippocampus were the most decisive Braak regions for predicting cognitive decline. Consistent with previous findings, this new approach suggests relationships between Aβ biomarkers and cognitive impairment. The proposed analytical framework can estimate cognitive impairment from Braak staging Aβ biomarkers and delineate the dominant brain regions collectively involved in AD pathophysiology.
Collapse
Affiliation(s)
- Puskar Bhattarai
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ahmed Taha
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Bhavin Soni
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Deepa S Thakuri
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- University of Missouri School of Medicine, Columbia, MO, USA
| | - Erin Ritter
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University McKelvey School of Engineering, St. Louis, MO, USA
| | - Ganesh B Chand
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA.
- Imaging Core, Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
- Institute of Clinical and Translational Sciences, Washington University School of Medicine, St. Louis, MO, USA.
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
45
|
Gao Y, Su D, Xue Z, Ji L, Wang S. Association Between Serum Neurofilament Light Chain and Cognitive Performance Among Older Adults in the United States: A Cross-Sectional Study. Neurol Ther 2023; 12:2147-2160. [PMID: 37845473 PMCID: PMC10630257 DOI: 10.1007/s40120-023-00555-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 09/29/2023] [Indexed: 10/18/2023] Open
Abstract
INTRODUCTION Serum neurofilament light chain (sNfL) is an emerging biomarker of neuronal damage in several neurological disorders. Its association with cognitive function in the general US population aged 60 years and above is unknown. The aim of this study was to investigate the correlation between sNfL and cognitive function in the general US population aged 60 and above. METHODS The data were obtained from the 2013-2014 National Health and Nutrition Examination Survey (NHANES), which include 506 individuals aged 60 or older who met our search criteria. In our study, sNfL levels were divided into two groups based on dichotomization (19.0 pg/mL). After adjusting for multiple covariates, it was found that the high sNfL group (≥ 19.0 pg/mL) had lower cognitive performance than the low sNfL group (< 19.0 pg/mL). This relationship was also stable in subgroup analysis. CONCLUSION In this sample of an American elderly population, higher sNfL levels are correlated with lower cognitive performance. Our findings suggest that sNfL may become a potential screening tool for early prediction and confirmation of cognitive damage.
Collapse
Affiliation(s)
- Yuanyuan Gao
- Affiliated Hospital Six of Nantong University, Yancheng Third People's Hospital, No. 75 Juchang Road, Yancheng, 224000, Jiangsu, China
- Yancheng Third People's Hospital, Yancheng, 224000, Jiangsu, China
| | - Dan Su
- Affiliated Hospital Six of Nantong University, Yancheng Third People's Hospital, No. 75 Juchang Road, Yancheng, 224000, Jiangsu, China
- Yancheng Third People's Hospital, Yancheng, 224000, Jiangsu, China
| | - Zhouya Xue
- The First People's Hospital of Yancheng, Yancheng, 224000, Jiangsu, China
| | - Lin Ji
- Affiliated Hospital Six of Nantong University, Yancheng Third People's Hospital, No. 75 Juchang Road, Yancheng, 224000, Jiangsu, China
- Yancheng Third People's Hospital, Yancheng, 224000, Jiangsu, China
| | - Shu Wang
- Affiliated Hospital Six of Nantong University, Yancheng Third People's Hospital, No. 75 Juchang Road, Yancheng, 224000, Jiangsu, China.
- Yancheng Third People's Hospital, Yancheng, 224000, Jiangsu, China.
| |
Collapse
|
46
|
Majeed J, Sabbagh MN, Kang MH, Lawrence JJ, Pruitt K, Bacus S, Reyna E, Brown M, Decourt B. Cancer drugs with high repositioning potential for Alzheimer's disease. Expert Opin Emerg Drugs 2023; 28:311-332. [PMID: 38100555 PMCID: PMC10877737 DOI: 10.1080/14728214.2023.2296079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/13/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION Despite the recent full FDA approval of lecanemab, there is currently no disease modifying therapy (DMT) that can efficiently slow down the progression of Alzheimer's disease (AD) in the general population. This statement emphasizes the need to identify novel DMTs in the shortest time possible to prevent a global epidemic of AD cases as the world population experiences an increase in lifespan. AREAS COVERED Here, we review several classes of anti-cancer drugs that have been or are being investigated in Phase II/III clinical trials for AD, including immunomodulatory drugs, RXR agonists, sex hormone therapies, tyrosine kinase inhibitors, and monoclonal antibodies. EXPERT OPINION Given the overall course of brain pathologies during the progression of AD, we express a great enthusiasm for the repositioning of anti-cancer drugs as possible AD DMTs. We anticipate an increasing number of combinatorial therapy strategies to tackle AD symptoms and their underlying pathologies. However, we strongly encourage improvements in clinical trial study designs to better assess target engagement and possible efficacy over sufficient periods of drug exposure.
