1
|
Si Y, Lu W, Holloway S, Wang H, Tucci AA, Brucker A, Cheng Y, Wang LS, Schellenberger G, Lee WP, Tzeng JY. CNV-Profile Regression: A New Approach for Copy Number Variant Association Analysis in Whole Genome Sequencing Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.23.624994. [PMID: 39651129 PMCID: PMC11623527 DOI: 10.1101/2024.11.23.624994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Copy number variants (CNVs) are DNA gains or losses involving >50 base pairs. Assessing CNV effects on disease risk requires consideration of several factors. First, there are no natural definitions for CNV loci. Second, CNV effects can depend on dosage and length. Third, CNV effects can be more accurately estimated when all CNV events in a genomic region are analyzed together to assess their joint effects. We propose a new framework for association analysis that directly models an individual's entire CNV profile within a genomic region. This framework represents an individual's CNVs using a CNV profile curve to capture variations in CNV length and dosage and to bypass the need to predefine CNV loci. CNV effects are estimated at each genome position, making the results comparable across different studies. To jointly estimate the effects of all CNVs, we use a Lasso penalty to select CNVs associated with the trait and integrate a weighted L2-fusion penalty to encourage similar effects of adjacent CNVs when supported by the data. Simulations show that the proposed model can more effectively identify causal CNVs while maintaining false positive rates comparable to baseline methods and yield more precise effect-size estimates across different settings. When applied to CNV derived from whole genome sequencing data of the Alzheimer's Disease Sequencing Project, the proposed methods identify additional CNVs associated with Alzheimer's Disease (AD). These identified CNVs overlap with several known AD-risk genes and are significantly enriched by biological processes related to neuron structures and functions crucial in AD development.
Collapse
|
2
|
Jaye S, Sandau US, McFarland TJ, Woltjer RL, Saugstad JA. A clathrin mediated endocytosis scaffolding protein, Intersectin 1, changes in an isoform, brain region, and sex specific manner in Alzheimer's disease. Front Neurosci 2024; 18:1426180. [PMID: 38915309 PMCID: PMC11195150 DOI: 10.3389/fnins.2024.1426180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 05/28/2024] [Indexed: 06/26/2024] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by the accumulation of amyloid-beta (Aβ) plaques and neurofibrillary Tau tangles in the brain. We previously identified a set of candidate AD microRNAs (miRNAs) in human cerebrospinal fluid (CSF) and used a target prediction pipeline to identify mRNAs and pathways that could potentially be regulated by the miRNAs. Of these pathways, clathrin mediated endocytosis (CME) was selected for further investigation. CME is altered in multiple brain cell types in AD and is implicated in early cellular phenotypes such as enlarged early endosomes and pathogenic processing of Aβ. However, a comprehensive evaluation of major CME hub proteins in humans with AD across multiple brain regions is lacking. Thus, we used immunoblots to evaluate human post-mortem AD and control (CTL) frontal cortex (FC; AD n = 22, CTL n = 23) and hippocampus (HP; AD n = 34, CTL n = 22) for changes in Intersectin 1 (ITSN1), Phosphatidylinositol Binding Clathrin Assembly Protein gene (PICALM), Clathrin Light Chain (CLT), FCH and Mu Domain Containing Endocytic Adaptor 1 (FCHO1), Adaptor Related Protein Complex 2 (AP2) Subunit Alpha 1 (AP2A1), and Dynamin 2 (DNM2). Of these, we found that in AD, ITSN1-long (ITSN1-L) was decreased in the FC of males and HP of females, while ITSN1-short was increased in the HP of both males and females. We further evaluated ITSN1-L levels in cortex (CTX) and HP of the 5xFAD mouse model of Aβ pathology at different timepoints during aging and disease progression by immunoblot (n = 5-8 per group). At 3 months, female 5xFAD exhibited an increase of ITSN1-L in CTX but a decrease at 6 and 9 months. Additionally, immunofluorescent staining of 5xFAD primary HP neurons showed an increase of ITSN1-L in matured 5xFAD neurons at 21 and 28 days in vitro. Together, our studies show that in AD, isoforms of ITSN1 change in a brain region-and sex-dependent manner. Further, changes in ITSN1-L are transient with levels increasing during early Aβ accumulation and decreasing during later progression. These findings suggest that ITSN1 expression, and consequently CME activity, may change depending on the stage of disease progression.
