1
|
Wang S, Xi J, Zhang M, Wang J. Identification of potential therapeutic targets for Alzheimer's disease from the proteomes of plasma and cerebrospinal fluid in a multicenter Mendelian randomization study. Int J Biol Macromol 2025; 294:139394. [PMID: 39755304 DOI: 10.1016/j.ijbiomac.2024.139394] [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: 09/19/2024] [Revised: 12/10/2024] [Accepted: 12/29/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Certain peripheral proteins are believed to be involved in the development of Alzheimer's disease (AD), but the roles of other new protein biomarkers are still unclear. Current treatments aim to manage symptoms, but they are not effective in stopping the progression of the disease. New drug targets are needed to prevent Alzheimer's disease. METHODS We used Mendelian randomization (MR) to study drug targets for Alzheimer's disease. We analyzed data from the European Alzheimer's and Dementia Biobank consortium and replicated our findings in GWAS data from IGAP and FinnGen cohorts. We identified genetic instruments for plasma and cerebrospinal fluid (CSF) proteins and conducted sensitivity analyses using various methods. Additionally, a comparison and analysis of protein-protein interactions (PPI) were conducted to identify potential causal proteins. The implications of these findings were further explored through an examination of existing AD drugs and their respective targets. RESULTS Through MR analysis, 10 protein AD pairs were identified as statistically significant at the Bonferroni level (P < 6.35 × 10-5). The specific findings indicate that elevated levels of plasma cathepsin H (CTSH) (OR = 1.06, 95%CI: 1.03-1.08, p = 6.12 × 10-6), plasma signal regulatory protein alpha (SIRPA) (OR = 1.03, 95%CI: 1.02-1.05, p = 1.37 × 10-5), plasma TMEM106B (OR = 1.16, 95%CI: 1.09-1.23, p = 1.92 × 10-6), and CSF bone sialoprotein (BSP) (OR = 1.33, 95%CI: 1.17-1.51, p = 9.34 × 10-6), CSF Interleukin-34 (IL-34) (OR = 2.13, 95%CI: 1.51-3.01, p = 1.85 × 10-5), CSF immunoglobulin-like transcript 2 (ILT-2) (OR = 1.33, 95%CI: 1.17-1.51, p = 9.34 × 10-6) are associated with an increased risk of AD, while increased levels of plasma progranulin gene (GRN) (OR = 0.79, 95%CI: 0.74-0.84, p = 2.19 × 10-12), plasma triggering receptor expressed on myeloid cells 2 (TREM2) (OR = 0.67, 95%CI: 0.58-0.78, p = 6.95 × 10-8), plasma sialic acid-bind immunoglobulin-like lectins (SIGLEC)-9 (OR = 0.67, 95%CI: 0.58-0.78, p = 6.95 × 10-8), and CSF SIGLEC7 (OR = 0.42, 95%CI: 0.28-0.64, p = 4.30 × 10-5) are associated with a decreased risk of AD. Bayesian colocalization found that the above protein-related genes shared the same mutation as AD. CONCLUSION Increased levels of plasma CTSH, SIRPA, TMEM106B, CSF BSP, CSF IL-34, and CSF ILT-2 have been found to be correlated with an elevated risk of AD, whereas elevated levels of plasma GRN, TREM2, SIGLEC9, and CSF SIGLEC7 are associated with a decreased risk of developing AD. Further investigation through clinical trials is needed.
Collapse
Affiliation(s)
- Shengnan Wang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China; Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianxin Xi
- Hepatobiliary and Pancreatic Surgery Department, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Mengyuan Zhang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jianglong Wang
- First Operating Room, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
2
|
Cheng Q, Fan Y, Zhang P, Liu H, Han J, Yu Q, Wang X, Wu S, Lu Z. Biomarkers of synaptic degeneration in Alzheimer's disease. Ageing Res Rev 2025; 104:102642. [PMID: 39701184 DOI: 10.1016/j.arr.2024.102642] [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: 03/27/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024]
Abstract
Synapse has been considered a critical neuronal structure in the procession of Alzheimer's disease (AD), attacked by two pathological molecule aggregates (amyloid-β and phosphorylated tau) in the brain, disturbing synaptic homeostasis before disease manifestation and subsequently causing synaptic degeneration. Recently, evidence has emerged indicating that soluble oligomeric amyloid-β (AβO) and tau exert direct toxicity on synapses, causing synaptic damage. Synaptic degeneration is closely linked to cognitive decline in AD, even in the asymptomatic stages of AD. Therefore, the identification of novel, specific, and sensitive biomarkers involved in synaptic degeneration holds significant promise for early diagnosis of AD, reducing synaptic degeneration and loss, and controlling the progression of AD. Currently, a range of biomarkers in cerebrospinal fluid (CSF), such as synaptosome-associated protein 25 (SNAP-25), synaptotagmin-1, growth-associated protein-43 (GAP-43), and neurogranin (Ng), along with functional brain imaging techniques, can detect variations in synaptic density, offering high sensitivity and specificity for AD diagnosis. However, these methods face challenges, including invasiveness, high cost, and limited accessibility. In contrast, biomarkers found in blood or urine provide a minimally invasive, cost-effective, and more accessible alternative to traditional diagnostic methods. Notably, neuron-derived exosomes in blood, which contain synaptic proteins, show variations in concentration that can serve as indicators of synaptic injury, providing an additional, less invasive approach to AD diagnosis and monitoring.
Collapse
Affiliation(s)
- Qian Cheng
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Yiou Fan
- Laboratory and Quality Management Department, Centers for Disease Control and Prevention of Shandong, Jinan, Shandong, China
| | - Pengfei Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Huan Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Jialin Han
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Qian Yu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xueying Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Shuang Wu
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Zhiming Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
3
|
Quan M. Timing of biomarker changes preceding Alzheimer's disease: for diagnosis, treatment or prevention? SCIENCE CHINA. LIFE SCIENCES 2025; 68:575-577. [PMID: 39422811 DOI: 10.1007/s11427-024-2733-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 09/15/2024] [Indexed: 10/19/2024]
Affiliation(s)
- Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing, 100053, China.
| |
Collapse
|
4
|
Zhou X, Xiao Z, Wu W, Chen Y, Yuan C, Leng Y, Yao Y, Zhao Q, Hofman A, Brunner E, Ding D. Closing the gap in dementia research by community-based cohort studies in the Chinese population. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2025; 55:101465. [PMID: 39902152 PMCID: PMC11788756 DOI: 10.1016/j.lanwpc.2025.101465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/20/2024] [Accepted: 01/02/2025] [Indexed: 02/05/2025]
Abstract
China accounts for 1/5 of the global population and China faces a particularly heavy dementia burden due to its rapidly ageing population. Unique historical events, genetic background, sociocultural factors, lifestyle, and the COVID-19 pandemic further influence cognitive outcomes in the Chinese population. We searched PubMed, Web of Science, and Embase for community-based cohort studies related to dementia in the Chinese population, and summarized the characteristics, methodologies, and major findings published over the last 25 years from 39 cohorts. We identified critical research gaps and propose future directions, including enhancing sample representativeness, investigating China-specific risk factors, expanding exposure measurements to the whole life-span, collecting objective data, conducting administer-friendly domain-specific cognitive assessments, adopting pathological diagnostic criteria, standardizing biobank construction, verifying multi-modal biomarkers, examining social and genetic-environmental aspects, and monitoring post-COVID cognitive health, to approach high quality of dementia studies that can provide solid evidence to policy making and promote global brain health research.
