1
|
Chen XH, Xia W, Ma JB, Chen J, Hu J, Shi X, Yu JJ, Gong J, Liu L, Sun YA, Liu ZG. Rare mixed dementia: A case report. World J Radiol 2025; 17:102579. [PMID: 39876884 PMCID: PMC11755906 DOI: 10.4329/wjr.v17.i1.102579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/22/2024] [Accepted: 01/14/2025] [Indexed: 01/21/2025] Open
Abstract
BACKGROUND Autoimmune encephalitis (AE) is a rare and recently described neuroinflammatory disease associated with specific autoantibodies. Anti-leucine-rich glioma inactivated 1 (anti-LGI1) encephalitis is a rare but treatable type of AE discovered in recent years. Alzheimer's disease (AD) is a degenerative brain disease and the most common cause of dementia. AD may undergo a series of pathological physiological changes in brain tissue 20 years before the onset of typical symptoms. The stage of mild cognitive impairment (MCI) that occurs during this process, known as MCI due to AD, is the earliest stage with clinical symptoms. MCI is typically categorized into two subtypes: Amnestic MCI (aMCI) and non-aMCI. CASE SUMMARY This report describes a patient with rapid cognitive impairment, diagnosed with anti-LGI1 antibody-mediated AE and aMCI, and treated at Peking University Shenzhen Hospital in March 2023. The patient was hospitalized with acute memory decline for more than 3 months. Both the cerebrospinal fluid and serum were positive for anti-LGI1 antibodies, biomarkers of AD coexisting in the patient's cerebrospinal fluid. Following combination treatment with immunoglobulin therapy and glucocorticoid, plus inhibition of acetylcholinesterase, the patient's cognitive function significantly improved. Throughout the 3-month follow-up period, a sustained improvement in cognitive function was observed. The results of serum anti-LGI1 antibody were negative. CONCLUSION This case has raised awareness of the possible interaction between AE and early AD (including MCI due to AD), and alerted clinicians to the possibility of concurrent rare and common diseases in patients presenting with cognitive impairment.
Collapse
Affiliation(s)
- Xu-Hui Chen
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen 518000, Guangdong Province, China
| | - Wen Xia
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen 518000, Guangdong Province, China
| | - Jia-Bin Ma
- Laboratory of Functional Chemistry and Nutrition of Food, Northwest A&F University, Yangling 712100, Shanxi Province, China
| | - Jiao Chen
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen 518000, Guangdong Province, China
| | - Jun Hu
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen 518000, Guangdong Province, China
| | - Xin Shi
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen 518000, Guangdong Province, China
| | - Jing-Jing Yu
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen 518000, Guangdong Province, China
| | - Jia Gong
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen 518000, Guangdong Province, China
| | - Lu Liu
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen 518000, Guangdong Province, China
| | - Yong-An Sun
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Zhi-Gang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, Northwest A&F University, Yangling 712100, Shanxi Province, China
| |
Collapse
|
2
|
Tang J, Cao Z, Lei M, Yu Q, Mai Y, Xu J, Liao W, Ruan Y, Shi L, Yang L, Liu J. Heterogeneity of cerebral atrophic rate in mild cognitive impairment and its interactive association with proteins related to microglia activity on longitudinal cognitive changes. Arch Gerontol Geriatr 2024; 127:105582. [PMID: 39079281 DOI: 10.1016/j.archger.2024.105582] [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: 04/21/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Heterogeneity of cerebral atrophic rate commonly exists in mild cognitive impairment (MCI), which may be associated with microglia-involved neuropathology and have an influence on cognitive outcomes. OBJECTIVE We aim to explore the heterogeneity of cerebral atrophic rate among MCI and its association with plasma proteins related to microglia activity, with further investigation of their interaction effects on long-term cognition. SUBJECTS A total of 630 MCI subjects in the ADNI database were included, of which 260 subjects were available with baseline data on plasma proteins. METHODS Group-based multi-trajectory modeling (GBMT) was used to identify the latent classes with heterogeneous cerebral atrophic rates. Associations between latent classes and plasma proteins related to microglia activity were investigated with generalized linear models. Linear mixed effect models (LME) were implemented to explore the interaction effects between proteins related to microglia activity and identified latent classes on longitudinal cognitive changes. RESULTS Two latent classes were identified and labeled as the slow-atrophy class and the fast-atrophy class. Associations were found between such heterogeneity of atrophic rates and plasma proteins related to microglia activity, especially AXL receptor tyrosine kinase (AXL), CD40 antigen (CD40), and tumor necrosis factor receptor-like 2 (TNF-R2). Interaction effects on longitudinal cognitive changes showed that higher CD40 was associated with faster cognitive decline in the slow-atrophy class and higher AXL or TNF-R2 was associated with slower cognitive decline in the fast-atrophy class. CONCLUSIONS Heterogeneity of atrophic rates at the MCI stage is associated with several plasma proteins related to microglia activity, which show either protective or adverse effects on long-term cognition depending on the variability of atrophic rates.
Collapse
Affiliation(s)
- Jingyi Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China
| | - Zhiyu Cao
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Ming Lei
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China
| | - Qun Yu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Yingren Mai
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Jiaxin Xu
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China
| | - Wang Liao
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Yuting Ruan
- Department of Rehabilitation, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Lin Shi
- BrainNow Research Institute, Shenzhen City, Guangdong Province, MN 518000, China; Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, MN 999077, China
| | - Lianhong Yang
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China.
| | - Jun Liu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China.
| |
Collapse
|
3
|
Suleiman Khoury Z, Sohail F, Wang J, Mendoza M, Raake M, Tahoor Silat M, Reddy Bathinapatta M, Sadeghzadegan A, Meghana P, Paul J. Neuroinflammation: A Critical Factor in Neurodegenerative Disorders. Cureus 2024; 16:e62310. [PMID: 39006715 PMCID: PMC11246070 DOI: 10.7759/cureus.62310] [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] [Accepted: 06/13/2024] [Indexed: 07/16/2024] Open
Abstract
This review offers a comprehensive review of the signals and the paramount role neuroinflammation plays in neurodegenerative diseases such as Alzheimer's, Parkinson's, Huntington's, and amyotrophic lateral sclerosis. The study explores the sophisticated interactions between microglial, astrocytic, and dendritic cells and how neuroinflammation affects long-term neuronal damage and dysfunction. There are specific pathways related to the mentioned inflammatory processes, including Janus kinases/signal transducer and activator of transcriptions, nuclear factor-κB, and mitogen-activated protein kinases pathways. Neuroinflammation is argued to be a double-edged sword, being not only a protective agent that prevents further neuron damage but also the causative factor in more cell injury development. This concept of contrasting inflammation with neuroprotection advocates for the use of therapeutic techniques that seek to modulate neuroinflammatory responses as part of the neurodegeneration treatment. The recent research findings are integrated with the established knowledge to help present a comprehensive image of neuroinflammation's impact on neurodegenerative diseases and its implications for future therapy.
Collapse
Affiliation(s)
| | - Fatima Sohail
- Department of Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, USA
| | - Jada Wang
- Department of Medicine, St. George's University, Brooklyn, USA
| | - Moises Mendoza
- Department of Health Sciences, Universidad Centroccidental Lisandro Alvarado, Barquisimeto, VEN
| | - Mohammed Raake
- Department of Medicine, Annamalai University, Chennai, IND
| | | | | | - Amirali Sadeghzadegan
- Department of General Practice, Marmara University School of Medicine, Istanbul, TUR
| | - Patel Meghana
- Department of Medicine, Ramaiah University of Applied Sciences, Bengaluru, IND
| | - Janisha Paul
- Department of Medicine, Punjab Institute of Medical Sciences, Jalandhar, IND
| |
Collapse
|
4
|
Padala S, Setti S, Raymick J, Hanig J, Sarkar S. Evaluation and Characterization of Modified K114 Method to Localize Plaques in Rodent and Plaques and Tangles in Human Brain Tissue. Curr Alzheimer Res 2024; 21:69-80. [PMID: 38566375 DOI: 10.2174/0115672050295561240327055835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/04/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND A plethora of studies has shown the utility of several chemical dyes due to their affinity to bind Aβ to enable visualization of plaques under light or fluorescence microscope, and some of them showed affinity to bind neurofibrillary tangles (NFT) as well. However, only a few of them have the propensity to bind both senile plaques (SP) and NFT simultaneously. OBJECTIVE In our current study, we aimed to modify the K114 dye and the staining procedure to substantially improve the staining of amyloid plaques in both human and rodent brains and neurofibrillary tangles in the human brain. METHODS We modified the K114 solution and the staining procedure using Sudan Black as a modifier. Additionally, to evaluate the target of the modified K114, we performed double labeling of K114 and increased Aβ against three different epitopes. We used 5 different antibodies to detect phosphorylated tau to understand the specific targets that modified K114 binds. RESULTS Dual labeling using hyperphosphorylated antibodies against AT8, pTau, and TNT1 revealed that more than 80% hyperphosphorylated tau colocalized with tangles that were positive for modified K114, whereas more than 70% of the hyperphosphorylated tau colocalized with modified K114. On the other hand, more than 80% of the plaques that were stained with Aβ MOAB-2 were colocalized with modified K114. CONCLUSION Our modified method can label amyloid plaques within 5 min in the rat brain and within 20 min in the human brain. Our results indicated that modified K114 could be used as a valuable tool for detecting amyloid plaques and tangles with high contrast and resolution relative to other conventional fluorescence markers.