Collapse
Affiliation(s)
- Sierra Jaye
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Ursula S. Sandau
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Trevor J. McFarland
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Randy L. Woltjer
- Division of Neuropathology, Department of Pathology, Oregon Health and Science University, Portland, OR, United States
| | - Julie A. Saugstad
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
3
|
Jaye S, Sandau US, Saugstad JA. Clathrin mediated endocytosis in Alzheimer's disease: cell type specific involvement in amyloid beta pathology. Front Aging Neurosci 2024; 16:1378576. [PMID: 38694257 PMCID: PMC11061891 DOI: 10.3389/fnagi.2024.1378576] [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: 01/29/2024] [Accepted: 04/03/2024] [Indexed: 05/04/2024] Open
Abstract
This review provides a comprehensive examination of the role of clathrin-mediated endocytosis (CME) in Alzheimer's disease (AD) pathogenesis, emphasizing its impact across various cellular contexts beyond neuronal dysfunction. In neurons, dysregulated CME contributes to synaptic dysfunction, amyloid beta (Aβ) processing, and Tau pathology, highlighting its involvement in early AD pathogenesis. Furthermore, CME alterations extend to non-neuronal cell types, including astrocytes and microglia, which play crucial roles in Aβ clearance and neuroinflammation. Dysregulated CME in these cells underscores its broader implications in AD pathophysiology. Despite significant progress, further research is needed to elucidate the precise mechanisms underlying CME dysregulation in AD and its therapeutic implications. Overall, understanding the complex interplay between CME and AD across diverse cell types holds promise for identifying novel therapeutic targets and interventions.
Collapse
Affiliation(s)
| | | | - Julie A. Saugstad
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
4
|
Tzavellas NP, Tsamis KI, Katsenos AP, Davri AS, Simos YV, Nikas IP, Bellos S, Lekkas P, Kanellos FS, Konitsiotis S, Labrakakis C, Vezyraki P, Peschos D. Firing Alterations of Neurons in Alzheimer's Disease: Are They Merely a Consequence of Pathogenesis or a Pivotal Component of Disease Progression? Cells 2024; 13:434. [PMID: 38474398 DOI: 10.3390/cells13050434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder, yet its underlying causes remain elusive. The conventional perspective on disease pathogenesis attributes alterations in neuronal excitability to molecular changes resulting in synaptic dysfunction. Early hyperexcitability is succeeded by a progressive cessation of electrical activity in neurons, with amyloid beta (Aβ) oligomers and tau protein hyperphosphorylation identified as the initial events leading to hyperactivity. In addition to these key proteins, voltage-gated sodium and potassium channels play a decisive role in the altered electrical properties of neurons in AD. Impaired synaptic function and reduced neuronal plasticity contribute to a vicious cycle, resulting in a reduction in the number of synapses and synaptic proteins, impacting their transportation inside the neuron. An understanding of these neurophysiological alterations, combined with abnormalities in the morphology of brain cells, emerges as a crucial avenue for new treatment investigations. This review aims to delve into the detailed exploration of electrical neuronal alterations observed in different AD models affecting single neurons and neuronal networks.
Collapse
Affiliation(s)
- Nikolaos P Tzavellas
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Konstantinos I Tsamis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, 455 00 Ioannina, Greece
| | - Andreas P Katsenos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Athena S Davri
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Yannis V Simos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Ilias P Nikas
- Medical School, University of Cyprus, 2029 Nicosia, Cyprus
| | - Stefanos Bellos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Panagiotis Lekkas
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Foivos S Kanellos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Spyridon Konitsiotis
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, 455 00 Ioannina, Greece
| | - Charalampos Labrakakis
- Department of Biological Applications and Technology, University of Ioannina, 451 10 Ioannina, Greece
| | - Patra Vezyraki
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Dimitrios Peschos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| |
Collapse
|
5
|
Lin NH, Goh A, Lin SH, Chuang KA, Chang CH, Li MH, Lu CH, Chen WY, Wei PH, Pan IH, Perng MD, Wen SF. Neuroprotective Effects of a Multi-Herbal Extract on Axonal and Synaptic Disruption in Vitro and Cognitive Impairment in Vivo. J Alzheimers Dis Rep 2023; 7:51-76. [PMID: 36777330 PMCID: PMC9912829 DOI: 10.3233/adr-220056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
Background Alzheimer's disease (AD) is a multifactorial disorder characterized by cognitive decline. Current available therapeutics for AD have limited clinical benefit. Therefore, preventive therapies for interrupting the development of AD are critically needed. Molecules targeting multifunction to interact with various pathlogical components have been considered to improve the therapeutic efficiency of AD. In particular, herbal medicines with multiplicity of actions produce cognitive benefits on AD. Bugu-M is a multi-herbal extract composed of Ganoderma lucidum (Antler form), Nelumbo nucifera Gaertn., Ziziphus jujuba Mill., and Dimocarpus longan, with the ability of its various components to confer resilience to cognitive deficits. Objective To evaluate the potential of Bugu-M on amyloid-β (Aβ) toxicity and its in vitro mechanisms and on in vivo cognitive function. Methods We illustrated the effect of Bugu-M on Aβ25-35-evoked toxicity as well as its possible mechanisms to diminish the pathogenesis of AD in rat cortical neurons. For cognitive function studies, 2-month-old female 3×Tg-AD mice were administered 400 mg/kg Bugu-M for 30 days. Behavioral tests were performed to assess the efficacy of Bugu-M on cognitive impairment. Results In primary cortical neuronal cultures, Bugu-M mitigated Aβ-evoked toxicity by reducing cytoskeletal aberrations and axonal disruption, restoring presynaptic and postsynaptic protein expression, suppressing mitochondrial damage and apoptotic signaling, and reserving neurogenic and neurotrophic factors. Importantly, 30-day administration of Bugu-M effectively prevented development of cognitive impairment in 3-month-old female 3×Tg-AD mice. Conclusion Bugu-M might be beneficial in delaying the progression of AD, and thus warrants consideration for its preventive potential for AD.