Collapse
Affiliation(s)
- Xiaowen Zhou
- Institute of Neurology, National Clinical Research Center for Aging and Medicine, National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhenxu Xiao
- Institute of Neurology, National Clinical Research Center for Aging and Medicine, National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Wanqing Wu
- Institute of Neurology, National Clinical Research Center for Aging and Medicine, National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
- Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
| | - Yuntao Chen
- Division of Psychiatry, Faculty of Brain Science, UCL, London, UK
| | - Changzheng Yuan
- School of Public Health, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
| | - Yue Leng
- Department of Psychiatry and Behavioural Sciences, University of California, San Francisco, USA
| | - Yao Yao
- China Center for Health Development Studies, School of Public Health, Peking University, Beijing, China
| | - Qianhua Zhao
- Institute of Neurology, National Clinical Research Center for Aging and Medicine, National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Albert Hofman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
| | - Eric Brunner
- Institute of Epidemiology and Health Care, UCL, London, UK
| | - Ding Ding
- Institute of Neurology, National Clinical Research Center for Aging and Medicine, National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Zhen M, Dang M, Cao Z, Xia X, Peng F, Wang S, Liu Y. Methylated cell-free DNA as a novel biomarker in Alzheimer's disease. Clin Chim Acta 2025; 566:120069. [PMID: 39622402 DOI: 10.1016/j.cca.2024.120069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/11/2024]
Abstract
Due to an aging population, Alzheimer's disease (AD), a neurodegenerative disorder, has affected more than 40 million people worldwide, a figure predicted to significantly increase in the coming decades. Despite much effort to understand AD pathogenesis, effective diagnosis and treatment remain a challenge. However, the development of liquid biopsy including the analysis of cell-free DNA (cfDNA) and methylation thereof has provided an alternative source of investigation to further explore the pathophysiology of AD. Herein, we discuss the research progress to date and highlight clinical applications of methylated cfDNA in AD.
Collapse
Affiliation(s)
- Mengyang Zhen
- Department of Clinical Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an, China; State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Miao Dang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Zexiang Cao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Xiaoying Xia
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Fan Peng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Siyuan Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Yang Liu
- Department of Clinical Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
6
|
Xin X, Liu Q, Jia S, Li S, Wang P, Wang X, Wang X. Correlation of muscle strength, information processing speed and cognitive function in the elderly with cognitive impairment--evidence from EEG. Front Aging Neurosci 2025; 17:1496725. [PMID: 39906715 PMCID: PMC11788344 DOI: 10.3389/fnagi.2025.1496725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 01/06/2025] [Indexed: 02/06/2025] Open
Abstract
Objective This study investigates the interplay between muscle strength, information processing speed, EEG-specific biomarkers, and cognitive function in elderly individuals with cognitive impairments, emphasizing the mediating roles of information processing speed and EEG-specific biomarkers. Method A cross-sectional study design was employed to recruit 151 elderly participants. The participants underwent grip strength and 30-s sit-to-stand tests to assess muscle strength, completed the Trail Making Test part A (TMT-A) and the Symbol Digit Modality Test (SDMT) to evaluate information processing speed, and utilized the Montreal Cognitive Assessment (MOCA) to gauge cognitive function. Additionally, EEG signals were recorded for 5 min to capture neural activity. Results The difference in information processing speed among elderly individuals with varying degrees of cognitive impairment was statistically significant (p < 0.001). A significant negative correlation was observed between the MoCA score and the time consumption of TMT-A (r = -0.402, p < 0.01), and a significant positive correlation was found between the MoCA score and the SDMT score (r = 0.609, p < 0.01). Grip strength was negatively correlated with the time consumption of TMT-A (r = -0.336, p < 0.01) and positively correlated with the SDMT score (r = 0.336, p < 0.01). A significant negative correlation was found between the 30-s sit-to-stand test and the time consumption of TMT-A (r = -0.273, p < 0.01), and a significant positive correlation was observed between the 30-s sit-to-stand test and the SDMT score (r = 0.372, p < 0.01). Additionally, we observed that the α1 power value indicators were significantly correlated with the MoCA score, the time consumption of TMT-A, and the SDMT score (all p < 0.01). The α1 power values at F7 + F8 and T5 + T6 were identified as sensitive EEG indicators for muscle strength and information processing speed. The EEG-specific indicators (B = 0.019, 95% CI: 0.003, 0.047) and information processing speed (B = 0.137, 95% CI: 0.096, 0.292) were found to partially mediate the relationship between grip strength and MoCA scores, with information processing speed exerting a stronger mediating effect. Conclusion Specific patterns were observed in the EEG of elderly individuals with cognitive impairments, which could objectively assess the risk of cognitive decline in this population. Muscle strength, information processing speed, and EEG-specific biomarkers were closely associated with cognitive function in elderly individuals. The potential pathway of interaction-muscle strength → EEG-specific biomarkers → information processing speed → cognitive function-provides valuable insights into advancing the field of cognitive research in the elderly.
Collapse
Affiliation(s)
- Xin Xin
- School of Physical Education, Shanghai University of Sport, Shanghai, China
| | - Qing Liu
- School of Physical Education, Shanghai University of Sport, Shanghai, China
| | - Shuqi Jia
- School of Physical Education, Shanghai University of Sport, Shanghai, China
| | - Shufan Li
- School of Physical Education, Shanghai University of Sport, Shanghai, China
| | - Peng Wang
- School of Physical Education, Shanghai University of Sport, Shanghai, China
| | - Xingze Wang
- School of Physical Education, Huzhou University, Huzhou, China
| | - Xing Wang
- School of Physical Education, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
7
|
Liu WS, You J, Chen SD, Zhang Y, Feng JF, Xu YM, Yu JT, Cheng W. Plasma proteomics identify biomarkers and undulating changes of brain aging. NATURE AGING 2025; 5:99-112. [PMID: 39653801 DOI: 10.1038/s43587-024-00753-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/17/2024] [Indexed: 12/15/2024]
Abstract
Proteomics enables the characterization of brain aging biomarkers and discernment of changes during brain aging. We leveraged multimodal brain imaging data from 10,949 healthy adults to estimate brain age gap (BAG), an indicator of brain aging. Proteome-wide association analysis across 4,696 participants of 2,922 proteins identified 13 significantly associated with BAG, implicating stress, regeneration and inflammation. Brevican (BCAN) (β = -0.838, P = 2.63 × 10-10) and growth differentiation factor 15 (β = 0.825, P = 3.48 × 10-11) showed the most significant, and multiple, associations with dementia, stroke and movement functions. Dysregulation of BCAN affected multiple cortical and subcortical structures. Mendelian randomization supported the causal association between BCAN and BAG. We revealed undulating changes in the plasma proteome across brain aging, and profiled brain age-related change peaks at 57, 70 and 78 years, implicating distinct biological pathways during brain aging. Our findings revealed the plasma proteomic landscape of brain aging and pinpointed biomarkers for brain disorders.