Collapse
Affiliation(s)
- Sanjana Padala
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food & Drug Administration, Jefferson, AR72079, USA
| | - Sharay Setti
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food & Drug Administration, Jefferson, AR72079, USA
| | - James Raymick
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food & Drug Administration, Jefferson, AR72079, USA
| | - Joseph Hanig
- Office of Testing & Research, Center for Drug Evaluation Research/FDA, Silver Spring, MD, USA
| | - Sumit Sarkar
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food & Drug Administration, Jefferson, AR72079, USA
| |
Collapse
|
5
|
Tubi MA, Wheeler K, Matsiyevskiy E, Hapenney M, Mack WJ, Chui HC, King K, Thompson PM, Braskie MN. White matter hyperintensity volume modifies the association between CSF vascular inflammatory biomarkers and regional FDG-PET along the Alzheimer's disease continuum. Neurobiol Aging 2023; 132:1-12. [PMID: 37708739 PMCID: PMC10843575 DOI: 10.1016/j.neurobiolaging.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 07/28/2023] [Accepted: 08/06/2023] [Indexed: 09/16/2023]
Abstract
In older adults with abnormal levels of Alzheimer's disease neuropathology, lower cerebrospinal fluid (CSF) vascular endothelial growth factor (VEGF) levels are associated with lower [¹⁸F]-fluorodeoxyglucose positron emission tomography (FDG-PET) signal, but whether this association is (1) specific to VEGF or broadly driven by vascular inflammation, or (2) modified by vascular risk (e.g., white matter hyperintensities [WMHs]) remains unknown. To address this and build upon our past work, we evaluated whether 5 CSF vascular inflammation biomarkers (vascular cell adhesion molecule 1, VEGF, C-reactive protein, fibrinogen, and von Willebrand factor)-previously associated with CSF amyloid levels-were related to FDG-PET signal and whether WMH volume modified these associations in 158 Alzheimer's Disease Neuroimaging Initiative participants (55-90 years old, 39 cognitively normal, 80 mild cognitive impairment, 39 Alzheimer's disease). We defined regions both by cortical boundary and by the 3 major vascular territories: anterior, middle, and posterior cerebral arteries. We found that WMH volume had interactive effects with CSF biomarkers (VEGF and C-reactive protein) on FDG-PET throughout the cortex in both vascular territories and conventionally defined regions of interest.
Collapse
Affiliation(s)
- Meral A Tubi
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Koral Wheeler
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Elizabeth Matsiyevskiy
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Matthew Hapenney
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Wendy J Mack
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Helena C Chui
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kevin King
- Department of Neuroradiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Meredith N Braskie
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA.
| |
Collapse
|
6
|
Li S, Chen X, Gao M, Zhang X, Han P, Cao L, Gao J, Tao Q, Zhai J, Liang D, Guo Q. The neutrophil-to-lymphocyte ratio is associated with mild cognitive impairment in community-dwelling older women aged over 70 years: a population-based cross-sectional study. Front Aging Neurosci 2023; 15:1261026. [PMID: 37781103 PMCID: PMC10539551 DOI: 10.3389/fnagi.2023.1261026] [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: 07/18/2023] [Accepted: 09/01/2023] [Indexed: 10/03/2023] Open
Abstract
Background The neutrophil-to-lymphocyte ratio (NLR) is a marker of inflammation that can be obtained quickly, conveniently, and cheaply from blood samples. However, there is no research to explore the effects of sex and age on the relationship between the NLR and mild cognitive impairment (MCI) in community-dwelling older adults. Methods A total of 3,126 individuals aged over 60 years in Shanghai were recruited for face-to-face interviews, and blood samples were collected. MCI was assessed by the Mini-Mental State Examination (MMSE) and the Instrumental Activities of Daily Living (IADL) scale, and neutrophil count and lymphocyte counts were measured in fasting blood samples. The NLR was calculated by dividing the absolute neutrophil count by the absolute lymphocyte count. Results In females, the NLR in the MCI group was significantly higher than that in the cognitively normal group (2.13 ± 0.94 vs. 1.85 ± 0.83, p < 0.001) but not in men. Logistic regression showed that a higher NLR was an independent risk factor for MCI in women [odds ratio (OR) = 1.33; 95% confidence interval (CI) = 1.14-1.55]. In addition, the elevated NLR quartile was associated with an increased risk of MCI, especially in women older than 70 years (p value for trend = 0.012). Conclusion Compared with males, female MCI patients had a significantly higher NLR than cognitively normal controls. In addition, elevated NLR was found to be significantly associated with MCI risk in women older than 70 years. Therefore, elderly Chinese women with a higher NLR value may be the target population for effective prevention of MCI.
Collapse
Affiliation(s)
- Shengjie Li
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Sports and Health, Tianjin University of Sport, Tianjin, China
| | - Xiaoyu Chen
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Mengze Gao
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Sports and Health, Tianjin University of Sport, Tianjin, China
| | - Xingyu Zhang
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Sports and Health, Tianjin University of Sport, Tianjin, China
| | - Peipei Han
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Liou Cao
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Gao
- General Practice Clinic, Pujiang Community Health Service Center in Minhang District, Shanghai, China
| | - Qiongying Tao
- Jiading Subdistrict Community Health Center, Shanghai, China
| | - Jiayi Zhai
- Jiading Subdistrict Community Health Center, Shanghai, China
| | - Dongyu Liang
- Clinical Research Center, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Qi Guo
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
7
|
de la Monte SM, Tong M, Hapel AJ. Concordant and Discordant Cerebrospinal Fluid and Plasma Cytokine and Chemokine Responses in Mild Cognitive Impairment and Early-Stage Alzheimer's Disease. Biomedicines 2023; 11:2394. [PMID: 37760836 PMCID: PMC10525668 DOI: 10.3390/biomedicines11092394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Neuroinflammation may be a pathogenic mediator and biomarker of neurodegeneration at the boundary between mild cognitive impairment (MCI) and early-stage Alzheimer's disease (AD). Whether neuroinflammatory processes are endogenous to the central nervous system (CNS) or originate from systemic (peripheral blood) sources could impact strategies for therapeutic intervention. To address this issue, we measured cytokine and chemokine immunoreactivities in simultaneously obtained lumbar puncture cerebrospinal fluid (CSF) and serum samples from 39 patients including 18 with MCI or early AD and 21 normal controls using a 27-plex XMAP bead-based enzyme-linked immunosorbent assay (ELISA). The MCI/AD combined group had significant (p < 0.05 or better) or statistically trend-wise (0.05 ≤ p ≤ 0.10) concordant increases in CSF and serum IL-4, IL-5, IL-9, IL-13, and TNF-α and reductions in GM-CSF, b-FGF, IL-6, IP-10, and MCP-1; CSF-only increases in IFN-y and IL-7 and reductions in VEGF and IL-12p70; serum-only increases in IL-1β, MIP-1α, and eotaxin and reductions in G-CSF, IL-2, IL-8 and IL-15; and discordant CSF-serum responses with reduced CSF and increased serum PDGF-bb, IL-17a, and RANTES. The results demonstrate simultaneously parallel mixed but modestly greater pro-inflammatory compared to anti-inflammatory or neuroprotective responses in CSF and serum. In addition, the findings show evidence that several cytokines and chemokines are selectively altered in MCI/AD CSF, likely corresponding to distinct neuroinflammatory responses unrelated to systemic pathologies. The aggregate results suggest that early management of MCI/AD neuroinflammation should include both anti-inflammatory and pro-neuroprotective strategies to help prevent disease progression.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Departments of Pathology (Neuropathology), Neurology, and Neurosurgery, Rhode Island Hospital, The Alpert Medical School of Brown University, Providence, RI 02903, USA
- Department of Medicine, Rhode Island Hospital, The Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, The Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Andrew J. Hapel
- Department of Genome Biology, John Curtin School of Medical Research, Australian National University, Canberra 2601, Australia;
| |
Collapse
|
8
|
Hu WT, Nayyar A, Kaluzova M. Charting the Next Road Map for CSF Biomarkers in Alzheimer's Disease and Related Dementias. Neurotherapeutics 2023; 20:955-974. [PMID: 37378862 PMCID: PMC10457281 DOI: 10.1007/s13311-023-01370-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 06/29/2023] Open
Abstract
Clinical prediction of underlying pathologic substrates in people with Alzheimer's disease (AD) dementia or related dementia syndromes (ADRD) has limited accuracy. Etiologic biomarkers - including cerebrospinal fluid (CSF) levels of AD proteins and cerebral amyloid PET imaging - have greatly modernized disease-modifying clinical trials in AD, but their integration into medical practice has been slow. Beyond core CSF AD biomarkers (including beta-amyloid 1-42, total tau, and tau phosphorylated at threonine 181), novel biomarkers have been interrogated in single- and multi-centered studies with uneven rigor. Here, we review early expectations for ideal AD/ADRD biomarkers, assess these goals' future applicability, and propose study designs and performance thresholds for meeting these ideals with a focus on CSF biomarkers. We further propose three new characteristics: equity (oversampling of diverse populations in the design and testing of biomarkers), access (reasonable availability to 80% of people at risk for disease, along with pre- and post-biomarker processes), and reliability (thorough evaluation of pre-analytical and analytical factors influencing measurements and performance). Finally, we urge biomarker scientists to balance the desire and evidence for a biomarker to reflect its namesake function, indulge data- as well as theory-driven associations, re-visit the subset of rigorously measured CSF biomarkers in large datasets (such as Alzheimer's disease neuroimaging initiative), and resist the temptation to favor ease over fail-safe in the development phase. This shift from discovery to application, and from suspended disbelief to cogent ingenuity, should allow the AD/ADRD biomarker field to live up to its billing during the next phase of neurodegenerative disease research.
Collapse
Affiliation(s)
- William T Hu
- Department of Neurology, Rutgers Biomedical and Health Sciences, Rutgers-Robert Wood Johnson Medical School, 125 Paterson Street, Suite 6200, New Brunswick, NJ, 08901, USA.
- Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
| | - Ashima Nayyar
- Department of Neurology, Rutgers Biomedical and Health Sciences, Rutgers-Robert Wood Johnson Medical School, 125 Paterson Street, Suite 6200, New Brunswick, NJ, 08901, USA
| | - Milota Kaluzova
- Department of Neurology, Rutgers Biomedical and Health Sciences, Rutgers-Robert Wood Johnson Medical School, 125 Paterson Street, Suite 6200, New Brunswick, NJ, 08901, USA
| |
Collapse
|
9
|
Pillai JA, Bena J, Bekris L, Kodur N, Kasumov T, Leverenz JB, Kashyap SR. Metabolic syndrome biomarkers relate to rate of cognitive decline in MCI and dementia stages of Alzheimer's disease. Alzheimers Res Ther 2023; 15:54. [PMID: 36927447 PMCID: PMC10018847 DOI: 10.1186/s13195-023-01203-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND The relationship between biomarkers of metabolic syndrome and insulin resistance, plasma triglyceride/HDL cholesterol (TG/HDL-C) ratio, on the rate of cognitive decline in mild cognitive impairment (MCI) and dementia stages of Alzheimer's disease (AD) is unknown. The role of peripheral and cerebrospinal fluid (CSF) levels of Apolipoprotein A1 (ApoA1), a key functional component of HDL, on cognitive decline also remains unclear among them. Here we evaluate baseline plasma TG/HDL-C ratio and CSF and plasma ApoA1 levels and their relation with cognitive decline in the MCI and Dementia stages of AD. PATIENTS AND METHODS A retrospective longitudinal study (156 participants; 106 MCI, 50 AD dementia) from the Alzheimer's Disease Neuroimaging Initiative, with an average of 4.0 (SD 2.8) years follow-up. Baseline plasma TG/HDL-C, plasma, and CSF ApoA1 and their relationship to inflammation and blood-brain barrier (BBB) biomarkers and longitudinal cognitive outcomes were evaluated. Multivariable linear mixed effect models were used to assess the effect of baseline analytes with longitudinal changes in Mini-Mental State Exam (MMSE), Clinical Dementia Rating-Sum of Boxes (CDR-SB), and Logical Memory delayed recall (LM) score after controlling for well-known covariates. RESULTS A total of 156 participants included 98 women, 63%; mean age was 74.9 (SD 7.3) years. At baseline, MCI and dementia groups did not differ significantly in TG/HDL-C (Wilcoxon W statistic = 0.39, p = 0.39) and CSF ApoA1 levels (W = 3642, p = 0.29), but the dementia group had higher plasma ApoA1 than the MCI group (W = 4615, p = 0.01). Higher TG/HDL-C ratio was associated with faster decline in CDR-SB among MCI and dementia groups. Higher plasma ApoA1 was associated with faster decline in MMSE and LM among MCI, while in contrast higher CSF ApoA1 levels related to slower cognitive decline in MMSE among MCI. CSF and plasma ApoA1 also show opposite directional correlations with biomarkers of BBB integrity. CSF but not plasma levels of ApoA1 positively correlated to inflammation analytes in the AGE-RAGE signaling pathway in diabetic complications (KEGG ID:KO04933). CONCLUSIONS Biomarkers of metabolic syndrome relate to rate of cognitive decline among MCI and dementia individuals. Elevated plasma TG/HDL-C ratio and plasma ApoA1 are associated with worse cognitive outcomes in MCI and dementia participants. CSF ApoA1 and plasma ApoA1 likely have different roles in AD progression in MCI stage.
Collapse
Affiliation(s)
- Jagan A Pillai
- Lou Ruvo Center for Brain Health, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, 9500 Euclid Ave/U10, Cleveland, OH, 44195, USA. .,Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA. .,Department of Neurology, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA. .,Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.
| | - James Bena
- Quantitative Health Sciences, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA
| | - Lynn Bekris
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.,Lerner Research Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA
| | - Nandan Kodur
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA
| | - Takhar Kasumov
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, 9500 Euclid Ave/U10, Cleveland, OH, 44195, USA.,Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.,Department of Neurology, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.,Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA
| | - Sangeeta R Kashyap
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.,Division of Endocrinology, Diabetes and Metabolism, Weill Cornell Medicine New York Presbyterian, New York, NY, 10021, USA
| | | |
Collapse
|
10
|
Mehta RI, Mehta RI. The Vascular-Immune Hypothesis of Alzheimer's Disease. Biomedicines 2023; 11:408. [PMID: 36830944 PMCID: PMC9953491 DOI: 10.3390/biomedicines11020408] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating and irreversible neurodegenerative disorder with unknown etiology. While its cause is unclear, a number of theories have been proposed to explain the pathogenesis of AD. In large part, these have centered around potential causes for intracerebral accumulation of beta-amyloid (βA) and tau aggregates. Yet, persons with AD dementia often exhibit autopsy evidence of mixed brain pathologies including a myriad of vascular changes, vascular brain injuries, complex brain inflammation, and mixed protein inclusions in addition to hallmark neuropathologic lesions of AD, namely insoluble βA plaques and neurofibrillary tangles (NFTs). Epidemiological data demonstrate that overlapping lesions diminish the βA plaque and NFT threshold necessary to precipitate clinical dementia. Moreover, a subset of persons who exhibit AD pathology remain resilient to disease while other persons with clinically-defined AD dementia do not exhibit AD-defining neuropathologic lesions. It is increasingly recognized that AD is a pathologically heterogeneous and biologically multifactorial disease with uncharacterized biologic phenomena involved in its genesis and progression. Here, we review the literature with regard to neuropathologic criteria and incipient AD changes, and discuss converging concepts regarding vascular and immune factors in AD.
Collapse
Affiliation(s)
- Rashi I. Mehta
- Department of Neuroradiology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Rupal I. Mehta
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
11
|
Karima S, Aghamollaii V, Mahmoodi Baram S, Balenci L, Lanctôt KL, Kiss A, Tafakhori A, Mahdavi M, Rajaei S, Shateri S, Yarhoseini A, Mokhtari F, Fotouhi A, Riazi A. Boswellic Acids Improve Clinical Cognitive Scores and Reduce Systemic Inflammation in Patients with Mild to Moderate Alzheimer's Disease. J Alzheimers Dis 2023; 94:359-370. [PMID: 37248896 DOI: 10.3233/jad-221026] [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: 05/31/2023]
Abstract
BACKGROUND Recent therapeutic approaches for Alzheimer's disease (AD) have had limited success. Considering the association of neuroinflammation with AD symptoms as demonstrated in multiple studies, assessment of the clinical efficacy of molecules that reduce systemic or brain inflammation is warranted. OBJECTIVE This clinical trial assessed whether boswellic acids can improve cognitive and neuropsychiatric symptoms while reducing inflammation in AD patients. METHODS A double-blind, placebo-controlled, study was conducted on 85 AD patients randomized to boswellic acids (K-Vie™ as the main ingredient in Memowell™) or placebo for 6 months. Clinical Dementia Rating-Sum of Boxes (CDR-SOB) and Mini-Mental State Examination (MMSE) scores were compared to baseline and between groups and constituted the co-primary clinical efficacy endpoints. Secondary outcomes included neuropsychiatric assessment (Neuropsychiatric Inventory-Questionnaire, NPI-Q) and assessment of AD and inflammation biomarkers. RESULTS Patients on K-Vie™ showed a 3.1- and 1.6-unit improvement in MMSE and CDR-SOB scores, respectively, when compared to patients on placebo. NPI-Q analysis revealed significant improvement in the K-Vie™ but not in the placebo group. Only mild gastrointestinal side effects were reported in a few patients. Patients on K-Vie™ showed improvement in plasma AD biomarkers and reduction of key inflammatory cytokines including IL-6 and TNF. CONCLUSION Our results support the positive cognitive effects of boswellic acids by reducing the systemic inflammation.
Collapse
Affiliation(s)
- Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vajiheh Aghamollaii
- Neurology Department, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Krista L Lanctôt
- Departments of Psychiatry and Pharmacology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Alex Kiss
- Department of Research Design and Biostatistics, Sunnybrook Research Institute, Toronto, Canada
| | - Abbas Tafakhori
- Department of Neurology, School of Medicine, Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Meisam Mahdavi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shima Rajaei
- Clinical Trial Department, Behbalin Inc., Tehran, Iran
| | - Somayeh Shateri
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Yarhoseini
- Neurology Department, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Akbar Fotouhi
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Riazi
- Kondor Pharma Inc. Mississauga, Ontario, Canada
| |
Collapse
|
12
|
Farvadi F, Hashemi F, Amini A, Alsadat Vakilinezhad M, Raee MJ. Early Diagnosis of Alzheimer's Disease with Blood Test; Tempting but Challenging. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2023; 12:172-210. [PMID: 38313372 PMCID: PMC10837916 DOI: 10.22088/ijmcm.bums.12.2.172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 11/25/2023] [Accepted: 12/13/2023] [Indexed: 02/06/2024]
Abstract
The increasing prevalence of Alzheimer's disease (AD) has led to a health crisis. According to official statistics, more than 55 million people globally have AD or other types of dementia, making it the sixth leading cause of death. It is still difficult to diagnose AD and there is no definitive diagnosis yet; post-mortem autopsy is still the only definite method. Moreover, clinical manifestations occur very late in the course of disease progression; therefore, profound irreversible changes have already occurred when the disease manifests. Studies have shown that in the preclinical stage of AD, changes in some biomarkers are measurable prior to any neurological damage or other symptoms. Hence, creating a reliable, fast, and affordable method capable of detecting AD in early stage has attracted the most attention. Seeking clinically applicable, inexpensive, less invasive, and much more easily accessible biomarkers for early diagnosis of AD, blood-based biomarkers (BBBs) seem to be an ideal option. This review is an inclusive report of BBBs that have been shown to be altered in the course of AD progression. The aim of this report is to provide comprehensive insight into the research status of early detection of AD based on BBBs.