Collapse
Affiliation(s)
- Jad Majeed
- University of Arizona Honors College, Tucson, Arizona, USA
| | - Marwan N. Sabbagh
- Alzheimer’s and Memory Disorders Division, Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Min H. Kang
- Department of Pediatrics, Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - J. Josh Lawrence
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Kevin Pruitt
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Ellie Reyna
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Maddy Brown
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Boris Decourt
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
- Roseman University of Health Sciences, Las Vegas, Nevada, USA
| |
Collapse
|
47
|
Doherty T, Yao Z, Khleifat AAL, Tantiangco H, Tamburin S, Albertyn C, Thakur L, Llewellyn DJ, Oxtoby NP, Lourida I, Ranson JM, Duce JA. Artificial intelligence for dementia drug discovery and trials optimization. Alzheimers Dement 2023; 19:5922-5933. [PMID: 37587767 DOI: 10.1002/alz.13428] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/26/2023] [Accepted: 07/05/2023] [Indexed: 08/18/2023]
Abstract
Drug discovery and clinical trial design for dementia have historically been challenging. In part these challenges have arisen from patient heterogeneity, length of disease course, and the tractability of a target for the brain. Applying big data analytics and machine learning tools for drug discovery and utilizing them to inform successful clinical trial design has the potential to accelerate progress. Opportunities arise at multiple stages in the therapy pipeline and the growing availability of large medical data sets opens possibilities for big data analyses to answer key questions in clinical and therapeutic challenges. However, before this goal is reached, several challenges need to be overcome and only a multi-disciplinary approach can promote data-driven decision-making to its full potential. Herein we review the current state of machine learning applications to clinical trial design and drug discovery, while presenting opportunities and recommendations that can break down the barriers to implementation.
Collapse
Affiliation(s)
- Thomas Doherty
- Eisai Europe Ltd, Hatfield, UK
- University of Westminster, London, UK
| | | | - Ahmad A L Khleifat
- Institute of Psychiatry, Psychology & Neuroscience, Department of Basic and Clinical Neuroscience, King's College London, London, UK
| | | | - Stefano Tamburin
- University of Verona, Department of Neurosciences, Biomedicine & Movement Sciences, Verona, Italy
| | - Chris Albertyn
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Lokendra Thakur
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David J Llewellyn
- University of Exeter Medical School, Exeter, UK
- Alan Turing Institute, London, UK
| | - Neil P Oxtoby
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | | | | | - James A Duce
- The ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
48
|
Gruia DC, Trender W, Hellyer P, Banerjee S, Kwan J, Zetterberg H, Hampshire A, Geranmayeh F. IC3 protocol: a longitudinal observational study of cognition after stroke using novel digital health technology. BMJ Open 2023; 13:e076653. [PMID: 38000822 PMCID: PMC10679983 DOI: 10.1136/bmjopen-2023-076653] [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: 06/15/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
INTRODUCTION Stroke is a major cause of death and disability worldwide, frequently resulting in persistent cognitive deficits among survivors. These deficits negatively impact recovery and therapy engagement, and their treatment is consistently rated as high priority by stakeholders and clinicians. Although clinical guidelines endorse cognitive screening for poststroke management, there is currently no gold-standard approach for identifying cognitive deficits after stroke, and clinical stroke services lack the capacity for long-term cognitive monitoring and care. Currently, available assessment tools are either not stroke-specific, not in-depth or lack scalability, leading to heterogeneity in patient assessments. METHODS AND ANALYSIS To address these challenges, a cost-effective, scalable and comprehensive screening tool is needed to provide a stroke-specific assessment of cognition. The current study presents such a novel digital tool, the Imperial Comprehensive Cognitive Assessment in Cerebrovascular Disease (IC3), designed to detect both domain-general and domain-specific cognitive deficits in patients after stroke with minimal input from a health professional. To ensure its reliability, we will use multiple validation approaches, and aim to recruit a large normative sample of age-matched, gender-matched and education-matched UK-based controls. Moreover, the IC3 assessment will be integrated within a larger prospective observational longitudinal clinical trial, where poststroke cognition will be examined in tandem with brain imaging and blood biomarkers to identify novel multimodal biomarkers of recovery after stroke. This study will enable deeper cognitive phenotyping of patients at a large scale, while identifying those with highest risk of progressive cognitive decline, as well as those with greatest potential for recovery. ETHICS AND DISSEMINATION This study has been approved by South West-Frenchay Research Ethics Committee (IRAS 299333) and authorised by the UK's Health Research Authority. Results from the study will be disseminated at conferences and within peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT05885295. Stage: Pre-results.