Collapse
Affiliation(s)
- Wei-Shi Liu
- Department of Neurology and National Center for Neurological diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia You
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Yu-Ming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Wei Cheng
- Department of Neurology and National Center for Neurological diseases, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
| |
Collapse
|
8
|
Hunter TR, Santos LE, Tovar-Moll F, De Felice FG. Alzheimer's disease biomarkers and their current use in clinical research and practice. Mol Psychiatry 2025; 30:272-284. [PMID: 39232196 DOI: 10.1038/s41380-024-02709-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/06/2024]
Abstract
While blood-based tests are readily available for various conditions, including cardiovascular diseases, type 2 diabetes, and common cancers, Alzheimer's disease (AD) and other neurodegenerative diseases lack an early blood-based screening test that can be used in primary care. Major efforts have been made towards the investigation of approaches that may lead to minimally invasive, cost-effective, and reliable tests capable of measuring brain pathological status. Here, we review past and current technologies developed to investigate biomarkers of AD, including novel blood-based approaches and the more established cerebrospinal fluid and neuroimaging biomarkers of disease. The utility of blood as a source of AD-related biomarkers in both clinical practice and interventional trials is discussed, supported by a comprehensive list of clinical trials for AD drugs and interventions that list biomarkers as primary or secondary endpoints. We highlight that identifying individuals in early preclinical AD using blood-based biomarkers will improve clinical trials and the optimization of therapeutic treatments as they become available. Lastly, we discuss challenges that remain in the field and address new approaches being developed, such as the examination of cargo packaged within extracellular vesicles of neuronal origin isolated from peripheral blood.
Collapse
Affiliation(s)
- Tai R Hunter
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Luis E Santos
- D'Or Institute for Research and Education, Rio de Janeiro, RJ, Brazil.
| | | | - Fernanda G De Felice
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
- D'Or Institute for Research and Education, Rio de Janeiro, RJ, Brazil.
- Centre for Neuroscience Studies and Department of Psychiatry, Queen's University, Kingston, ON, Canada.
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
9
|
Zhang F, Han X, Mu Q, Zailani H, Liu WC, Do QL, Wu Y, Wu N, Kang Y, Su L, Liu Y, Su KP, Wang F. Elevated cerebrospinal fluid biomarkers of neuroinflammation and neuronal damage in essential hypertension with secondary insomnia: Implications for Alzheimer's disease risk. Brain Behav Immun 2024; 125:158-167. [PMID: 39733863 DOI: 10.1016/j.bbi.2024.12.157] [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: 11/15/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 12/31/2024] Open
Abstract
Essential hypertension (EH) with secondary insomnia is associated with increased risks of neuroinflammation, neuronal damage, and Alzheimer's disease (AD). However, its relationship with specific cerebrospinal fluid (CSF) biomarkers of neuronal damage and neuroinflammation remains unclear. This case-control study compared CSF biomarker levels across three groups: healthy controls (HC, n = 64), hypertension-controlled (HTN-C, n = 54), and hypertension-uncontrolled (HTN-U, n = 107) groups, all EH participants experiencing secondary insomnia. CSF samples from knee replacement patients were analyzed for key biomarkers, and sleep quality was assessed via the Pittsburgh Sleep Quality Index (PSQI). Our findings showed that the HTN-U group had significantly higher CSF levels of proinflammatory cytokines IL-6, TNF-α, and IL-17 than the HC and HTN-C groups (all p < 0.01). These cytokines correlated positively with secondary insomnia measures, with IL-6 (r = 0.285, p = 0.003), IL-17 (r = 0.324, p = 0.001), and TNF-α (r = 0.274, p = 0.005) linked to PSQI scores. In the HTN-U group, elevated IL-6, TNF-α, and IL-17 levels were also positively associated with neurofilament light (NF-L) and negatively with β-amyloid 42 (Aβ42), both key AD markers (all p < 0.05). Additionally, secondary insomnia was negatively correlated with Aβ42 (r = -0.225, p = 0.021) and positively with NF-L (r = 0.261, p = 0.007). Higher CSF palmitic acid (PA) levels observed in the HTN-U group were linked to poorer sleep quality (r = 0.208, p = 0.033). In conclusion, EH with secondary insomnia is associated with CSF biomarkers of neuronal damage, neuroinflammation, and neurodegeneration, suggesting a potential increase in AD risk among this population.
Collapse
Affiliation(s)
- Feng Zhang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing 100096, China
| | - Xiaoli Han
- Clinical Nutrition Department, Friendship Hospital of Urumqi, Urumqi 830049, China
| | - Qingshuang Mu
- Xinjiang Key Laboratory of Neurological Disorder Research, the Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, China
| | - Halliru Zailani
- Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Nutrition, China Medical University, Taichung, Taiwan; Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | - Wen-Chun Liu
- Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; Department of Nursing, National Tainan Junior College of Nursing, Tainan, Taiwan
| | - Quang Le Do
- Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Nutrition, China Medical University, Taichung, Taiwan
| | - Yan Wu
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing 100096, China
| | - Nan Wu
- Institute of Polygenic Disease, Qiqihar Medical University, Qiqihar 161006, China
| | - Yimin Kang
- Medical Neurobiology Lab, Inner Mongolia Medical University, Huhhot 010110, China
| | - Lidong Su
- Medical Neurobiology Lab, Inner Mongolia Medical University, Baotou 014010, China
| | - Yanlong Liu
- School of Mental Health, Wenzhou Medical University, Wenzhou 325035, China.
| | - Kuan-Pin Su
- Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; College of Medicine, China Medical University, Taichung, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; An-Nan Hospital, China Medical University, Tainan, Taiwan.
| | - Fan Wang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing 100096, China.