Collapse
Affiliation(s)
- Fakhrossadat Farvadi
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Hashemi
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, the University of Newcastle, Newcastle, Australia
| | - Azadeh Amini
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical sciences, Tehran, Iran
| | | | - Mohammad Javad Raee
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
13
|
Zhang Y, Ghose U, Buckley NJ, Engelborghs S, Sleegers K, Frisoni GB, Wallin A, Lleó A, Popp J, Martinez-Lage P, Legido-Quigley C, Barkhof F, Zetterberg H, Visser PJ, Bertram L, Lovestone S, Nevado-Holgado AJ, Shi L. Predicting AT(N) pathologies in Alzheimer's disease from blood-based proteomic data using neural networks. Front Aging Neurosci 2022; 14:1040001. [PMID: 36523958 PMCID: PMC9746615 DOI: 10.3389/fnagi.2022.1040001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/04/2022] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Blood-based biomarkers represent a promising approach to help identify early Alzheimer's disease (AD). Previous research has applied traditional machine learning (ML) to analyze plasma omics data and search for potential biomarkers, but the most modern ML methods based on deep learning has however been scarcely explored. In the current study, we aim to harness the power of state-of-the-art deep learning neural networks (NNs) to identify plasma proteins that predict amyloid, tau, and neurodegeneration (AT[N]) pathologies in AD. METHODS We measured 3,635 proteins using SOMAscan in 881 participants from the European Medical Information Framework for AD Multimodal Biomarker Discovery study (EMIF-AD MBD). Participants underwent measurements of brain amyloid β (Aβ) burden, phosphorylated tau (p-tau) burden, and total tau (t-tau) burden to determine their AT(N) statuses. We ranked proteins by their association with Aβ, p-tau, t-tau, and AT(N), and fed the top 100 proteins along with age and apolipoprotein E (APOE) status into NN classifiers as input features to predict these four outcomes relevant to AD. We compared NN performance of using proteins, age, and APOE genotype with performance of using age and APOE status alone to identify protein panels that optimally improved the prediction over these main risk factors. Proteins that improved the prediction for each outcome were aggregated and nominated for pathway enrichment and protein-protein interaction enrichment analysis. RESULTS Age and APOE alone predicted Aβ, p-tau, t-tau, and AT(N) burden with area under the curve (AUC) scores of 0.748, 0.662, 0.710, and 0.795. The addition of proteins significantly improved AUCs to 0.782, 0.674, 0.734, and 0.831, respectively. The identified proteins were enriched in five clusters of AD-associated pathways including human immunodeficiency virus 1 infection, p53 signaling pathway, and phosphoinositide-3-kinase-protein kinase B/Akt signaling pathway. CONCLUSION Combined with age and APOE genotype, the proteins identified have the potential to serve as blood-based biomarkers for AD and await validation in future studies. While the NNs did not achieve better scores than the support vector machine model used in our previous study, their performances were likely limited by small sample size.
Collapse
Affiliation(s)
- Yuting Zhang
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Upamanyu Ghose
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Noel J. Buckley
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Sebastiaan Engelborghs
- Department of Biomedical Sciences, Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Department of Neurology, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Center for Neurociences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Kristel Sleegers
- Complex Genetics Group, VIB Center for Molecular Neurology, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Anders Wallin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Alberto Lleó
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Julius Popp
- Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
- Department of Geriatric Psychiatry, University Hospital of Psychiatry and University of Zürich, Zürich, Switzerland
| | | | - Cristina Legido-Quigley
- Kings College London, London, United Kingdom
- The Systems Medicine Group, Steno Diabetes Center, Gentofte, Denmark
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands
- University College London (UCL) Institutes of Neurology and Healthcare Engineering, London, United Kingdom
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, Netherlands
- Alzheimer Center, VU University Medical Center, Amsterdam, Netherlands
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Janssen R&D, High Wycombe, United Kingdom
| | | | - Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
14
|
Ye Q, Liu L, Wang Y, Li L, Wang Z, Liu G, Lin P, Li Q. Association of Type D personality and mild cognitive impairment in patients with hypertension. Front Psychol 2022; 13:974430. [PMID: 36467148 PMCID: PMC9709486 DOI: 10.3389/fpsyg.2022.974430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 10/27/2022] [Indexed: 11/15/2023] Open
Abstract
OBJECTIVE The aim of this study was to evaluate the association between Type D personality and mild cognitive impairment (MCI) in patients with hypertension. METHODS A total of 324 subjects with hypertension were included in the study. All of them completed questionnaires on demographic characteristics, Type D personality Scale, Montreal Cognitive Assessment (MoCA), Beck Anxiety Inventory (BAI) and Beck Depression Inventory (BDI). The Type D personality effect was analyzed as both dichotomous and continuous methods. RESULTS The incidence of MCI was 56.5% in hypertensive individuals. Type D personality presenting as a dichotomous construct was an independent risk factor of MCI (odds ratio [OR] = 2.814, 95% confidence interval [CI] = 1.577-5.021, p < 0.001), after adjusting for ages, sex and some clinical factors. Meanwhile, main effect of negative affectivity component was independently related to the prevalence of MCI (OR = 1.087, 95%CI = 1.014-1.165, p = 0.019). However, associations between the main effect of social inhibition component (OR = 1.011, 95%CI = 0.924-1.107, p = 0.811) as well as the interaction of negative affectivity and social inhibition (OR = 1.013, 95%CI = 0.996-1.030, p = 0.127) with MCI were not found. CONCLUSION The findings suggest that Type D personality is strongly associated with MCI in patients with hypertension. The negative affectivity component of the Type D appears to drive the correlations between Type D and MCI. These findings provide new ideas for studying the mechanisms underlying the relationship between personality and cognitive decline in hypertensive individuals.
Collapse
Affiliation(s)
- Qingfang Ye
- College of Nursing of Harbin Medical University, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Basic Nursing, School of Nursing, Harbin Medical University, Daqing, China
| | - Li Liu
- Department of Basic Nursing, School of Nursing, Harbin Medical University, Daqing, China
| | - Yini Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ling Li
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhengjun Wang
- Department of Basic Nursing, School of Nursing, Harbin Medical University, Daqing, China
| | - Guojie Liu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ping Lin
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiujie Li
- College of Nursing of Harbin Medical University, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
15
|
Oh SL, Zhou M, Chin EWM, Amarnath G, Cheah CH, Ng KP, Kandiah N, Goh ELK, Chiam KH. Alzheimer's Disease Blood Biomarkers Associated With Neuroinflammation as Therapeutic Targets for Early Personalized Intervention. Front Digit Health 2022; 4:875895. [PMID: 35899035 PMCID: PMC9309434 DOI: 10.3389/fdgth.2022.875895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022] Open
Abstract
The definitive diagnosis of Alzheimer's Disease (AD) without the need for neuropathological confirmation remains a challenge in AD research today, despite efforts to uncover the molecular and biological underpinnings of the disease process. Furthermore, the potential for therapeutic intervention is limited upon the onset of symptoms, providing motivation for studying and treating the AD precursor mild cognitive impairment (MCI), the prodromal stage of AD instead. Applying machine learning classification to transcriptomic data of MCI, AD, and cognitively normal (CN) control patients, we identified differentially expressed genes that serve as biomarkers for the characterization and classification of subjects into MCI or AD groups. Predictive models employing these biomarker genes exhibited good classification performances for CN, MCI, and AD, significantly above random chance. The PI3K-Akt, IL-17, JAK-STAT, TNF, and Ras signaling pathways were also enriched in these biomarker genes, indicating their diagnostic potential and pathophysiological roles in MCI and AD. These findings could aid in the recognition of MCI and AD risk in clinical settings, allow for the tracking of disease progression over time in individuals as part of a therapeutic approach, and provide possible personalized drug targets for early intervention of MCI and AD.
Collapse
Affiliation(s)
- Sher Li Oh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- IGP-Neuroscience, Interdisciplinary Graduate Programme, Nanyang Technological University, Singapore, Singapore
| | - Meikun Zhou
- Bioinformatics Institute, ASTAR, Singapore, Singapore
| | - Eunice W. M. Chin
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Gautami Amarnath
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Chee Hoe Cheah
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Kok Pin Ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Nagaendran Kandiah
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Eyleen L. K. Goh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- *Correspondence: Eyleen L. K. Goh
| | - Keng-Hwee Chiam
- Bioinformatics Institute, ASTAR, Singapore, Singapore
- Keng-Hwee Chiam
| |
Collapse
|
16
|
Single-Cell RNA-Seq Analysis of Olfactory Mucosal Cells of Alzheimer’s Disease Patients. Cells 2022; 11:cells11040676. [PMID: 35203328 PMCID: PMC8870160 DOI: 10.3390/cells11040676] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 12/17/2022] Open
Abstract
Olfaction is orchestrated by olfactory mucosal cells located in the upper nasal cavity. Olfactory dysfunction manifests early in several neurodegenerative disorders including Alzheimer’s disease, however, disease-related alterations to the olfactory mucosal cells remain poorly described. The aim of this study was to evaluate the olfactory mucosa differences between cognitively healthy individuals and Alzheimer’s disease patients. We report increased amyloid-beta secretion in Alzheimer’s disease olfactory mucosal cells and detail cell-type-specific gene expression patterns, unveiling 240 differentially expressed disease-associated genes compared to the cognitively healthy controls, and five distinct cell populations. Overall, alterations of RNA and protein metabolism, inflammatory processes, and signal transduction were observed in multiple cell populations, suggesting their role in Alzheimer’s disease-related olfactory mucosa pathophysiology. Furthermore, the single-cell RNA-sequencing proposed alterations in gene expression of mitochondrially located genes in AD OM cells, which were verified by functional assays, demonstrating altered mitochondrial respiration and a reduction of ATP production. Our results reveal disease-related changes of olfactory mucosal cells in Alzheimer’s disease and demonstrate the utility of single-cell RNA sequencing data for investigating molecular and cellular mechanisms associated with the disease.