Collapse
Affiliation(s)
- Ni-Hsuan Lin
- Institute of Molecular Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Angela Goh
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Shyh-Horng Lin
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Kai-An Chuang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chih-Hsuan Chang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Ming-Han Li
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chu-Hsun Lu
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Wen-Yin Chen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Pei-Hsuan Wei
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - I-Hong Pan
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Ming-Der Perng
- Institute of Molecular Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan,
School of Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan,Correspondence to: Shu-Fang Wen, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, 321, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan. Tel.: +886 35743946; E-mail: and Ming-Der Perng, College of Life Sciences, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan. Tel.: +886 35742024; E-mail:
| | - Shu-Fang Wen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan,Correspondence to: Shu-Fang Wen, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, 321, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan. Tel.: +886 35743946; E-mail: and Ming-Der Perng, College of Life Sciences, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan. Tel.: +886 35742024; E-mail:
| |
Collapse
|
6
|
Moore JA, Tuladhar A, Ismail Z, Mouches P, Wilms M, Forkert ND. Dementia in Convolutional Neural Networks: Using Deep Learning Models to Simulate Neurodegeneration of the Visual System. Neuroinformatics 2023; 21:45-55. [PMID: 36083416 DOI: 10.1007/s12021-022-09602-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2022] [Indexed: 11/27/2022]
Abstract
Although current research aims to improve deep learning networks by applying knowledge about the healthy human brain and vice versa, the potential of using such networks to model and study neurodegenerative diseases remains largely unexplored. In this work, we present an in-depth feasibility study modeling progressive dementia in silico with deep convolutional neural networks. Therefore, networks were trained to perform visual object recognition and then progressively injured by applying neuronal as well as synaptic injury. After each iteration of injury, network object recognition accuracy, saliency map similarity between the intact and injured networks, and internal activations of the degenerating models were evaluated. The evaluation revealed that cognitive function of the network progressively decreased with increasing injury load whereas this effect was much more pronounced for synaptic damage. The effects of neurodegeneration found for the in silico model are especially similar to the loss of visual cognition seen in patients with posterior cortical atrophy.
Collapse
Affiliation(s)
- Jasmine A Moore
- Department of Radiology, University of Calgary, Calgary, AB, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
- Biomedical Engineering Program, University of Calgary, Calgary, AB, Canada.
| | - Anup Tuladhar
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Zahinoor Ismail
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada
- O'Brien Institute for Public Health, University of Calgary, Calgary, AB, Canada
| | - Pauline Mouches
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Program, University of Calgary, Calgary, AB, Canada
| | - Matthias Wilms
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Nils D Forkert
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Department of Electrical and Software Engineering, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
7
|
Lv N, Wang Y, Liu Y, Tang J, Lei Q, Wang Y, Wei H. Decreased Microglia in Pax2 Mutant Mice Leads to Impaired Learning and Memory. ACS Chem Neurosci 2022; 13:2490-2502. [PMID: 35929805 DOI: 10.1021/acschemneuro.2c00352] [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: 01/20/2023] Open
Abstract
Impaired learning and memory ability is one of the characteristics of a variety of neurological diseases, and its molecular mechanisms are complex and diverse and are regulated by a variety of factors. It is generally believed that synaptic plasticity plays an important role in the process of learning and memory. The protein encoded by the Pax2 gene is a transcription factor involved in neuron migration and cell fate determination during neural development. Mice knocked out of BDNF in the Pax2 lineage-derived interneuron precursor exhibited learning disabilities and severe cognitive impairment. In this study, Pax2 heterozygous gene (Pax2+/- mice) deletion mice were used as the research objects and behavioral tests were used to observe the effect of Pax2 gene deletion on learning and memory ability; morphological and molecular biological methods were used to observe the effect of Pax2 gene deletion on the neural structure. Single-cell transcriptome sequencing was used to observe the cell subtypes and differentially expressed genes (DEGs) and signaling pathways affected by Pax2 gene deletion and the possible molecular mechanisms. The results showed that Pax2+/- mice had impaired learning and memory ability, abnormal synaptic structure, and significantly reduced number of microglia clusters, and DEGs were associated with pro-inflammatory chemokines. Finally, we speculate that Pax2 gene deletion may lead to abnormal chemokines and chemokine receptors by affecting microglia.