| |
Collapse
|
10
|
Lin J, Liu X, Lin X, Liu N, Pei H, Zhao Y, Yu G, Wang W, Chen C, Hou T, Li X, Lin X, Li H. Effectiveness and Safety of Shenxiong Huanglian Detoxification Granule Combined with Donepezil for the Treatment of Alzheimer's Disease: Study Protocol for a Multicenter, Pragmatic, Randomized Controlled Clinical Trial. Int J Gen Med 2024; 17:6153-6164. [PMID: 39691835 PMCID: PMC11651136 DOI: 10.2147/ijgm.s485314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
Background Alzheimer's disease is a degenerative condition that causes patients to experience progressive memory decline and a significant decline in overall cognitive ability at any given moment. The increase in the elderly population has resulted in a notable surge in the prevalence of Alzheimer's disease, as has the global impact of the disease. Significant clinical efficacy of traditional Chinese medicine in combination with Western medicine for the treatment of Alzheimer's disease has been demonstrated in previous studies. The main purpose of this trial is to assess the effectiveness and safety of Shenxiong Huanglian Detoxification Granule combined with donepezil in individuals diagnosed with mild-to-moderate Alzheimer's disease. Methods This is a multicenter, pragmatic, randomized controlled trial. A total of 386 eligible individuals with mild to moderate Alzheimer's disease will receive random assignment and equal access to the test or control group. The effectiveness and safety of Shenxiong Huanglian Detoxification Granule in combination with donepezil will be observed. The primary outcome is the alteration in scores acquired from the Alzheimer's Disease Assessment Scale-Cognitive Subscale. Secondary outcomes include the assessments of the Traditional Chinese Medicine Syndrome score scale, Mini-Mental State Examination, Clinical Dementia Rating, and Activity of Daily Living scale. We will also analyze blood biomarkers of Alzheimer's disease, inflammatory indicators, oxidative stress indicators, and hemorheology indicators. In addition, safety assessments will be conducted at baseline, after 12 weeks, and after 24 weeks of treatment. Discussion These findings will offer reliable clinical evidence regarding the effectiveness and safety of Shenxiong Huanglian Detoxification Granule in combination with donepezil for treating patients with mild-to-moderate Alzheimer's disease. Additionally, this study will support the integration of traditional Chinese and Western medicine into mainstream treatment for Alzheimer's disease, promoting a multitarget strategy. Trial Registration Chinese Clinical Trial Registry, Registration Number: ChiCTR2300072768. Registered on 25 June 2023. https://www.chictr.org.cn/showproj.html?proj=195457.
Collapse
Affiliation(s)
- Jian Lin
- Department of Encephalopathy, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine: Guangzhou University of Chinese Medicine Third Clinical Medical College, Guangzhou, 510378, People’s Republic of China
| | - Xinghua Liu
- Department of Encephalopathy, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine: Guangzhou University of Chinese Medicine Third Clinical Medical College, Guangzhou, 510378, People’s Republic of China
| | - Xi Lin
- Department of Encephalopathy, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine: Guangzhou University of Chinese Medicine Third Clinical Medical College, Guangzhou, 510378, People’s Republic of China
| | - Nanyang Liu
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, People’s Republic of China
| | - Hui Pei
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, People’s Republic of China
| | - Yichun Zhao
- Department of Neurology, Wuxi Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214045, People’s Republic of China
| | - Guran Yu
- Department of Neurology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People’s Republic of China
| | - Wei Wang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, People’s Republic of China
| | - Chuan Chen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, People’s Republic of China
| | - Tingting Hou
- Department of Neurology, Shandong First Medical University Affiliated Provincial Hospital, Jinan, 250021, People’s Republic of China
| | - Xun Li
- Centre for Evidence-Based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Xingdong Lin
- Department of Encephalopathy, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine: Guangzhou University of Chinese Medicine Third Clinical Medical College, Guangzhou, 510378, People’s Republic of China
| | - Hao Li
- Department of Geriatrics, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, People’s Republic of China
| |
Collapse
|
11
|
Zhang R, Sun L, Jia AN, Wu SY, Wang Y, Wang SW, Guo Q, Meng YJ, Liang J, Huang W, Li YH, Wu J. Impact of ambient temperatures on Alzheimer's disease and other dementia mortality among elderly patients aged 60 years and older in China. ADVANCES IN CLIMATE CHANGE RESEARCH 2024. [DOI: 10.1016/j.accre.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
An C, Cai H, Ren Z, Fu X, Quan S, Jia L. Biofluid biomarkers for Alzheimer's disease: past, present, and future. MEDICAL REVIEW (2021) 2024; 4:467-491. [PMID: 39664082 PMCID: PMC11629312 DOI: 10.1515/mr-2023-0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 09/04/2024] [Indexed: 12/13/2024]
Abstract
Alzheimer's disease (AD) is a gradually progressive neurodegenerative disease with tremendous social and economic burden. Therefore, early and accurate diagnosis is imperative for effective treatment or prevention of the disease. Cerebrospinal fluid and blood biomarkers emerge as favorable diagnostic tools due to their relative accessibility and potential for widespread clinical use. This review focuses on the AT(N) biomarker system, which includes biomarkers reflecting AD core pathologies, amyloid deposition, and pathological tau, as well as neurodegeneration. Novel biomarkers associated with inflammation/immunity, synaptic dysfunction, vascular pathology, and α-synucleinopathy, which might contribute to either the pathogenesis or the clinical progression of AD, have also been discussed. Other emerging candidates including non-coding RNAs, metabolites, and extracellular vesicle-based markers have also enriched the biofluid biomarker landscape for AD. Moreover, the review discusses the current challenges of biofluid biomarkers in AD diagnosis and offers insights into the prospective future development.
Collapse
Affiliation(s)
- Chengyu An
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Huimin Cai
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Ziye Ren
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Xiaofeng Fu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shuiyue Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| |
Collapse
|
13
|
Cheng YW, Lin YJ, Lin YS, Hong WP, Kuan YC, Wu KY, Hsu JL, Wang PN, Pai MC, Chen CS, Fuh JL, Hu CJ, Chiu MJ. Application of blood-based biomarkers of Alzheimer's disease in clinical practice: Recommendations from Taiwan Dementia Society. J Formos Med Assoc 2024; 123:1210-1217. [PMID: 38296698 DOI: 10.1016/j.jfma.2024.01.018] [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: 05/11/2023] [Revised: 11/29/2023] [Accepted: 01/14/2024] [Indexed: 02/02/2024] Open
Abstract
Blood-based biomarkers (BBM) are potentially powerful tools that assist in the biological diagnosis of Alzheimer's disease (AD) in vivo with minimal invasiveness, relatively low cost, and good accessibility. This review summarizes current evidence for using BBMs in AD, focusing on amyloid, tau, and biomarkers for neurodegeneration. Blood-based phosphorylated tau and the Aβ42/Aβ40 ratio showed consistent concordance with brain pathology measured by CSF or PET in the research setting. In addition, glial fibrillary acidic protein (GFAP) and neurofilament light chain (NfL) are neurodegenerative biomarkers that show the potential to assist in the differential diagnosis of AD. Other pathology-specific biomarkers, such as α-synuclein and TAR DNA-binding protein 43 (TDP-43), can potentially detect AD concurrent pathology. Based on current evidence, the working group from the Taiwan Dementia Society (TDS) achieved consensus recommendations on the appropriate use of BBMs for AD in clinical practice. BBMs may assist clinical diagnosis and prognosis in AD subjects with cognitive symptoms; however, the results should be interpreted by dementia specialists and combining biochemical, neuropsychological, and neuroimaging information. Further studies are needed to evaluate BBMs' real-world performance and potential impact on clinical decision-making.
