1
|
Williams JR, DeConne TM, Pewowaruk R, Korcarz C, Tanley J, Carlsson C, Heckbert SR, Habes M, Nasrallah I, Lockhart SN, Luchsinger JA, Ding J, Hayden KM, Hughes TM, Gepner AD. Total and Structural Carotid Artery Stiffness Are Associated With Cognitive Decline and Structural Brain Abnormalities Related to Alzheimer Disease and Alzheimer Disease-Related Dementias Pathology: The Multi-Ethnic Study of Atherosclerosis. J Am Heart Assoc 2025; 14:e039925. [PMID: 40314392 DOI: 10.1161/jaha.124.039925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/28/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Arterial stiffness is associated with pathological changes underlying Alzheimer disease and related dementias. Total pulse wave velocity can be subdivided into 2 main mechanisms: structural arterial stiffness (S-PWV) due to intrinsic remodeling of the artery wall and load-dependent arterial stiffness due to increased blood pressure. METHODS AND RESULTS In this prospective cohort study, MESA (Multi-Ethnic Study of Atherosclerosis) participants completed B-mode carotid ultrasounds from which carotid total pulse wave velocity was calculated. S-PWV was calculated by adjusting pulse wave velocity to 120/80 mmHg using a nonlinear pressure-diameter relationship, and load-dependent arterial stiffness was derived by subtracting S-PWV from total pulse wave velocity. Participants had repeated cognitive assessments with the Cognitive Abilities Screening Instrument, Digit Symbol Coding, and Digit Span combined into a global cognitive composite (N=2489). Brain magnetic resonance imaging was used to generate total gray matter volume (N=906), white matter hyperintensity volume (N=896), and total white matter fractional anisotropy (N=810). Multivariable linear fixed and mixed effects regression models related standardized pulse wave velocity components to neuroimaging and cognitive decline parameters, respectively. Greater S-PWV was associated with greater longitudinal cognitive decline in global cognitive composite score (β=-0.05, P=0.002) and subtests, whereas greater load-dependent arterial stiffness was not associated with longitudinal cognitive decline. Greater S-PWV was associated with lower gray matter volume (β=-3183.4, P=0.013) and higher log white matter hyperintensity volume (β=0.20, P<0.001), whereas load-dependent arterial stiffness was associated with lower total white matter fractional anisotropy (β=-0.004, P≤0.001). CONCLUSIONS Higher structural stiffness of the carotid artery is associated with cognitive decline, whereas both structural and load-dependent stiffness are associated with brain structural abnormalities common in Alzheimer disease-related dementias.
Collapse
Affiliation(s)
- Jeremy R Williams
- Department of Cardiovascular Medicine University of Wisconsin-School of Medicine and Public Health Madison WI USA
- William S. Middleton Memorial Veterans Hospital Madison WI USA
| | - Theodore M DeConne
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine Wake Forest School of Medicine Winton-Salem NC USA
| | | | - Claudia Korcarz
- Department of Cardiovascular Medicine University of Wisconsin-School of Medicine and Public Health Madison WI USA
| | - Jordan Tanley
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine Wake Forest School of Medicine Winton-Salem NC USA
| | - Cynthia Carlsson
- Department of Cardiovascular Medicine University of Wisconsin-School of Medicine and Public Health Madison WI USA
- William S. Middleton Memorial Veterans Hospital Madison WI USA
| | - Susan R Heckbert
- Department of Epidemiology University of Washington-School of Public Health Seattle WA USA
| | - Mohamad Habes
- Neuroimage Analytics Laboratory, and Biggs Institute Neuroimaging Core Biggs Alzheimer's Institute, University of Texas San Antonio Health Science Center San Antonio TX USA
| | - Ilya Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania Philadelphia PA USA
- Department of Radiology University of Pennsylvania Philadelphia PA USA
| | - Samuel N Lockhart
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine Wake Forest School of Medicine Winton-Salem NC USA
| | - José A Luchsinger