Collapse
|
17
|
Li P, Wu Y, Hamlett ED, Goodwin AJ, Halushka PV, Carroll SL, Liu M, Fan H. Suppression of Fli-1 protects against pericyte loss and cognitive deficits in Alzheimer's disease. Mol Ther 2022; 30:1451-1464. [PMID: 35038582 PMCID: PMC9077320 DOI: 10.1016/j.ymthe.2022.01.023] [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] [Received: 07/20/2021] [Revised: 12/11/2021] [Accepted: 01/12/2022] [Indexed: 11/16/2022] Open
Abstract
Brain pericytes regulate cerebral blood flow, maintain the integrity of the blood-brain barrier (BBB) and facilitate the removal of amyloid β (Aβ) which is critical to healthy brain activity. Pericyte loss has been observed in brains from patients with Alzheimer's disease (AD) and animal models. Our previous data demonstrated that friend leukemia virus integration 1 (Fli-1), an ETS transcription factor, governs pericyte viability in murine sepsis; however, the role of Fli-1 and its impact on pericyte loss in AD remains unknown. Here, we demonstrated that Fli-1 expression was up-regulated in postmortem brains from a cohort of human AD donors and in 5xFAD mice, which corresponded with a decreased pericyte number, elevated inflammatory mediators, and increased Aβ accumulation as compared to cognitively normal individuals and WT mice. Antisense oligonucleotide Fli-1 Gapmer administrated via intrahippocampal injection decelerated pericyte loss, decreased inflammatory response, ameliorated cognitive deficits, improved BBB dysfunction, and reduced Aβ deposition in 5xFAD mice. Fli-1 Gapmer-mediated inhibition of Fli-1 protected against Aβ accumulation-induced human brain pericyte apoptosis in vitro. Overall, these studies indicate that Fli-1 contributes to pericyte loss, inflammatory response, Aβ deposition, vascular dysfunction and cognitive decline, and suggest that inhibition of Fli-1 may represent novel therapeutic strategies for AD.
Collapse
Affiliation(s)
- Pengfei Li
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425
| | - Yan Wu
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425
| | - Eric D Hamlett
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425
| | - Andrew J Goodwin
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Medical University of South Carolina, Charleston, SC, 29425
| | - Perry V Halushka
- Department of Medicine and Medical University of South Carolina, Charleston, SC, 29425; Department of Pharmacology and, Medical University of South Carolina, Charleston, SC, 29425
| | - Steven L Carroll
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425
| | - Meng Liu
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, 29425
| | - Hongkuan Fan
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425.
| |
Collapse
|
18
|
Alvarez XA, Winston CN, Barlow JW, Sarsoza FM, Alvarez I, Aleixandre M, Linares C, García-Fantini M, Kastberger B, Winter S, Rissman RA. Modulation of Amyloid-β and Tau in Alzheimer's Disease Plasma Neuronal-Derived Extracellular Vesicles by Cerebrolysin® and Donepezil. J Alzheimers Dis 2022; 90:705-717. [PMID: 36155516 PMCID: PMC9697063 DOI: 10.3233/jad-220575] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Plasma neuronal-derived extracellular vesicles (NDEV) contain proteins of pathological, diagnostic, and therapeutic relevance. OBJECTIVE We investigated the associations of six plasma NDEV markers with Alzheimer's disease (AD) severity, cognition and functioning, and changes in these biomarkers after Cerebrolysin®, donepezil, and a combination therapy in AD. METHODS Plasma NDEV levels of Aβ42, total tau, P-T181-tau, P-S393-tau, neurogranin, and REST were determined in: 1) 116 mild to advanced AD patients and in 20 control subjects; 2) 110 AD patients treated with Cerebrolysin®, donepezil, or combination therapy in a randomized clinical trial (RCT). Samples for NDEV determinations were obtained at baseline in the NDEV study and at baseline and study endpoint in the RCT. Cognition and functioning were assessed at the same time points. RESULTS NDEV levels of Aβ42, total tau, P-T181-tau, and P-S393-tau were higher and those of neurogranin and REST were lower in mild-to-moderate AD than in controls (p < 0.05 to p < 0.001). NDEV total tau, neurogranin, and REST increased with AD severity (p < 0.05 to p < 0.001). NDEV Aβ42 and P-T181-tau correlated negatively with serum BDNF (p < 0.05), and total-tau levels were associated to plasma TNF-α (p < 0.01) and cognitive impairment (p < 0.05). Combination therapy reduced NDEV Aβ42 with respect to monotherapies (p < 0.05); and NDEV total tau, P-T181-tau, and P-S396-tau were decreased in Cerebrolysin-treated patients compared to those on donepezil monotherapy (p < 0.05). CONCLUSION The present results demonstrate the utility of NDEV determinations of pathologic and synaptic proteins as effective AD biomarkers, as markers of AD severity, and as potential tools for monitoring the effects of anti-AD drugs.
Collapse
Affiliation(s)
- X. Anton Alvarez
- Medinova Institute of Neurosciences, Clinica Reha Salud, A Coruña, Spain
- Clinical Research Department, QPS Holdings, A Coruña, Spain
| | | | - James W. Barlow
- Department of Neurosciences, University of California, San Diego, CA, USA
| | - Floyd M. Sarsoza
- Department of Neurosciences, University of California, San Diego, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Irene Alvarez
- Medinova Institute of Neurosciences, Clinica Reha Salud, A Coruña, Spain
| | | | | | | | | | | | - Robert A. Rissman
- Department of Neurosciences, University of California, San Diego, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
19
|
Erhardt EB, Adair JC, Knoefel JE, Caprihan A, Prestopnik J, Thompson J, Hobson S, Siegel D, Rosenberg GA. Inflammatory Biomarkers Aid in Diagnosis of Dementia. Front Aging Neurosci 2021; 13:717344. [PMID: 34489684 PMCID: PMC8416621 DOI: 10.3389/fnagi.2021.717344] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/22/2021] [Indexed: 11/17/2022] Open
Abstract
Dual pathology of Alzheimer's disease (AD) and vascular cognitive impairment and dementia (VCID) commonly are found together at autopsy, but mixed dementia (MX) is difficult to diagnose during life. Biological criteria to diagnose AD have been defined, but are not available for vascular disease. We used the biological criteria for AD and white matter injury based on MRI to diagnose MX. Then we measured multiple biomarkers in CSF and blood with multiplex biomarker kits for proteases, angiogenic factors, and cytokines to explore pathophysiology in each group. Finally, we used machine learning with the Random forest algorithm to select the biomarkers of maximal importance; that analysis identified three proteases, matrix metalloproteinase-10 (MMP-10), MMP-3 and MMP-1; three angiogenic factors, VEGF-C, Tie-2 and PLGF, and three cytokines interleukin-2 (IL-2), IL-6, IL-13. To confirm the clinical importance of the variables, we showed that they correlated with results of neuropsychological testing.
Collapse
Affiliation(s)
- Erik B Erhardt
- Department of Mathematics and Statistics, University of New Mexico, Albuquerque, NM, United States
| | - John C Adair
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States.,Center for Memory and Aging, Albuquerque, NM, United States
| | - Janice E Knoefel
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States.,Center for Memory and Aging, Albuquerque, NM, United States
| | | | | | | | - Sasha Hobson
- Center for Memory and Aging, Albuquerque, NM, United States
| | - David Siegel
- Department of Anesthesiology, University of New Mexico, Albuquerque, NM, United States
| | - Gary A Rosenberg
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States.,Center for Memory and Aging, Albuquerque, NM, United States
| |
Collapse
|
20
|
Sharma HS, Muresanu DF, Castellani RJ, Nozari A, Lafuente JV, Buzoianu AD, Sahib S, Tian ZR, Bryukhovetskiy I, Manzhulo I, Menon PK, Patnaik R, Wiklund L, Sharma A. Alzheimer's disease neuropathology is exacerbated following traumatic brain injury. Neuroprotection by co-administration of nanowired mesenchymal stem cells and cerebrolysin with monoclonal antibodies to amyloid beta peptide. PROGRESS IN BRAIN RESEARCH 2021; 265:1-97. [PMID: 34560919 DOI: 10.1016/bs.pbr.2021.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Military personnel are prone to traumatic brain injury (TBI) that is one of the risk factors in developing Alzheimer's disease (AD) at a later stage. TBI induces breakdown of the blood-brain barrier (BBB) to serum proteins into the brain and leads to extravasation of plasma amyloid beta peptide (ΑβP) into the brain fluid compartments causing AD brain pathology. Thus, there is a need to expand our knowledge on the role of TBI in AD. In addition, exploration of the novel roles of nanomedicine in AD and TBI for neuroprotection is the need of the hour. Since stem cells and neurotrophic factors play important roles in TBI and in AD, it is likely that nanodelivery of these agents exert superior neuroprotection in TBI induced exacerbation of AD brain pathology. In this review, these aspects are examined in details based on our own investigations in the light of current scientific literature in the field. Our observations show that TBI exacerbates AD brain pathology and TiO2 nanowired delivery of mesenchymal stem cells together with cerebrolysin-a balanced composition of several neurotrophic factors and active peptide fragments, and monoclonal antibodies to amyloid beta protein thwarted the development of neuropathology following TBI in AD, not reported earlier.