Collapse
Affiliation(s)
- Na Lv
- Department of Neurology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China.,Department of Physiology, School of Basic Medicine, Shanxi Medical University, Taiyuan 030012, China.,Shanxi Key Laboratory of Brain Disease Control, Shanxi Provincial People's Hospital, Taiyuan 030012, China
| | - Ying Wang
- Department of Neurology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China.,Shanxi Key Laboratory of Brain Disease Control, Shanxi Provincial People's Hospital, Taiyuan 030012, China
| | - Yongfeng Liu
- Department of Neurology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China
| | - Jiaming Tang
- Department of Neurology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China.,Graduate College, Shanxi University of Chinese Medicine, Taiyuan 030024, China
| | - Qiang Lei
- Department of Neurology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China.,Department of Physiology, School of Basic Medicine, Shanxi Medical University, Taiyuan 030012, China.,Shanxi Key Laboratory of Brain Disease Control, Shanxi Provincial People's Hospital, Taiyuan 030012, China
| | - Yizhuo Wang
- Department of Neurology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China.,Shanxi Key Laboratory of Brain Disease Control, Shanxi Provincial People's Hospital, Taiyuan 030012, China
| | - Hongen Wei
- Department of Neurology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China.,Shanxi Key Laboratory of Brain Disease Control, Shanxi Provincial People's Hospital, Taiyuan 030012, China
| |
Collapse
|
8
|
Podvin S, Jiang Z, Boyarko B, Rossitto LA, O’Donoghue A, Rissman RA, Hook V. Dysregulation of Neuropeptide and Tau Peptide Signatures in Human Alzheimer's Disease Brain. ACS Chem Neurosci 2022; 13:1992-2005. [PMID: 35758417 PMCID: PMC9264367 DOI: 10.1021/acschemneuro.2c00222] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Synaptic dysfunction and loss occur in Alzheimer's disease (AD) brains, which results in cognitive deficits and brain neurodegeneration. Neuropeptides comprise the major group of synaptic neurotransmitters in the nervous system. This study evaluated neuropeptide signatures that are hypothesized to differ in human AD brain compared to age-matched controls, achieved by global neuropeptidomics analysis of human brain cortex synaptosomes. Neuropeptidomics demonstrated distinct profiles of neuropeptides in AD compared to controls consisting of neuropeptides derived from chromogranin A (CHGA) and granins, VGF (nerve growth factor inducible), cholecystokinin, and others. The differential neuropeptide signatures indicated differences in proteolytic processing of their proneuropeptides. Analysis of cleavage sites showed that dibasic residues at the N-termini and C-termini of neuropeptides were the main sites for proneuropeptide processing, and data also showed that the AD group displayed differences in preferred residues adjacent to the cleavage sites. Notably, tau peptide signatures differed in the AD compared to age-matched control human brain cortex synaptosomes. Unique tau peptides were derived from the tau protein through proteolysis using similar and differential cleavage sites in the AD brain cortex compared to the control. Protease profiles differed in the AD compared to control, indicated by proteomics data. Overall, these results demonstrate that dysregulation of neuropeptides and tau peptides occurs in AD brain cortex synaptosomes compared to age-matched controls, involving differential cleavage site properties for proteolytic processing of precursor proteins. These dynamic changes in neuropeptides and tau peptide signatures may be associated with the severe cognitive deficits of AD.