- Department of Medicine and Epidemiology Columbia University Irving Medical Center New York NY USA
| | - Jingzhong Ding
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine Wake Forest School of Medicine Winton-Salem NC USA
| | - Kathleen M Hayden
- Department of Social Sciences and Health Policy, Division of Public Health Sciences Wake Forest School of Medicine Winston-Salem NC USA
| | - Timothy M Hughes
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine Wake Forest School of Medicine Winton-Salem NC USA
| | - Adam D Gepner
- Department of Cardiovascular Medicine University of Wisconsin-School of Medicine and Public Health Madison WI USA
- William S. Middleton Memorial Veterans Hospital Madison WI USA
| |
Collapse
|
2
|
Vollhardt A, Frölich L, Stockbauer AC, Danek A, Schmitz C, Wahl AS. Towards a better diagnosis and treatment of dementia: Identifying common and distinct neuropathological mechanisms in Alzheimer's and vascular dementia. Neurobiol Dis 2025; 208:106845. [PMID: 39999928 DOI: 10.1016/j.nbd.2025.106845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) and vascular dementia (VaD) together contribute to almost 90 % of all dementia cases leading to major health challenges of our time with a substantial global socioeconomic burden. While in AD, the improved understanding of Amyloid beta (Aß) mismetabolism and tau hyperphosphorylation as pathophysiological hallmarks has led to significant clinical breakthroughs, similar advances in VaD are lacking. After comparing the clinical presentation, including risk factors, disease patterns, course of diseases and further diagnostic parameters for both forms of dementia, we highlight the importance of shared pathomechanisms found in AD and VaD: Endothelial damage, blood brain barrier (BBB) breakdown and hypoperfusion inducing oxidative stress and inflammation and thus trophic uncoupling in the neurovascular unit. A dysfunctional endothelium and BBB lead to the accumulation of neurotoxic molecules and Aß through impaired clearance, which in turn leads to neurodegeneration. In this context we discuss possible neuropathological parameters, which might serve as biomarkers and thus improve diagnostic accuracy or reveal targets for novel therapeutic strategies for both forms of dementia.
Collapse
Affiliation(s)
- Alisa Vollhardt
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany
| | - Lutz Frölich
- Central Institute of Mental Health, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - Anna Christina Stockbauer
- Department of Neurology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Adrian Danek
- Department of Neurology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Christoph Schmitz
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany
| | - Anna-Sophia Wahl
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany; Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany.
| |
Collapse
|
3
|
Huang Y, Tang J, Chen G, Wu Q, Wang Y, Chen J, Chen S, Liu J, Huang X. HDL-Apolipoprotein in Alzheimer's Disease Revisited: From Periphery to CNS. Aging Med (Milton) 2025; 8:164-177. [PMID: 40353050 PMCID: PMC12064998 DOI: 10.1002/agm2.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/14/2024] [Accepted: 01/23/2025] [Indexed: 05/14/2025] Open
Abstract
High-density lipoprotein (HDL), as a crucial component of lipid metabolism, have roles in regulating Alzheimer's disease (AD) core pathology amyloid β (Aβ) and phosphorylated tau (p-tau) through its apolipoproteins, which are associated with brain structures, cognition, and risk of dementia. The pool of HDL apolipoproteins-in the brain and in the periphery-has its own distinct origin, composition, and regulatory mechanisms. It remains unclear whether these apolipoproteins in the periphery and CNS play distinct roles in the pathogenesis of AD. Specifically, this review focus on the distinct associations of apolipoprotein AI and apolipoprotein E-the major components of HDL in the blood and CSF-with pathological proteins, brain integrity, cognition, and dementia progression in AD. We summarize and examine the current state of knowledge on the values of these apolipoproteins in AD pathogenesis and clinical potential.