Collapse
Affiliation(s)
- Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Manzhulo
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Preeti K Menon
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
21
|
Pillai JA, Khrestian M, Bena J, Leverenz JB, Bekris LM. Temporal Ordering of Inflammatory Analytes sTNFR2 and sTREM2 in Relation to Alzheimer's Disease Biomarkers and Clinical Outcomes. Front Aging Neurosci 2021; 13:676744. [PMID: 34276339 PMCID: PMC8279003 DOI: 10.3389/fnagi.2021.676744] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/28/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammatory changes are among the key markers of Alzheimer's disease (AD) related pathological changes. Pro-inflammatory analytes have been related to cognitive decline while others have been related to attenuating neuronal death. Among them, changes in cerebrospinal fluid (CSF) levels of soluble triggering receptor expressed on myeloid cells 2 (sTREM2) and soluble tumor necrosis factor receptor 2 (sTNFR2) have been described as impacting favorable clinical outcomes in AD. We therefore evaluate the effect of CSF sTREM2 and sTNFR2 when taken together on AD biomarkers and longitudinal clinical decline to understand their relative role on impacting AD clinical biomarkers and subsequent clinical outcomes. This longitudinal observational cohort study included 168 amyloid-positive (A+) and p-tau-positive (T+) participants with mild cognitive impairment (MCI) or AD dementia from the Alzheimer's Disease Neuroimaging Initiative (ADNI) with 109 of them having concomitant CSF sTREM2 and sTNFR2 data and 48 A+ T+ participants with MCI from a tertiary memory clinic cohort. An exploratory analysis was performed using data from 86 cognitively normal (CN) participants from ADNI with 72 of them having concomitant CSF AD biomarkers and CSF sTREM2 and sTNFR2 data. General linear models were used to evaluate the effect of sTREM2 and sTNFR2 levels on baseline CSF Aβ42, t-tau, and p-tau, and a linear mixed-effects model was used to assess longitudinal cognitive change after controlling for well-known covariates. Among ADNI A+ T+ MCI and AD dementia participants, CSF sTNFR2 had a stronger association, than CSF sTREM2, with CSF t-tau and p-tau. This was replicated among A+ T+ MCI participants from the memory clinic cohort. On the contrary, among A+ T+ CN participants, CSF sTREM2 explained significant variance in CSF t-tau and p-tau, while CSF sTNFR2 did not. When the effects of CSF sTNFR2 and t-tau on longitudinal cognitive change were taken into account, higher CSF sTREM2 predicted slower cognitive decline in A+ T+ AD dementia participants and faster decline in A+ T+ CN participants. Our results show that given the dynamic changes in sTREM2 and sTNFR2, the clinical impact of these distinct inflammation related biomarkers in tracking AD temporal progression across disease stages are likely to differ.
Collapse
Affiliation(s)
- Jagan A. Pillai
- Department of Neurology, Cleveland Clinic, Cleveland, OH, United States
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, United States
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Maria Khrestian
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, United States
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - James Bena
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - James B. Leverenz
- Department of Neurology, Cleveland Clinic, Cleveland, OH, United States
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, United States
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Lynn M. Bekris
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, United States
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
22
|
Della Vecchia A, Mucci F, Pozza A, Marazziti D. Negative Air Ions in Neuropsychiatric Disorders. Curr Med Chem 2021; 28:2521-2539. [PMID: 32603272 DOI: 10.2174/0929867327666200630104550] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/04/2020] [Accepted: 05/14/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Air ions (AIs) are clusters of ionized particles present in the atmosphere, carrying an electrical charge of negative or positive polarity. Past speculations suggested that exposure to positive air ions may be harmful, while exposure to negative air ions (NAIs) may be associated with beneficial health effects. Increasing attention has been directed towards investigating the potential effect of NAIs on human brain activities since initial observations of their beneficial effects on some cognitive processes and mood. AIMS Given the paucity and scattered literature, our paper aims to review the available studies on potential positive effects of NAIs exposure on cognitive performances and depression. DISCUSSION The review of the literature seems to confirm the effects of NAIs on several brain functions. Indeed, a significant association between NAIs exposure and both well-being and high cognitive performances has been described. Furthermore, exposure to high concentrations of NAIs could be related to the improvement of depressive symptoms. CONCLUSION A growing evidence of data, although not yet conclusive, would suggest that NAIs might improve cognitive processes. These findings require specific and urgent controlled trials adopting systems based on AIs release to possibly prevent and treat cognitive dysfunctions present in a broad range of neuropsychiatric conditions.
Collapse
Affiliation(s)
- Alessandra Della Vecchia
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma 67, 56100 Pisa, Italy
| | - Federico Mucci
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma 67, 56100 Pisa, Italy
| | - Andrea Pozza
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Donatella Marazziti
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma 67, 56100 Pisa, Italy
| |
Collapse
|
23
|
Pillai JA, Bebek G, Khrestian M, Bena J, Bergmann CC, Bush WS, Leverenz JB, Bekris LM. TNFRSF1B Gene Variants and Related Soluble TNFR2 Levels Impact Resilience in Alzheimer's Disease. Front Aging Neurosci 2021; 13:638922. [PMID: 33716716 PMCID: PMC7947258 DOI: 10.3389/fnagi.2021.638922] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor necrosis factor receptor 2 (TNFR2) promotes neuronal survival downstream. This longitudinal study evaluated whether the TNFRSF1B gene encoding TNFR2 and levels of its soluble form (sTNFR2) affect Alzheimer disease (AD) biomarkers and clinical outcomes. Data analyzed included 188 patients in the Alzheimer's Disease Neuroimaging Initiative (ADNI) who had mild cognitive impairment (MCI) and AD dementia. Further, a replication study was performed in 48 patients with MCI with positive AD biomarkers who were treated at a memory clinic. Cerebrospinal fluid (CSF) sTNFR2 levels along with two related TNFRSF1B gene single nucleotide polymorphisms (SNPs) rs976881 and rs1061622 were assessed. General linear models were used to evaluate the effect of CSF sTNFR2 levels and each SNP in relationship to CSF t-tau and p-tau, cognitive domains, MRI brain measures, and longitudinal cognitive changes after adjustments were made for covariates such as APOE ε4 status. In the ADNI cohort, a significant interaction between rs976881 and CSF sTNFR2 modulates CSF t-tau and p-tau levels; hippocampal and whole brain volumes; and Digit Span Forwards subtest scores. In the replication cohort, a significant interaction between rs976881 and CSF sTNFR2 modulates CSF p-tau. A significant interaction between rs976881 and CSF sTNFR2 also impacts Clinical Dementia Rating Sum of Boxes scores over 12 months in the ADNI cohort. The interaction between TNFRSF1B variant rs976881 and CSF sTNFR2 levels was noted to modulate multiple AD-associated severity markers and cognitive domains. This interaction impacts resilience-related clinical outcomes in AD and lends support to sTNFR2 as a promising candidate for therapeutic targeting to improve clinical outcomes of interest.
Collapse
Affiliation(s)
- Jagan A. Pillai
- Department of Neurology, Cleveland Clinic, Cleveland, OH, United States
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, United States
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Gurkan Bebek
- Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, OH, United States
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, United States
| | - Maria Khrestian
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - James Bena
- Department of Quantitative Health Science, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Cornelia C. Bergmann
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - William S. Bush
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - James B. Leverenz
- Department of Neurology, Cleveland Clinic, Cleveland, OH, United States
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, United States
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Lynn M. Bekris
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
24
|
What is 'Alzheimer's disease'? The neuropathological heterogeneity of clinically defined Alzheimer's dementia. Curr Opin Neurol 2021; 34:237-245. [PMID: 33591030 DOI: 10.1097/wco.0000000000000912] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW Beta-amyloid with paired helical filaments (PHF)-tau neurofibrillary tangles define hallmark Alzheimer's disease neuropathologic changes (AD-NC). Yet persons with Alzheimer's dementia, defined broadly as an amnestic multidomain progressive dementia, often exhibit postmortem evidence of other neuropathologies including other neurodegenerative (Lewy body disease and transactive response DNA-binding protein disease) and vascular-related brain lesions. Clinicopathologic and epidemiologic analyses demonstrate the significance of these substrates, as coinciding neuropathologies mitigate the threshold for diagnosis of Alzheimer's dementia. In addition, other biologic processes may also independently underlie a progressive amnestic dementia. Advances in research on the relationship between age-related cognitive decline and the underlying neuropathologic substrates indicate that consensus neuropathologic criteria or disease nomenclature may need new considerations or refinement. This review appraises seminal literature as well as mixed pathologies and biological factors that may be determinants of clinical and pathologic disease. RECENT FINDINGS Cognition in aging (spanning from normal cognition to dementia) represents a clinical continuum. Traditional neuropathologic substrates of dementia however do not explain the variability of cognitive decline. Conversely, not all patients with AD-NC exhibit symptomatology of Alzheimer's dementia. In addition to diagnostic plaques and tangles, other neurodegenerative, cerebrovascular, and perivascular substrates manifest through discrete tissue lesions. Factors related to energetics, neurogenetics, neuroimmunology, resilience, proteinopathies, and waste clearance are increasingly suggested to be general drivers of disease. Recognition of novel neuroimmune pathways and brain-body connections further suggest there may be broader extracranial determinants of person-specific disease. SUMMARY Alzheimer's dementia is a pathologically heterogeneous and biologically multilayered disease. Recent studies and exercises in nomenclature reveal shortcomings in existing terminologies. Recognizing and overcoming these limitations is required for experts to effectively communicate about and ultimately prevent and treat Alzheimer's dementia.