Collapse
Affiliation(s)
- Sonia Podvin
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - Zhenze Jiang
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - Ben Boyarko
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - Leigh-Ana Rossitto
- Biomedical
Sciences Graduate Program, University of
California, San Diego, La Jolla, California 92093, United States
| | - Anthony O’Donoghue
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - Robert A. Rissman
- Department
of Neurosciences, University of California
San Diego, La Jolla, California 92093, United States
- Veterans
Affairs San Diego Health System, La Jolla, California 92093, United States
| | - Vivian Hook
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
- Biomedical
Sciences Graduate Program, University of
California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
9
|
Gupta C, Xu J, Jin T, Khullar S, Liu X, Alatkar S, Cheng F, Wang D. Single-cell network biology characterizes cell type gene regulation for drug repurposing and phenotype prediction in Alzheimer's disease. PLoS Comput Biol 2022; 18:e1010287. [PMID: 35849618 PMCID: PMC9333448 DOI: 10.1371/journal.pcbi.1010287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/28/2022] [Accepted: 06/07/2022] [Indexed: 12/03/2022] Open
Abstract
Dysregulation of gene expression in Alzheimer's disease (AD) remains elusive, especially at the cell type level. Gene regulatory network, a key molecular mechanism linking transcription factors (TFs) and regulatory elements to govern gene expression, can change across cell types in the human brain and thus serve as a model for studying gene dysregulation in AD. However, AD-induced regulatory changes across brain cell types remains uncharted. To address this, we integrated single-cell multi-omics datasets to predict the gene regulatory networks of four major cell types, excitatory and inhibitory neurons, microglia and oligodendrocytes, in control and AD brains. Importantly, we analyzed and compared the structural and topological features of networks across cell types and examined changes in AD. Our analysis shows that hub TFs are largely common across cell types and AD-related changes are relatively more prominent in some cell types (e.g., microglia). The regulatory logics of enriched network motifs (e.g., feed-forward loops) further uncover cell type-specific TF-TF cooperativities in gene regulation. The cell type networks are also highly modular and several network modules with cell-type-specific expression changes in AD pathology are enriched with AD-risk genes. The further disease-module-drug association analysis suggests cell-type candidate drugs and their potential target genes. Finally, our network-based machine learning analysis systematically prioritized cell type risk genes likely involved in AD. Our strategy is validated using an independent dataset which showed that top ranked genes can predict clinical phenotypes (e.g., cognitive impairment) of AD with reasonable accuracy. Overall, this single-cell network biology analysis provides a comprehensive map linking genes, regulatory networks, cell types and drug targets and reveals cell-type gene dysregulation in AD.
Collapse
Affiliation(s)
- Chirag Gupta
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jielin Xu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Ting Jin
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Saniya Khullar
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Xiaoyu Liu
- Department of Statistics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Sayali Alatkar
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Daifeng Wang
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
10
|
Chino K, Izuo N, Noike H, Uno K, Kuboyama T, Tohda C, Muramatsu SI, Nitta A. Shati/Nat8l Overexpression Improves Cognitive Decline by Upregulating Neuronal Trophic Factor in Alzheimer's Disease Model Mice. Neurochem Res 2022; 47:2805-2814. [PMID: 35759136 DOI: 10.1007/s11064-022-03649-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/11/2022] [Accepted: 06/01/2022] [Indexed: 11/25/2022]
Abstract
Alzheimer's disease (AD) is a type of dementia characterized by the deposition of amyloid β, a causative protein of AD, in the brain. Shati/Nat8l, identified as a psychiatric disease related molecule, is a responsive enzyme of N-acetylaspartate (NAA) synthesis. In the hippocampi of AD patients and model mice, the NAA content and Shati/Nat8l expression were reported to be reduced. Having recently clarified the involvement of Shati/Nat8l in cognitive function, we examined the recovery effect of the hippocampal overexpression of Shati/Nat8l in AD model mice (5XFAD). Shati/Nat8l overexpression suppressed cognitive dysfunction without affecting the Aβ burden or number of NeuN-positive neurons. In addition, brain-derived neurotrophic factor mRNA was upregulated by Shati/Nat8l overexpression in 5XFAD mice. These results suggest that Shati/Nat8l overexpression prevents cognitive dysfunction in 5XFAD mice, indicating that Shati/Nat8l could be a therapeutic target for AD.