Collapse
Affiliation(s)
- Yu-Wen Cheng
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Ju Lin
- Department of Psychiatry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yung-Shuan Lin
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan; Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Pin Hong
- Department of Neurology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Yi-Chun Kuan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan; Department of Neurology and Dementia Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yi Wu
- Department of Psychiatry, Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Jung-Lung Hsu
- Department of Neurology, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital and Chang Gung University, New Taipei City, Taiwan; Graduate Institute of Mind, Brain, & Consciousness, Taipei Medical University, Taipei, Taiwan; Brain & Consciousness Research Center, Shuang Ho Hospital, New Taipei City, Taiwan
| | - Pei-Ning Wang
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Chyi Pai
- Division of Behavioral Neurology, Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Alzheimer's Disease Research Center, National Cheng Kung University Hospital, Tainan, Taiwan; Institute of Gerontology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Sheng Chen
- Department of Psychiatry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Psychiatry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jong-Ling Fuh
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chaur-Jong Hu
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan; Department of Neurology and Dementia Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
14
|
Merino-Serrais P, Soria JM, Arrabal CA, Ortigado-López A, Esparza MÁG, Muñoz A, Hernández F, Ávila J, DeFelipe J, León-Espinosa G. Protein tau phosphorylation in the proline rich region and its implication in the progression of Alzheimer's disease. Exp Neurol 2024; 383:115049. [PMID: 39522802 DOI: 10.1016/j.expneurol.2024.115049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/28/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Tau has a wide variety of essential functions in the brain, but this protein also plays a determining role in the development of Alzheimer's disease (AD) and other neurodegenerative diseases called tauopathies. This is due to its abnormal aggregation and the subsequent formation of neurofibrillary tangles. Tau hyperphosphorylation appears to be a critical step in its transformation into an aggregated protein. However, the exact process, including the cellular events that trigger it, remains unclear. In this study, we employed immunocytochemistry assays on hippocampal sections from AD cases and from tauopathy cases (Braak stage III) with no evidence of cognitive decline, and the P301S mouse model to investigate the colocalization patterns of Tau phosphorylated (p) at specific residues (S202-T205, S214, and T231) within the proline-rich region. Our results show pyramidal neurons in the hippocampus of P301S mice in which Tau is intensely phosphorylated at residues S202 and T205 (recognized by the AT8 antibody), but with no detectable phosphorylation at S214 or T231. These non-colocalizing neurons displayed intensely labeled aggregated pTau deposits distributed through the soma and dendritic processes. However, most of the hippocampal pyramidal neurons are labeled with pTauS214 or pTauT231 antibodies and typically showed a homogeneous and diffuse pTau distribution (not aggregated). This different labeling likely reflects a Tau conformational step, potentially related to the transition from a diffuse tau phosphorylation phenotype (Type 2) into an NFT-like or Type 1 phenotype. We further observed that dendrites of CA3 pyramidal cells are intensely labeled with pTau214 in the stratum lucidum, but not with AT8 or pTauT231. By contrast, analysis of tissue from AD patients or other human tauopathy cases (Braak stage III) with no evidence of cognitive decline revealed extensive colocalization with both antibody combinations in CA1. The complete or mature tangle development may follow a different mechanism in the P301S mouse model or may require more time to achieve the maturity state found in AD cases. Further studies would be necessary to address this question.
Collapse
Affiliation(s)
- Paula Merino-Serrais
- Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain; Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus Montegancedo S/N, Pozuelo de Alarcón, 28223 Madrid, Spain; CIBER de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - José Miguel Soria
- Department of Biomedical Sciences, Cardenal Herrera University-CEU Universities, 46001, Valencia, Spain
| | - Cristina Aguirre Arrabal
- Departamento de Matemática Aplicada y Estadística, Universidad San Pablo-CEU, CEU Universities, Julian Romea 22, 28003 Madrid, Spain
| | - Alfonso Ortigado-López
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Madrid, Spain
| | | | - Alberto Muñoz
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus Montegancedo S/N, Pozuelo de Alarcón, 28223 Madrid, Spain; Department of Cell Biology, Universidad Complutense de Madrid, Madrid, Spain
| | - Félix Hernández
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Jesús Ávila
- CIBER de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain; Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Javier DeFelipe
- Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain; Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus Montegancedo S/N, Pozuelo de Alarcón, 28223 Madrid, Spain; CIBER de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Gonzalo León-Espinosa
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus Montegancedo S/N, Pozuelo de Alarcón, 28223 Madrid, Spain; Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Madrid, Spain.
| |
Collapse
|
15
|
Kwon HS, Yu HJ, Koh SH. Revolutionizing Alzheimer's Diagnosis and Management: The Dawn of Biomarker-Based Precision Medicine. Dement Neurocogn Disord 2024; 23:188-201. [PMID: 39512700 PMCID: PMC11538857 DOI: 10.12779/dnd.2024.23.4.188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024] Open
Abstract
Alzheimer's disease (AD), a leading cause of dementia, presents a formidable global health challenge intensified by the aging population. This review encapsulates the evolving landscape of AD diagnosis and treatment with a special focus on the innovative role of fluid biomarkers. Pathologically, AD is marked by amyloid beta (Aβ) plaques and neurofibrillary tangles of hyperphosphorylated tau, which lead to synaptic dysfunction, neuronal loss, and cognitive decline. These pathological changes, commencing decades before symptom onset, underscore the need for early detection and intervention. Diagnosis traditionally relies on clinical assessment, neuropsychological testing, and neuroimaging techniques. However, fluid biomarkers in cerebrospinal fluid and blood, such as various forms of Aβ, total tau, phosphorylated tau, and neurofilament light chain, are emerging as less invasive, cost-effective diagnostic tools. These biomarkers are pivotal for early diagnosis, differential diagnosis, disease progression monitoring, and treatment response evaluation. The treatment landscape is shifting toward personalized medicine, highlighted by advancements in Aβ immunotherapies, such as lecanemab and donanemab. Demonstrating efficacy in phase III clinical trials, these therapies hold promise as tailored treatment strategies based on individual biomarker profiles. The integration of fluid biomarkers into clinical practice represents a significant advance in AD management, providing the potential for early and precise diagnosis, coupled with personalized therapeutic approaches. This heralds a new era in combating this debilitating disease.