Collapse
Affiliation(s)
- Dragos-Cristian Gruia
- Department of Brain Sciences, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - William Trender
- Department of Brain Sciences, Imperial College London, London, UK
| | - Peter Hellyer
- Centre for Neuroimaging Sciences, IoPPN, King's College London, London, UK
| | - Soma Banerjee
- Department of Brain Sciences, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Joseph Kwan
- Department of Brain Sciences, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, Institute of Neurology, UCL, London, UK
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Goteborg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute, UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Adam Hampshire
- Department of Brain Sciences, Imperial College London, London, UK
| | - Fatemeh Geranmayeh
- Department of Brain Sciences, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
49
|
Cáceres C, Heusser B, Garnham A, Moczko E. The Major Hypotheses of Alzheimer's Disease: Related Nanotechnology-Based Approaches for Its Diagnosis and Treatment. Cells 2023; 12:2669. [PMID: 38067098 PMCID: PMC10705786 DOI: 10.3390/cells12232669] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/25/2023] [Accepted: 09/19/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease (AD) is a well-known chronic neurodegenerative disorder that leads to the progressive death of brain cells, resulting in memory loss and the loss of other critical body functions. In March 2019, one of the major pharmaceutical companies and its partners announced that currently, there is no drug to cure AD, and all clinical trials of the new ones have been cancelled, leaving many people without hope. However, despite the clear message and startling reality, the research continued. Finally, in the last two years, the Food and Drug Administration (FDA) approved the first-ever medications to treat Alzheimer's, aducanumab and lecanemab. Despite researchers' support of this decision, there are serious concerns about their effectiveness and safety. The validation of aducanumab by the Centers for Medicare and Medicaid Services is still pending, and lecanemab was authorized without considering data from the phase III trials. Furthermore, numerous reports suggest that patients have died when undergoing extended treatment. While there is evidence that aducanumab and lecanemab may provide some relief to those suffering from AD, their impact remains a topic of ongoing research and debate within the medical community. The fact is that even though there are considerable efforts regarding pharmacological treatment, no definitive cure for AD has been found yet. Nevertheless, it is strongly believed that modern nanotechnology holds promising solutions and effective clinical strategies for the development of diagnostic tools and treatments for AD. This review summarizes the major hallmarks of AD, its etiological mechanisms, and challenges. It explores existing diagnostic and therapeutic methods and the potential of nanotechnology-based approaches for recognizing and monitoring patients at risk of irreversible neuronal degeneration. Overall, it provides a broad overview for those interested in the evolving areas of clinical neuroscience, AD, and related nanotechnology. With further research and development, nanotechnology-based approaches may offer new solutions and hope for millions of people affected by this devastating disease.
Collapse
Affiliation(s)
| | | | | | - Ewa Moczko
- Facultad de Ingeniería y Ciencias, Universidad Adolfo Ibáñez, Viña del Mar 2562307, Chile; (C.C.)
| |
Collapse
|
50
|
Kang JH, Korecka M, Lee EB, Cousins KAQ, Tropea TF, Chen-Plotkin AA, Irwin DJ, Wolk D, Brylska M, Wan Y, Shaw LM. Alzheimer Disease Biomarkers: Moving from CSF to Plasma for Reliable Detection of Amyloid and tau Pathology. Clin Chem 2023; 69:1247-1259. [PMID: 37725909 PMCID: PMC10895336 DOI: 10.1093/clinchem/hvad139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/07/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Development of validated biomarkers to detect early Alzheimer disease (AD) neuropathology is needed for therapeutic AD trials. Abnormal concentrations of "core" AD biomarkers, cerebrospinal fluid (CSF) amyloid beta1-42, total tau, and phosphorylated tau correlate well with neuroimaging biomarkers and autopsy findings. Nevertheless, given the limitations of established CSF and neuroimaging biomarkers, accelerated development of blood-based AD biomarkers is underway. CONTENT Here we describe the clinical significance of CSF and plasma AD biomarkers to detect disease pathology throughout the Alzheimer continuum and correlate with imaging biomarkers. Use of the AT(N) classification by CSF and imaging biomarkers provides a more objective biologically based diagnosis of AD than clinical diagnosis alone. Significant progress in measuring CSF AD biomarkers using extensively validated highly automated assay systems has facilitated their transition from research use only to approved in vitro diagnostics tests for clinical use. We summarize development of plasma AD biomarkers as screening tools for enrollment and monitoring participants in therapeutic trials and ultimately in clinical care. Finally, we discuss the challenges for AD biomarkers use in clinical trials and precision medicine, emphasizing the possible ethnocultural differences in the levels of AD biomarkers. SUMMARY CSF AD biomarker measurements using fully automated analytical platforms is possible. Building on this experience, validated blood-based biomarker tests are being implemented on highly automated immunoassay and mass spectrometry platforms. The progress made developing analytically and clinically validated plasma AD biomarkers within the AT(N) classification scheme can accelerate use of AD biomarkers in therapeutic trials and routine clinical practice.
Collapse
Affiliation(s)
- Ju Hee Kang
- Department of Pharmacology and Clinical Pharmacology, Research Center for Controlling Intercellular Communication, Inha University, Incheon, South Korea
| | - Magdalena Korecka
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Katheryn A Q Cousins
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Thomas F Tropea
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alice A Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David Wolk
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Magdalena Brylska
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yang Wan
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|