Collapse
Affiliation(s)
- Kakeru Chino
- Department of Pharmaceutical Therapy and Neuropharmacology, School of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Naotaka Izuo
- Department of Pharmaceutical Therapy and Neuropharmacology, School of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Hiroshi Noike
- Department of Pharmaceutical Therapy and Neuropharmacology, School of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Kyosuke Uno
- Department of Pharmaceutical Therapy and Neuropharmacology, School of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
- Laboratory of Molecular Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata-shi, Osaka, Japan
| | - Tomoharu Kuboyama
- Laboratory of Pharmacognosy, Daiichi University of Pharmacy, 22-1 Tamagawa-cho, Minami-ku, Fukuoka, 815-8511, Japan
| | - Chihiro Tohda
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, Sugitani 2630, Toyama, 930-0194, Japan
| | - Shin-Ichi Muramatsu
- Division of Neurological Gene Therapy, Open Innovation Center, Jichi Medical University, Shimotsuke, 329-0498, Japan
- Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Atsumi Nitta
- Department of Pharmaceutical Therapy and Neuropharmacology, School of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| |
Collapse
|
11
|
Bashir S, Uzair M, Abualait T, Arshad M, Khallaf RA, Niaz A, Thani Z, Yoo WK, Túnez I, Demirtas-Tatlidede A, Meo SA. Effects of transcranial magnetic stimulation on neurobiological changes in Alzheimer's disease (Review). Mol Med Rep 2022; 25:109. [PMID: 35119081 PMCID: PMC8845030 DOI: 10.3892/mmr.2022.12625] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/15/2021] [Indexed: 11/05/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline and brain neuronal loss. A pioneering field of research in AD is brain stimulation via electromagnetic fields (EMFs), which may produce clinical benefits. Noninvasive brain stimulation techniques, such as transcranial magnetic stimulation (TMS), have been developed to treat neurological and psychiatric disorders. The purpose of the present review is to identify neurobiological changes, including inflammatory, neurodegenerative, apoptotic, neuroprotective and genetic changes, which are associated with repetitive TMS (rTMS) treatment in patients with AD. Furthermore, it aims to evaluate the effect of TMS treatment in patients with AD and to identify the associated mechanisms. The present review highlights the changes in inflammatory and apoptotic mechanisms, mitochondrial enzymatic activities, and modulation of gene expression (microRNA expression profiles) associated with rTMS or sham procedures. At the molecular level, it has been suggested that EMFs generated by TMS may affect the cell redox status and amyloidogenic processes. TMS may also modulate gene expression by acting on both transcriptional and post‑transcriptional regulatory mechanisms. TMS may increase brain cortical excitability, induce specific potentiation phenomena, and promote synaptic plasticity and recovery of impaired functions; thus, it may re‑establish cognitive performance in patients with AD.
Collapse
Affiliation(s)
- Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Eastern Province 32253, Saudi Arabia
| | - Mohammad Uzair
- Department of Biological Sciences, Faculty of Basic and Applied Sciences, International Islamic University Islamabad, Islamabad 44000, Pakistan
| | - Turki Abualait
- College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province 34212, Saudi Arabia
| | - Muhammad Arshad
- Department of Biological Sciences, Faculty of Basic and Applied Sciences, International Islamic University Islamabad, Islamabad 44000, Pakistan
| | - Roaa A. Khallaf
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Eastern Province 32253, Saudi Arabia
| | - Asim Niaz
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Eastern Province 32253, Saudi Arabia
| | - Ziyad Thani
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Eastern Province 32253, Saudi Arabia
| | - Woo-Kyoung Yoo
- Department of Physical Medicine and Rehabilitation, Hallym University College of Medicine, Anyang, Gyeonggi-do 24252, Republic of Korea
| | - Isaac Túnez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing/ Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), University of Cordoba, Cordoba 14071, Spain
- Cooperative Research Thematic Excellent Network on Brain Stimulation (REDESTIM), Ministry for Economy, Industry and Competitiveness, 28046 Madrid, Spain
| | | | - Sultan Ayoub Meo
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
12
|
Li T, Wei Y, Qu M, Mou L, Miao J, Xi M, Liu Y, He R. Formaldehyde and De/Methylation in Age-Related Cognitive Impairment. Genes (Basel) 2021; 12:genes12060913. [PMID: 34199279 PMCID: PMC8231798 DOI: 10.3390/genes12060913] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/31/2021] [Accepted: 06/10/2021] [Indexed: 12/19/2022] Open
Abstract
Formaldehyde (FA) is a highly reactive substance that is ubiquitous in the environment and is usually considered as a pollutant. In the human body, FA is a product of various metabolic pathways and participates in one-carbon cycle, which provides carbon for the synthesis and modification of bio-compounds, such as DNA, RNA, and amino acids. Endogenous FA plays a role in epigenetic regulation, especially in the methylation and demethylation of DNA, histones, and RNA. Recently, epigenetic alterations associated with FA dysmetabolism have been considered as one of the important features in age-related cognitive impairment (ARCI), suggesting the potential of using FA as a diagnostic biomarker of ARCI. Notably, FA plays multifaceted roles, and, at certain concentrations, it promotes cell proliferation, enhances memory formation, and elongates life span, effects that could also be involved in the aetiology of ARCI. Further investigation of and the regulation of the epigenetics landscape may provide new insights about the aetiology of ARCI and provide novel therapeutic targets.