Collapse
Affiliation(s)
- Hyuk Sung Kwon
- Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
| | - Hyun-Jung Yu
- Department of Neurology, Bundang Jesaeng General Hospital, Seongnam, Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Korea
| |
Collapse
|
16
|
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
|
17
|
Sessa F, Polito R, Li Rosi G, Salerno M, Esposito M, Pisanelli D, Ministeri F, Messina A, Carotenuto M, Chieffi S, Messina G, Monda M. Neurobiology and medico-legal aspects of suicides among older adults: a narrative review. Front Psychiatry 2024; 15:1449526. [PMID: 39290301 PMCID: PMC11405742 DOI: 10.3389/fpsyt.2024.1449526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
The task of preventing suicide in older adults is an important social burden as older adults aged above 65 are exposed to singular psychological aspects that increase suicide risks. Moreover, when an older adult corpse is found, the medico-legal inspection represents a fundamental tool to identify the exact cause of death, classifying or excluding it as suicide. In this scenario, this review aims to explore the neurobiological factors that could be related to suicidal behavior in older adults. A further goal of this review is the exploration of the medico-legal aspects surrounding older adult suicides, clarifying the importance of forensic investigation. Particularly, this review examines issues such as neurotransmitter imbalances, cognitive impairment, neuroinflammation, psychosocial factors related to geriatric suicide, and neurodegenerative diseases. Additionally, medico-legal aspects such as policy considerations, legal frameworks, mental health assessments, ethical implications and forensic investigation were explored. Considering the importance of this phenomenon, especially in western countries, a need has emerged for focused screening tools on suicidal behavior among older adults, in order to contain it. Therefore, this review makes an exhaustive appraisal of the literature giving insights into the delicate interplay between neurobiology as well as mental health in relation to older adult suicide within a medico-legal context. The comprehension of different aspects about this complex phenomenon is fundamental to propose new and more effective interventions, supporting tailored initiatives such as family support and improving healthcare, specifically towards vulnerable ageing societies to reduce older adult suicide risks.
Collapse
Affiliation(s)
- Francesco Sessa
- Department of Medical, Surgical and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| | - Rita Polito
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Giuseppe Li Rosi
- Department of Medical, Surgical and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| | - Monica Salerno
- Department of Medical, Surgical and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| | | | - Daniela Pisanelli
- Microbiology and Virology Unit, Ospedali Riuniti, Viale Luigi Pinto, Foggia, Italy
| | - Federica Ministeri
- Department of Medical, Surgical and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| | - Antonietta Messina
- Department of Precision Medicine, University of Campania, Luigi Vanvitelli, Napoli, Italy
| | - Marco Carotenuto
- Clinic of Child and Adolescent Neuropsychiatry, Department of Mental Health, Physical and Preventive Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Sergio Chieffi
- Department of Clinical Medicine, University of Campania, Luigi Vanvitelli, Napoli, Italy
| | - Giovanni Messina
- Department of Clinical Medicine, University of Campania, Luigi Vanvitelli, Napoli, Italy
| | - Marcellino Monda
- Department of Clinical Medicine, University of Campania, Luigi Vanvitelli, Napoli, Italy
| |
Collapse
|
18
|
Cyr B, Curiel Cid R, Loewenstein D, Vontell RT, Dietrich WD, Keane RW, de Rivero Vaccari JP. The Inflammasome Adaptor Protein ASC in Plasma as a Biomarker of Early Cognitive Changes. Int J Mol Sci 2024; 25:7758. [PMID: 39063000 PMCID: PMC11276719 DOI: 10.3390/ijms25147758] [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: 05/31/2024] [Revised: 07/13/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
Dementia is a group of symptoms including memory loss, language difficulties, and other types of cognitive and functional impairments that affects 57 million people worldwide, with the incidence expected to double by 2040. Therefore, there is an unmet need to develop reliable biomarkers to diagnose early brain impairments so that emerging interventions can be applied before brain degeneration. Here, we performed biomarker analyses for apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), and amyloid-β 42/40 (Aβ42/40) ratio in the plasma of older adults. Participants had blood drawn at baseline and underwent two annual clinical and cognitive evaluations. The groups tested either cognitively normal on both evaluations (NN), cognitively normal year 1 but cognitively impaired year 2 (NI), or cognitively impaired on both evaluations (II). ASC was elevated in the plasma of the NI group compared to the NN and II groups. Additionally, Aβ42 was increased in the plasma in the NI and II groups compared to the NN group. Importantly, the area under the curve (AUC) for ASC in participants older than 70 years old in NN vs. NI groups was 0.81, indicating that ASC is a promising plasma biomarker for early detection of cognitive decline.
Collapse
Affiliation(s)
- Brianna Cyr
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
| | - Rosie Curiel Cid
- Center for Cognitive Neuroscience and Aging, University of Miami, Miami, FL 33136, USA; (R.C.C.); (D.L.)
| | - David Loewenstein
- Center for Cognitive Neuroscience and Aging, University of Miami, Miami, FL 33136, USA; (R.C.C.); (D.L.)
| | | | - W. Dalton Dietrich
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
| | - Robert W. Keane
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
- Department of Physiology and Biophysics, University of Miami, Miami, FL 33136, USA
| | - Juan Pablo de Rivero Vaccari
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
- Center for Cognitive Neuroscience and Aging, University of Miami, Miami, FL 33136, USA; (R.C.C.); (D.L.)
- Department of Physiology and Biophysics, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
19
|
McInvale JJ, Canoll P, Hargus G. Induced pluripotent stem cell models as a tool to investigate and test fluid biomarkers in Alzheimer's disease and frontotemporal dementia. Brain Pathol 2024; 34:e13231. [PMID: 38246596 PMCID: PMC11189780 DOI: 10.1111/bpa.13231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/29/2023] [Indexed: 01/23/2024] Open
Abstract
Neurodegenerative diseases are increasing in prevalence and comprise a large socioeconomic burden on patients and their caretakers. The need for effective therapies and avenues for disease prevention and monitoring is of paramount importance. Fluid biomarkers for neurodegenerative diseases have gained a variety of uses, including informing participant selection for clinical trials, lending confidence to clinical diagnosis and disease staging, determining prognosis, and monitoring therapeutic response. Their role is expected to grow as disease-modifying therapies start to be available to a broader range of patients and as prevention strategies become established. Many of the underlying molecular mechanisms of currently used biomarkers are incompletely understood. Animal models and in vitro systems using cell lines have been extensively employed but face important translatability limitations. Induced pluripotent stem cell (iPSC) technology, where a theoretically unlimited range of cell types can be reprogrammed from peripheral cells sampled from patients or healthy individuals, has gained prominence over the last decade. It is a promising avenue to study physiological and pathological biomarker function and response to experimental therapeutics. Such systems are amenable to high-throughput drug screening or multiomics readouts such as transcriptomics, lipidomics, and proteomics for biomarker discovery, investigation, and validation. The present review describes the current state of biomarkers in the clinical context of neurodegenerative diseases, with a focus on Alzheimer's disease and frontotemporal dementia. We include a discussion of how iPSC models have been used to investigate and test biomarkers such as amyloid-β, phosphorylated tau, neurofilament light chain or complement proteins, and even nominate novel biomarkers. We discuss the limitations of current iPSC methods, mentioning alternatives such as coculture systems and three-dimensional organoids which address some of these concerns. Finally, we propose exciting prospects for stem cell transplantation paradigms using animal models as a preclinical tool to study biomarkers in the in vivo context.