Collapse
|
25
|
Day GS, Gordon BA, McCullough A, Bucelli RC, Perrin RJ, Benzinger TLS, Ances BM. Flortaucipir (tau) PET in LGI1 antibody encephalitis. Ann Clin Transl Neurol 2021; 8:491-497. [PMID: 33410601 PMCID: PMC7886030 DOI: 10.1002/acn3.51297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/27/2020] [Accepted: 12/16/2020] [Indexed: 01/05/2023] Open
Abstract
The contributors to persistent cognitive impairment and hippocampal atrophy in leucine-rich glioma-inactivated 1 antibody encephalitis (LGI1) patients are unknown. We evaluated whether tau neuropathology measured with [18 F]flortaucipir PET neuroimaging associated with persistent cognitive impairment and hippocampal atrophy in four recovering LGI1 patients (3 men; median age, 67 [37-88] years). Imaging findings in cases were compared with those observed in age- and gender-similar cognitively normal individuals (n = 124) and individuals with early-symptomatic Alzheimer disease (n = 11). Elevated [18 F]flortaucipir retention was observed in the two LGI1 patients with hippocampal atrophy and persistent cognitive impairment, including one with autopsy-confirmed Alzheimer disease. Tau neuropathology may associate with cognitive complaints and hippocampal atrophy in recovering LGI1 patients.
Collapse
Affiliation(s)
- Gregory S. Day
- Department of NeurologyMayo ClinicJacksonvilleFloridaUSA
| | - Brian A. Gordon
- Washington University School of MedicineSaint LouisMissouriUSA
- Mallinckrodt Institute of RadiologySaint LouisMissouriUSA
| | - Austin McCullough
- Washington University School of MedicineSaint LouisMissouriUSA
- Mallinckrodt Institute of RadiologySaint LouisMissouriUSA
| | - Robert C. Bucelli
- Washington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of Medicine in Saint Louis JacksonvilleSaint LouisMissouriUSA
| | - Richard J. Perrin
- Washington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of Medicine in Saint Louis JacksonvilleSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University in Saint Louis School of MedicineSaint LouisMissouriUSA
| | - Tammie L. S. Benzinger
- Washington University School of MedicineSaint LouisMissouriUSA
- Mallinckrodt Institute of RadiologySaint LouisMissouriUSA
| | - Beau M. Ances
- Washington University School of MedicineSaint LouisMissouriUSA
- Mallinckrodt Institute of RadiologySaint LouisMissouriUSA
- Department of NeurologyWashington University School of Medicine in Saint Louis JacksonvilleSaint LouisMissouriUSA
| |
Collapse
|
26
|
Pillai JA, Bena J, Bekris LM, Foldvary-Schaefer N, Heinzinger C, Rao S, Rao SM, Leverenz JB, Mehra R. Unique Sleep and Circadian Rhythm Dysfunction Neuroinflammatory and Immune Profiles in Alzheimer's Disease with Mild Cognitive Impairment. J Alzheimers Dis 2021; 81:487-492. [PMID: 33814445 PMCID: PMC8179975 DOI: 10.3233/jad-201573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Sleep dysfunction has been identified in the pathophysiology of Alzheimer's disease (AD); however, the role and mechanism of circadian rhythm dysfunction is less well understood. In a well-characterized cohort of patients with AD at the mild cognitive impairment stage (MCI-AD), we identify that circadian rhythm irregularities were accompanied by altered humoral immune responses detected in both the cerebrospinal fluid and plasma as well as alterations of cerebrospinal fluid biomarkers of neurodegeneration. On the other hand, sleep disruption was more so associated with abnormalities in circulating markers of immunity and inflammation and decrements in cognition.
Collapse
Affiliation(s)
- Jagan A. Pillai
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, USA
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Neurology, Cleveland Clinic, Cleveland, OH, USA
| | - James Bena
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Lynn M. Bekris
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Catherine Heinzinger
- Sleep Disorders Center, Neurologic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sujata Rao
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Stephen M. Rao
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, USA
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - James B. Leverenz
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, USA
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Neurology, Cleveland Clinic, Cleveland, OH, USA
| | - Reena Mehra
- Sleep Disorders Center, Neurologic Institute, Cleveland Clinic, Cleveland, OH, USA
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA
- Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
27
|
Ayton S, Janelidze S, Roberts B, Palmqvist S, Kalinowski P, Diouf I, Belaidi AA, Stomrud E, Bush AI, Hansson O. Acute phase markers in CSF reveal inflammatory changes in Alzheimer's disease that intersect with pathology, APOE ε4, sex and age. Prog Neurobiol 2020; 198:101904. [PMID: 32882319 DOI: 10.1016/j.pneurobio.2020.101904] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 01/31/2023]
Abstract
It is unknown how neuroinflammation may feature in the etiology of Alzheimer's disease (AD). We profiled acute phase response (APR) proteins (α1-antitrypsin, α1-antichymotrypsin, ceruloplasmin, complement C3, ferritin, α-fibrinogen, β-fibrinogen, γ-fibrinogen, haptoglobin, hemopexin) in CSF of 1291 subjects along the clinical and biomarker spectrum of AD to investigate the association between inflammatory changes, disease outcomes, and demographic variables. Subjects were stratified by Aβ42/t-tau as well as the following clinical diagnoses: cognitively normal (CN); subjective cognitive decline (SCD); mild cognitive impairment (MCI); and AD dementia. In separate multiple regressions (adjusting for diagnosis, age, sex, APOE-ε4) of each APR protein and a composite of all APR proteins, CSF Aβ42/t-tau status was associated with elevated ferritin, but not any other APR protein in CN and SCD subjects. Rather, the APR was elevated along with symptomatic progression (CN < SCD < MCI < AD), and this was elevation was mediated by CSF p-tau181. APOE ε4 status did not affect levels of any APR proteins in CSF, while these were elevated in males and with increased age. The performance of the APR in predicting clinical diagnosis was influenced by APOE ε4 status, sex, and age. These data provide new insight into inflammatory changes in AD and how this intersects with pathology changes and patient demographics.
Collapse
Affiliation(s)
- Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Blaine Roberts
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Pawel Kalinowski
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Ibrahima Diouf
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Abdel A Belaidi
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia.
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
28
|
Pillai JA, Bena J, Bebek G, Bekris LM, Bonner‐Jackson A, Kou L, Pai A, Sørensen L, Neilsen M, Rao SM, Chance M, Lamb BT, Leverenz JB. Inflammatory pathway analytes predicting rapid cognitive decline in MCI stage of Alzheimer's disease. Ann Clin Transl Neurol 2020; 7:1225-1239. [PMID: 32634865 PMCID: PMC7359114 DOI: 10.1002/acn3.51109] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/20/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To determine the inflammatory analytes that predict clinical progression and evaluate their performance against biomarkers of neurodegeneration. METHODS A longitudinal study of MCI-AD patients in a Discovery cohort over 15 months, with replication in the Alzheimer's Disease Neuroimaging Initiative (ADNI) MCI cohort over 36 months. Fifty-three inflammatory analytes were measured in the CSF and plasma with a RBM multiplex analyte platform. Inflammatory analytes that predict clinical progression on Clinical Dementia Rating Scale-Sum of Boxes (CDR-SB) and Mini Mental State Exam scores were assessed in multivariate regression models. To provide context, key analyte results in ADNI were compared against biomarkers of neurodegeneration, hippocampal volume, and CSF neurofilament light (NfL), in receiver operating characteristic (ROC) analyses evaluating highest quartile of CDR-SB change over two years (≥3 points). RESULTS Cerebrospinal fluid inflammatory analytes in relation to cognitive decline were best described by gene ontology terms, natural killer cell chemotaxis, and endothelial cell apoptotic process and in plasma, extracellular matrix organization, blood coagulation, and fibrin clot formation described the analytes. CSF CCL2 was most robust in predicting rate of cognitive change and analytes that correlated to CCL2 suggest IL-10 pathway dysregulation. The ROC curves for ≥3 points change in CDR-SB over 2 years when comparing baseline hippocampal volume, CSF NfL, and CCL2 were not significantly different. INTERPRETATION Baseline levels of immune cell chemotactic cytokine CCL2 in the CSF and IL-10 pathway dysregulation impact longitudinal cognitive and functional decline in MCI-AD. CCL2's utility appears comparable to biomarkers of neurodegeneration in predicting rapid decline.