Collapse
Affiliation(s)
- Ting Li
- Bayannur Hospital, Bayannur 015000, China;
| | - Yan Wei
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China; (Y.W.); (L.M.); (J.M.)
| | - Meihua Qu
- Translational Medical Center, Weifang Second People’s Hospital, The Second Affiliated Hospital of Weifang Medical University, Weifang 261041, China;
| | - Lixian Mou
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China; (Y.W.); (L.M.); (J.M.)
| | - Junye Miao
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China; (Y.W.); (L.M.); (J.M.)
| | - Mengqi Xi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (M.X.); (Y.L.)
| | - Ying Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (M.X.); (Y.L.)
| | - Rongqiao He
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China; (Y.W.); (L.M.); (J.M.)
- Correspondence:
| |
Collapse
|
13
|
Prins ND, Harrison JE, Chu HM, Blackburn K, Alam JJ, Scheltens P. A phase 2 double-blind placebo-controlled 24-week treatment clinical study of the p38 alpha kinase inhibitor neflamapimod in mild Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2021; 13:106. [PMID: 34044875 PMCID: PMC8157623 DOI: 10.1186/s13195-021-00843-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND In preclinical studies, p38⍺ kinase is implicated in Alzheimer's disease (AD) pathogenesis. In animal models, it mediates impaired synaptic dysfunction in the hippocampus, causing memory deficits, and is involved in amyloid-beta (Aβ) production and tau pathology. METHODS The REVERSE-SD (synaptic dysfunction) study was a multi-center phase 2, randomized, double-blind, placebo-controlled trial of the p38⍺ kinase inhibitor neflamapimod; conducted December 29, 2017, to June 17, 2019; 464 participants screened, and 161 randomized to either 40 mg neflamapimod (78 study participants) or matching placebo (83 study participants), orally twice daily for 24 weeks. Study participants are as follows: CSF AD-biomarker confirmed, Clinical Dementia Rating (CDR)-global score 0.5 or 1.0, CDR-memory score ≥0.5, and Mini-Mental State Examination (MMSE) 20-28. The primary endpoint was the improvement in episodic memory, assessed by combined change in Z-scores of Hopkins Verbal Learning Test-Revised (HVLT-R) Total and Delayed Recall. Secondary endpoints included change in Wechsler Memory Scale-IV (WMS) Immediate and Delayed Recall composites, CDR-SB, MMSE, and CSF biomarkers [total and phosphorylated tau (T-tau and p-tau181), Aβ1-40, Aβ1-42, neurogranin, and neurofilament light chain]. RESULTS At randomization, the mean age is 72, 50% female, 77% with CDR-global score 0.5, and mean MMSE score 23.8. The incidence of discontinuation for adverse events and serious adverse events (all considered unrelated) was 3% each. No significant differences between treatment groups were observed in the primary or secondary clinical endpoints. Significantly reduced CSF levels with neflamapimod treatment, relative to placebo, were evident for T-tau [difference (95% CI): -18.8 (-35.8, -1.8); P=0.031] and p-tau181 [-2.0 (-3.6, -0.5); P=0.012], with a trend for neurogranin [-21.0 (-43.6, 1.6); P=0.068]. In pre-specified pharmacokinetic-pharmacodynamic (PK-PD) analyses, subjects in the highest quartile of trough plasma neflamapimod levels demonstrated positive trends, compared with placebo, in HLVT-R and WMS. CONCLUSIONS AND RELEVANCE A 24-week treatment with 40 mg neflamapimod twice daily did not improve episodic memory in patients with mild AD. However, neflamapimod treatment lowered CSF biomarkers of synaptic dysfunction. Combined with PK-PD findings, the results indicate that a longer duration study of neflamapimod at a higher dose level to assess effects on AD progression is warranted. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT03402659 . Registered on January 18, 2018.
Collapse
Affiliation(s)
- Niels D Prins
- Alzheimer Center, Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands. .,Brain Research Center, Amsterdam, The Netherlands.