Collapse
Affiliation(s)
- Julie J. McInvale
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia UniversityNew YorkNew YorkUSA
- Medical Scientist Training Program, Columbia UniversityNew YorkNew YorkUSA
| | - Peter Canoll
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
| | - Gunnar Hargus
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
20
|
Strobel J, Yousefzadeh-Nowshahr E, Deininger K, Bohn KP, von Arnim CAF, Otto M, Solbach C, Anderl-Straub S, Polivka D, Fissler P, Glatting G, Riepe MW, Higuchi M, Beer AJ, Ludolph A, Winter G. Exploratory Tau PET/CT with [11C]PBB3 in Patients with Suspected Alzheimer's Disease and Frontotemporal Lobar Degeneration: A Pilot Study on Correlation with PET Imaging and Cerebrospinal Fluid Biomarkers. Biomedicines 2024; 12:1460. [PMID: 39062033 PMCID: PMC11274645 DOI: 10.3390/biomedicines12071460] [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: 05/08/2024] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Accurately diagnosing Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD) is challenging due to overlapping symptoms and limitations of current imaging methods. This study investigates the use of [11C]PBB3 PET/CT imaging to visualize tau pathology and improve diagnostic accuracy. Given diagnostic challenges with symptoms and conventional imaging, [11C]PBB3 PET/CT's potential to enhance accuracy was investigated by correlating tau pathology with cerebrospinal fluid (CSF) biomarkers, positron emission tomography (PET), computed tomography (CT), amyloid-beta, and Mini-Mental State Examination (MMSE). We conducted [11C]PBB3 PET/CT imaging on 24 patients with suspected AD or FTLD, alongside [11C]PiB PET/CT (13 patients) and [18F]FDG PET/CT (15 patients). Visual and quantitative assessments of [11C]PBB3 uptake using standardized uptake value ratios (SUV-Rs) and correlation analyses with clinical assessments were performed. The scans revealed distinct tau accumulation patterns; 13 patients had no or faint uptake (PBB3-negative) and 11 had moderate to pronounced uptake (PBB3-positive). Significant inverse correlations were found between [11C]PBB3 SUV-Rs and MMSE scores, but not with CSF-tau or CSF-amyloid-beta levels. Here, we show that [11C]PBB3 PET/CT imaging can reveal distinct tau accumulation patterns and correlate these with cognitive impairment in neurodegenerative diseases. Our study demonstrates the potential of [11C]PBB3-PET imaging for visualizing tau pathology and assessing disease severity, offering a promising tool for enhancing diagnostic accuracy in AD and FTLD. Further research is essential to validate these findings and refine the use of tau-specific PET imaging in clinical practice, ultimately improving patient care and treatment outcomes.
Collapse
Affiliation(s)
- Joachim Strobel
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Katharina Deininger
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Karl Peter Bohn
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Markus Otto
- Department of Neurology, Halle University, 06120 Halle, Germany
| | - Christoph Solbach
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Dörte Polivka
- Department of Neurology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Patrick Fissler
- Psychiatric Services Thurgau (Academic Teaching Hospital of the University of Konstanz), 8596 Münsterlingen, Switzerland
| | - Gerhard Glatting
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Matthias W. Riepe
- Department of Psychiatry and Psychotherapy II, Ulm University, 89075 Ulm, Germany
| | - Makoto Higuchi
- National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Ambros J. Beer
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Albert Ludolph
- Department of Neurology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Gordon Winter
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| |
Collapse
|
21
|
Bonanni R, Cariati I, Cifelli P, Frank C, Annino G, Tancredi V, D'Arcangelo G. Exercise to Counteract Alzheimer's Disease: What Do Fluid Biomarkers Say? Int J Mol Sci 2024; 25:6951. [PMID: 39000060 PMCID: PMC11241657 DOI: 10.3390/ijms25136951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/14/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Neurodegenerative diseases (NDs) represent an unsolved problem to date with an ever-increasing population incidence. Particularly, Alzheimer's disease (AD) is the most widespread ND characterized by an accumulation of amyloid aggregates of beta-amyloid (Aβ) and Tau proteins that lead to neuronal death and subsequent cognitive decline. Although neuroimaging techniques are needed to diagnose AD, the investigation of biomarkers within body fluids could provide important information on neurodegeneration. Indeed, as there is no definitive solution for AD, the monitoring of these biomarkers is of strategic importance as they are useful for both diagnosing AD and assessing the progression of the neurodegenerative state. In this context, exercise is known to be an effective non-pharmacological management strategy for AD that can counteract cognitive decline and neurodegeneration. However, investigation of the concentration of fluid biomarkers in AD patients undergoing exercise protocols has led to unclear and often conflicting results, suggesting the need to clarify the role of exercise in modulating fluid biomarkers in AD. Therefore, this critical literature review aims to gather evidence on the main fluid biomarkers of AD and the modulatory effects of exercise to clarify the efficacy and usefulness of this non-pharmacological strategy in counteracting neurodegeneration in AD.