Collapse
Affiliation(s)
- Jagan A. Pillai
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhio44195
- Neurological InstituteCleveland ClinicClevelandOhio44195
- Department of NeurologyCleveland ClinicClevelandOhio44195
| | - James Bena
- Quantitative Health SciencesCleveland ClinicClevelandOhio44195
| | - Gurkan Bebek
- Center for Proteomics and BioinformaticsCase Western Reserve UniversityClevelandOhio44195
- Department of NutritionCase Western Reserve UniversityClevelandOhio44195
| | - Lynn M. Bekris
- Genomic Medicine InstituteCleveland ClinicClevelandOhio44195
| | - Aaron Bonner‐Jackson
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhio44195
- Neurological InstituteCleveland ClinicClevelandOhio44195
- Department of NeurologyCleveland ClinicClevelandOhio44195
| | - Lei Kou
- Quantitative Health SciencesCleveland ClinicClevelandOhio44195
| | - Akshay Pai
- Department of Computer ScienceUniversity of CopenhagenCopenhagenDenmark
- Biomediq A/SCopenhagenDenmark
- Cerebriu A/SCopenhagenDenmark
| | - Lauge Sørensen
- Department of Computer ScienceUniversity of CopenhagenCopenhagenDenmark
- Biomediq A/SCopenhagenDenmark
- Cerebriu A/SCopenhagenDenmark
| | - Mads Neilsen
- Department of Computer ScienceUniversity of CopenhagenCopenhagenDenmark
- Biomediq A/SCopenhagenDenmark
- Cerebriu A/SCopenhagenDenmark
| | - Stephen M. Rao
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhio44195
- Neurological InstituteCleveland ClinicClevelandOhio44195
- Department of NeurologyCleveland ClinicClevelandOhio44195
| | - Mark Chance
- Center for Proteomics and BioinformaticsCase Western Reserve UniversityClevelandOhio44195
- Department of NutritionCase Western Reserve UniversityClevelandOhio44195
| | - Bruce T. Lamb
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisIN46202
| | - James B. Leverenz
- Lou Ruvo Center for Brain HealthCleveland ClinicClevelandOhio44195
- Neurological InstituteCleveland ClinicClevelandOhio44195
- Department of NeurologyCleveland ClinicClevelandOhio44195
| | | |
Collapse
|
29
|
Rehiman SH, Lim SM, Neoh CF, Majeed ABA, Chin AV, Tan MP, Kamaruzzaman SB, Ramasamy K. Proteomics as a reliable approach for discovery of blood-based Alzheimer's disease biomarkers: A systematic review and meta-analysis. Ageing Res Rev 2020; 60:101066. [PMID: 32294542 DOI: 10.1016/j.arr.2020.101066] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 02/08/2023]
Abstract
In order to gauge the impact of proteomics in discovery of Alzheimer's disease (AD) blood-based biomarkers, this study had systematically reviewed articles published between 1984-2019. Articles that fulfilled the inclusion criteria were assessed for risk of bias. A meta-analysis was performed for replicable candidate biomarkers (CB). Of the 1651 articles that were identified, 17 case-control and two cohort studies, as well as three combined case-control and longitudinal designs were shortlisted. A total of 207 AD and mild cognitive impairment (MCI) CB were discovered, with 48 reported in >2 studies. This review highlights six CB, namely alpha-2-macroglobulin (α2M)ps, pancreatic polypeptide (PP)ps, apolipoprotein A-1 (ApoA-1)ps, afaminp, insulin growth factor binding protein-2 (IGFBP-2)ps and fibrinogen-γ-chainp, all of which exhibited consistent pattern of regulation in >three independent cohorts. They are involved in AD pathogenesis via amyloid-beta (Aβ), neurofibrillary tangles, diabetes and cardiovascular diseases (CVD). Meta-analysis indicated that ApoA-1ps was significantly downregulated in AD (SMD = -1.52, 95% CI: -1.89, -1.16, p < 0.00001), with low inter-study heterogeneity (I2 = 0%, p = 0.59). α2Mps was significantly upregulated in AD (SMD = 0.83, 95% CI: 0.05, 1.62, p = 0.04), with moderate inter-study heterogeneity (I2 = 41%, p = 0.19). Both CB are involved in Aβ formation. These findings provide important insights into blood-based AD biomarkers discovery via proteomics.
Collapse
|
30
|
Yamada Y, Weis CA, Thelen J, Sticht C, Schalke B, Ströbel P, Marx A. Thymoma Associated Myasthenia Gravis (TAMG): Differential Expression of Functional Pathways in Relation to MG Status in Different Thymoma Histotypes. Front Immunol 2020; 11:664. [PMID: 32373124 PMCID: PMC7176899 DOI: 10.3389/fimmu.2020.00664] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/23/2020] [Indexed: 01/29/2023] Open
Abstract
A unique feature of thymomas is their unrivaled frequency of associated myasthenia gravis (MG). Previous studies reported that MG+ thymomas contain a larger number of mature “pre-emigrant” CD4+ T cells than MG- thymomas and that most thymomas do not contain AIRE expressing cells irrespective of MG status. These findings suggest that CD4+ T cells that mature inside the abnormal microenvironment of thymomas and egress to the blood are critical to the development of thymoma-associated MG (TAMG) irrespective of thymoma histotype. However, underlying mechanisms have remained enigmatic. To get hints to mechanisms underlying TAMG, we pursue three hypotheses: (i) Functional pathways with metabolic and immunological relevance might be differentially expressed in TAMG(+) compared to TAMG(-) thymomas; (ii) differentially enriched pathways might be more evident in immature lymphocyte-poor (i.e., tumor cell/stroma-rich) thymoma subgroups; and (iii) mechanisms leading to TAMG might be different among thymoma histological subtypes. To test these hypotheses, we compared the expression of functional pathways with potential immunological relevance (N = 380) in relation to MG status separately in type AB and B2 thymomas and immature lymphocyte-rich and lymphocyte-poor subgroups of these thymoma types using the TCGA data set. We found that <10% of the investigated pathways were differentially upregulated or downregulated in MG+ compared to MG- thymomas with significant differences between AB and B2 thymomas. The differences were particularly evident, when epithelial cell/stroma-rich subsets of type AB and B2 thymomas were analyzed. Unexpectedly, some MG-associated pathways that were significantly upregulated in AB thymomas were significantly downregulated in B2 thymomas, as exemplified by the oxidative phosphorylation pathway. Conversely, the MG-associated pathway related to macrophage polarization was downregulated in MG+ AB thymoma and upregulated in MG+ B2 thymoma. We conclude that functional pathways are significantly associated with TAMG, and that some mechanisms leading to TAMG might be different among thymoma histological subtypes. Functions related to metabolisms, vascular and macrophage biology are promising new candidate mechanisms potentially involved in the pathogenesis of TAMG. More generally, the results imply that future studies addressing pathomechanisms of TAMG should take the histotype and abundance of tumor cells and non-neoplastic stromal components of thymomas into account.
Collapse
Affiliation(s)
- Yosuke Yamada
- Institute of Pathology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
| | - Cleo-Aron Weis
- Institute of Pathology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
| | - Julian Thelen
- Institute of Pathology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
| | - Carsten Sticht
- Medical Faculty Mannheim, Medical Research Center, Heidelberg University, Mannheim, Germany
| | - Berthold Schalke
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Alexander Marx
- Institute of Pathology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
31
|
Early intraneuronal amyloid triggers neuron-derived inflammatory signaling in APP transgenic rats and human brain. Proc Natl Acad Sci U S A 2020; 117:6844-6854. [PMID: 32144141 PMCID: PMC7104377 DOI: 10.1073/pnas.1914593117] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
This work provides evidence that soluble and oligomeric amyloid protein stokes neuronal inflammation during the earliest stages of Alzheimer’s disease. Identifying neuron-derived factors that engage the brain’s immune system will provide insight into how vulnerable neurons might interact with other immune cells to propagate cytotoxic signaling cascades and cellular dysfunction during disease development. Chronic inflammation during Alzheimer’s disease (AD) is most often attributed to sustained microglial activation in response to amyloid-β (Aβ) plaque deposits and cell death. However, cytokine release and microgliosis are consistently observed in AD transgenic animal models devoid of such pathologies, bringing into question the underlying processes that may be at play during the earliest AD-related immune response. We propose that this plaque-independent inflammatory reaction originates from neurons burdened with increasing levels of soluble and oligomeric Aβ, which are known to be the most toxic amyloid species within the brain. Laser microdissected neurons extracted from preplaque amyloid precursor protein (APP) transgenic rats were found to produce a variety of potent immune factors, both at the transcript and protein levels. Neuron-derived cytokines correlated with the extent of microglial activation and mobilization, even in the absence of extracellular plaques and cell death. Importantly, we identified an inflammatory profile unique to Aβ-burdened neurons, since neighboring glial cells did not express similar molecules. Moreover, we demonstrate within disease-vulnerable regions of the human brain that a neuron-specific inflammatory response may precede insoluble Aβ plaque and tau tangle formation. Thus, we reveal the Aβ-burdened neuron as a primary proinflammatory agent, implicating the intraneuronal accumulation of Aβ as a significant immunological component in the AD pathogenesis.
Collapse
|
32
|
|
33
|
Santiago JA, Bottero V, Potashkin JA. Transcriptomic and Network Analysis Highlight the Association of Diabetes at Different Stages of Alzheimer's Disease. Front Neurosci 2019; 13:1273. [PMID: 31849586 PMCID: PMC6895844 DOI: 10.3389/fnins.2019.01273] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/11/2019] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) and type 2 diabetes (T2D) are among the most prevalent chronic diseases affecting the aging population. Extensive research evidence indicates that T2D is a well-established risk factor for AD; however, the molecular mechanisms underlying this association have not been fully elucidated. Furthermore, how T2D may contribute to the progression of AD is a subject of extensive investigation. In this study, we compared the blood transcriptome of patients with mild cognitive impairment (MCI), AD, and advanced AD to those afflicted with T2D to unveil shared and unique pathways and potential therapeutic targets. Blood transcriptomic analyses revealed a positive correlation between gene expression profiles of MCI, AD, and T2D in seven independent microarrays. Interestingly, gene expression profiles from women with advanced AD correlated negatively with T2D, suggesting sex-specific differences in T2D as a risk factor for AD. Network and pathway analysis revealed that shared molecular networks between MCI and T2D were predominantly enriched in inflammation and infectious diseases whereas those networks shared between overt AD and T2D were involved in the phosphatidylinositol 3-kinase and protein kinase B/Akt (PI3K-AKT) signaling pathway, a major mediator of insulin signaling in the body. The PI3K-AKT signaling pathway became more significantly dysregulated in the advanced AD and T2D shared network. Furthermore, endocrine resistance and atherosclerosis pathways emerged as dysregulated pathways in the advanced AD and T2D shared network. Interestingly, network analysis of shared differentially expressed genes between children with T2D and MCI subjects identified forkhead box O3 (FOXO3) as a central transcriptional regulator, suggesting that it may be a potential therapeutic target for early intervention in AD. Collectively, these results suggest that T2D may be implicated at different stages of AD through different molecular pathways disrupted during the preclinical phase of AD and more advanced stages of the disease.
Collapse
Affiliation(s)
| | - Virginie Bottero
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Judith A Potashkin
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|