| | - John E Harrison
- Alzheimer Center, Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands.,Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.,Metis Cognition Ltd., Wiltshire, UK
| | | | | | | | - Philip Scheltens
- Alzheimer Center, Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands.,Brain Research Center, Amsterdam, The Netherlands
| | | |
Collapse
|
14
|
Resveratrol, Metabolic Dysregulation, and Alzheimer's Disease: Considerations for Neurogenerative Disease. Int J Mol Sci 2021; 22:ijms22094628. [PMID: 33924876 PMCID: PMC8125227 DOI: 10.3390/ijms22094628] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) has traditionally been discussed as a disease where serious cognitive decline is a result of Aβ-plaque accumulation, tau tangle formation, and neurodegeneration. Recently, it has been shown that metabolic dysregulation observed with insulin resistance and type-2 diabetes actively contributes to the progression of AD. One of the pathologies linking metabolic disease to AD is the release of inflammatory cytokines that contribute to the development of brain neuroinflammation and mitochondrial dysfunction, ultimately resulting in amyloid-beta peptide production and accumulation. Improving these metabolic impairments has been shown to be effective at reducing AD progression and improving cognitive function. The polyphenol resveratrol (RSV) improves peripheral metabolic disorders and may provide similar benefits centrally in the brain. RSV reduces inflammatory cytokine release, improves mitochondrial energetic function, and improves Aβ-peptide clearance by activating SIRT1 and AMPK. RSV has also been linked to improved cognitive function; however, the mechanisms of action are less defined. However, there is evidence to suggest that chronic RSV-driven AMPK activation may be detrimental to synaptic function and growth, which would directly impact cognition. This review will discuss the benefits and adverse effects of RSV on the brain, highlighting the major signaling pathways and some of the gaps surrounding the use of RSV as a treatment for AD.
Collapse
|
15
|
Yang J, Jia L, Li Y, Qiu Q, Quan M, Jia J. Fluid Biomarkers in Clinical Trials for Alzheimer's Disease: Current and Future Application. J Alzheimers Dis 2021; 81:19-32. [PMID: 33749646 DOI: 10.3233/jad-201068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Alzheimer's disease (AD) research is entering a unique moment in which enormous information about the molecular basis of this disease is being translated into therapeutics. However, almost all drug candidates have failed in clinical trials over the past 30 years. These many trial failures have highlighted a need for the incorporation of biomarkers in clinical trials to help improve the trial design. Fluid biomarkers measured in cerebrospinal fluid and circulating blood, which can reflect the pathophysiological process in the brain, are becoming increasingly important in AD clinical trials. In this review, we first succinctly outline a panel of fluid biomarkers for neuropathological changes in AD. Then, we provide a comprehensive overview of current and future application of fluid biomarkers in clinical trials for AD. We also summarize the many challenges that have been encountered in efforts to integrate fluid biomarkers in clinical trials, and the barriers that have begun to be overcome. Ongoing research efforts in the field of fluid biomarkers will be critical to make significant progress in ultimately unveiling disease-modifying therapies in AD.
Collapse
Affiliation(s)
- Jianwei Yang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of 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, People's Republic of China.,National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, People's Republic of China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, People's Republic of China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China
| | - Qiongqiong Qiu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China
| | - Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China.,National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, People's Republic of China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, People's Republic of China
| |
Collapse
|
16
|
Khalsa DS, Newberg AB. Spiritual Fitness: A New Dimension in Alzheimer's Disease Prevention. J Alzheimers Dis 2021; 80:505-519. [PMID: 33554917 PMCID: PMC8075383 DOI: 10.3233/jad-201433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2021] [Indexed: 01/17/2023]
Abstract
BACKGROUND Religious and spiritual interventions may have an effect on Alzheimer's disease prevention. Kirtan Kriya meditation has been shown to mitigate the deleterious effects of chronic stress on cognition, reverse memory loss, and create psychological and spiritual wellbeing, which may reduce multiple drivers of Alzheimer's disease risk. OBJECTIVE To detail a new concept in medicine called Spiritual Fitness, a merging of stress reduction, basic wellbeing, and psycho/spiritual wellbeing to prevent Alzheimer's disease. METHODS The literature on the topics mentioned above is described, including an in-depth discussion on why and how each are critical to advancing the future of Alzheimer's disease prevention. The many negative effects of chronic stress, and the benefits of Kirtan Kriya, are reviewed. The four pillars of basic wellbeing, six practical aspects of psychological wellbeing, and the four new non-sectarian features of spiritual fitness are then disclosed. Moreover, instructions on practicing Kirtan Kriya are offered in the Supplementary Material. CONCLUSION Religious and spiritual practices, including Kirtan Kriya, are crucial components in the development of enhanced cognition and well-being, which may help prevent and, in some cases, reverse cognitive decline. The key point of this review is that making a commitment to live a brain longevity lifestyle including spiritual fitness is a critically important way for aging Alzheimer's disease free. We hope that this article will inspire scientists, clinicians, and patients to embrace this new concept of spiritual fitness and make it a part of every multidomain program for the prevention of cognitive disability.
Collapse
Affiliation(s)
| | - Andrew B. Newberg
- Department of Integrative Medicine and Nutritional Sciences, Department of Radiology, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|