Collapse
Affiliation(s)
- Roberto Bonanni
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Ida Cariati
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Pierangelo Cifelli
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Claudio Frank
- UniCamillus-Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| | - Giuseppe Annino
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Sports Engineering Laboratory, Department of Industrial Engineering, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Virginia Tancredi
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Giovanna D'Arcangelo
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
| |
Collapse
|
22
|
Mantellatto Grigoli M, Pelegrini LNC, Whelan R, Cominetti MR. Present and Future of Blood-Based Biomarkers of Alzheimer's Disease: Beyond the Classics. Brain Res 2024; 1830:148812. [PMID: 38369085 DOI: 10.1016/j.brainres.2024.148812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/13/2023] [Accepted: 02/13/2024] [Indexed: 02/20/2024]
Abstract
The field of blood-based biomarkers for Alzheimer's disease (AD) has advanced at an incredible pace, especially after the development of sensitive analytic platforms that can facilitate large-scale screening. Such screening will be important when more sophisticated diagnostic methods are scarce and expensive. Thus, blood-based biomarkers can potentially reduce diagnosis inequities among populations from different socioeconomic contexts. This large-scale screening can be performed so that older adults at risk of cognitive decline assessed using these methods can then undergo more complete assessments with classic biomarkers, increasing diagnosis efficiency and reducing costs to the health systems. Blood-based biomarkers can also aid in assessing the effect of new disease-modifying treatments. This paper reviews recent advances in the area, focusing on the following leading candidates for blood-based biomarkers: amyloid-beta (Aβ), phosphorylated tau isoforms (p-tau), neurofilament light (NfL), and glial fibrillary acidic (GFAP) proteins, as well as on new candidates, Neuron-Derived Exosomes contents (NDEs) and Transactive response DNA-binding protein-43 (TDP-43), based on data from longitudinal observational cohort studies. The underlying challenges of validating and incorporating these biomarkers into routine clinical practice and primary care settings are also discussed. Importantly, challenges related to the underrepresentation of ethnic minorities and socioeconomically disadvantaged persons must be considered. If these challenges are overcome, a new time of cost-effective blood-based biomarkers for AD could represent the future of clinical procedures in the field and, together with continued prevention strategies, the beginning of an era with a lower incidence of dementia worldwide.
Collapse
Affiliation(s)
| | | | - Robert Whelan
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland; School of Psychology, Trinity College Dublin, Dublin, Ireland
| | - Marcia R Cominetti
- Department of Gerontology, Federal University of São Carlos, Brazil; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
23
|
Wang J, Xu L, Chen X, Wu J, Chen Y, Feng Z, Dong L, Yao D, Cai Q, Jian W, Li H, Duan M, Wang Z. Correlation Analysis of ApoB, ApoA1, and ApoB/ApoA1 with Cortical Morphology in Patients with Memory Complaints. J Alzheimers Dis 2024; 101:1137-1150. [PMID: 39302359 DOI: 10.3233/jad-230863] [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: 09/22/2024]
Abstract
Background Apolipoproteins and cortical morphology are closely associated with memory complaints, and both may contribute to the development of Alzheimer's disease. Objective To examine whether apolipoprotein B (ApoB), apolipoprotein A-1 (ApoA1), and their ratio (ApoB/ApoA1) are associated with cortical morphology in patients with memory complaints. Methods Ninety-seven patients underwent neuropsychological testing, measurements of ApoB, ApoA1, ApoB/ApoA1, plasma Alzheimer's biomarker, apolipoprotein E (ApoE) genotyping, and 3T structural magnetic resonance imaging (sMRI) scans. Based on sMRI scanning locations, patients were categorized into the University of Electronic Science and Technology (UESTC) and the Fourth People's Hospital of Chengdu (FPHC). The Computational Anatomy Toolbox within Statistical Parametric Mapping was used to calculate each patient's cortical morphology index based on sMRI data. The cortical morphology index and apolipoproteins were also analyzed. Results Significant positive correlations were found between ApoB and sulcal depth in the lateral occipital cortex among the UESTC, the FPHC, and the total sample groups, and negative correlations were observed between sulcal depth in the lateral occipital cortex and the scores of the Shape Trails Test Part A and B. In the FPHC group, the scores of the Montreal Cognitive Assessment Basic, delayed recall of the Auditory Verbal Learning Test, Animal Fluency Test and Boston Naming Test were positively correlated with the sulcal depth. Conclusions ApoB is associated with the sulcal depth in the lateral occipital cortex, potentially relating to speed/executive function in individuals with memory complaints.
Collapse
Affiliation(s)
- Jiayu Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
- Nursing School of Zunyi Medical University, Zunyi, China
| | - Lisi Xu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
| | - Xuemei Chen
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
| | - Jiajing Wu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
- Nursing School of Zunyi Medical University, Zunyi, China
| | - Yu Chen
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Radiology, the Fourth People's Hospital of Chengdu, Chengdu, China
| | - Ziqian Feng
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
- Nursing School of Zunyi Medical University, Zunyi, China
| | - Li Dong
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit of NeuroInformation, Chinese Academy of Medical Sciences, Chengdu, China
- Sichuan Institute for Brain Science and Brain-Inspired Intelligence, Chengdu, China
| | - Dezhong Yao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit of NeuroInformation, Chinese Academy of Medical Sciences, Chengdu, China
- Sichuan Institute for Brain Science and Brain-Inspired Intelligence, Chengdu, China
| | - Qingyan Cai
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
| | - Wei Jian
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
| | - Hongyi Li
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
| | - MingJun Duan
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
| | - Ziqi Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
| |
Collapse
|
24
|
Chen T, Hutchison RM, Rubel C, Murphy J, Xie J, Montenigro P, Cheng W, Fraser K, Dent G, Hendrix S, Hansson O, Aisen P, Tian Y, O'Gorman J. A Statistical Framework for Assessing the Relationship between Biomarkers and Clinical Endpoints in Alzheimer's Disease. J Prev Alzheimers Dis 2024; 11:1228-1240. [PMID: 39350368 PMCID: PMC11436399 DOI: 10.14283/jpad.2024.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/11/2024] [Indexed: 10/04/2024]
Abstract
Changes in biomarker levels of Alzheimer's disease (AD) reflect underlying pathophysiological changes in the brain and can provide evidence of direct and downstream treatment effects linked to disease modification. Recent results from clinical trials of anti-amyloid β (Aβ) treatments have raised the question of how to best characterize the relationship between AD biomarkers and clinical endpoints. Consensus methodology for assessing such relationships is lacking, leading to inconsistent evaluation and reporting. In this review, we provide a statistical framework for reporting treatment effects on early and late accelerating AD biomarkers and assessing their relationship with clinical endpoints at the subject and group levels. Amyloid positron emission tomography (PET), plasma p-tau, and tau PET follow specific trajectories during AD and are used as exemplar cases to contrast biomarkers with early and late progression. Subject-level correlation was assessed using change from baseline in biomarkers versus change from baseline in clinical endpoints, and interpretation of the correlation is dependent on the biomarker and disease stage. Group-level correlation was assessed using the placebo-adjusted treatment effects on biomarkers versus those on clinical endpoints in each trial. This correlation leverages the fundamental advantages of randomized placebo-controlled trials and assesses the predictivity of a treatment effect on a biomarker or clinical benefit. Harmonization in the assessment of treatment effects on biomarkers and their relationship to clinical endpoints will provide a wealth of comparable data across clinical trials and may yield new insights for the treatment of AD.
Collapse
Affiliation(s)
- T Chen
- Tianle Chen, Biogen Inc., 225 Binney St., Cambridge, MA 02142, Email address: , Phone: 617-914-7278
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|