Collapse
Affiliation(s)
- Yihong Huang
- Department of NeurologyHoujie Hospital and Clinical College of Guangdong Medical UniversityDongguanGuangdongChina
| | - Jingyi Tang
- Department of NeurologySun Yat‐Sen Memorial Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Guohua Chen
- Department of NeurologyHoujie Hospital and Clinical College of Guangdong Medical UniversityDongguanGuangdongChina
| | - Qiangqiang Wu
- Department of NeurologyHoujie Hospital and Clinical College of Guangdong Medical UniversityDongguanGuangdongChina
| | - Yongfei Wang
- Department of NeurologyHoujie Hospital and Clinical College of Guangdong Medical UniversityDongguanGuangdongChina
| | - Jianjun Chen
- Department of NeurologyHoujie Hospital and Clinical College of Guangdong Medical UniversityDongguanGuangdongChina
| | - Simei Chen
- Department of NeurologyHoujie Hospital and Clinical College of Guangdong Medical UniversityDongguanGuangdongChina
| | - Jun Liu
- Department of NeurologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Xiaoyun Huang
- Department of NeurologySun Yat‐Sen Memorial Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
4
|
Johnson TP, Tsoy E, Shen J, Rivera W, Lieu K, Salazar C, Tse M, Li Y, Goldberger L, Soo B, Kramer J, Rosen HJ, Miller BL, Sirkis DW, Bonham LW, Yokoyama JS, Alzheimer’s Disease Neuroimaging Initiative. Reduced levels of angiogenesis biomarkers predict increased symptom severity in Chinese Americans with Alzheimer's disease with demographic-specific effect. Neurocase 2025; 31:82-91. [PMID: 39924667 PMCID: PMC11919552 DOI: 10.1080/13554794.2025.2455759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/13/2025] [Indexed: 02/11/2025]
Abstract
Alzheimer's disease (AD) symptomatology, while classically studied through the lens of amyloid-β and tau burden, is likely also influenced by multiple-interacting co-pathologies like vascular disease and dysmetabolism. These co-pathologies, especially vascular disease, occur disparately in the Chinese-American population and are often treatable via therapeutics and lifestyle modifications. Given this, we explored whether plasma biomarkers, including an array of vascular-related proteins, associate with cognition in a cohort of 34 Chinese Americans clinically diagnosed as cognitively normal, with mild cognitive impairment, or with AD. We found that a composite score of plasma angiogenesis biomarkers (MMP-1, bFGF, VEGF, and VEGF-C) were positively associated with total Mini Mental State Examination scores (p = 0.045) as well as memory performance (p = 0.006), and that this relationship was most pronounced in AD (biomarker composite score within AD vs MMSE & memory, both p < 0.001). To explore whether these findings were specific to the Chinese-American population, we repeated the above analyses in 73 demographically matched non-Hispanic White American participants and found no significant associations between angiogenesis biomarkers and MMSE or memory, highlighting the potential relevance of vascular dysregulation in Chinese Americans at risk for AD.
Collapse
Affiliation(s)
- Taylor P Johnson
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Elena Tsoy
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Global Brain Health Institute, University of California, San Francisco, San Francisco, CA 94158, USA and Trinity College Dublin, Dublin, Ireland
- Department of Humanities and Social Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Jeffrey Shen
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Will Rivera
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kevin Lieu
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Cande Salazar
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Marian Tse
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yi Li
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lauren Goldberger
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Brendan Soo
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Joel Kramer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Howard J Rosen
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Global Brain Health Institute, University of California, San Francisco, San Francisco, CA 94158, USA and Trinity College Dublin, Dublin, Ireland
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Global Brain Health Institute, University of California, San Francisco, San Francisco, CA 94158, USA and Trinity College Dublin, Dublin, Ireland
| | - Daniel W Sirkis
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Luke W Bonham
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jennifer S Yokoyama
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Global Brain Health Institute, University of California, San Francisco, San Francisco, CA 94158, USA and Trinity College Dublin, Dublin, Ireland
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | | |
Collapse
|
5
|
Cognacq G, Attwood JE, DeLuca GC. Traumatic Brain Injury and Alzheimer's Disease: A Shared Neurovascular Hypothesis. Neurosci Insights 2025; 20:26331055251323292. [PMID: 40124421 PMCID: PMC11926848 DOI: 10.1177/26331055251323292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Traumatic brain injury (TBI) is a modifiable risk factor for Alzheimer's disease (AD). TBI and AD share several histopathological hallmarks: namely, beta-amyloid aggregation, tau hyperphosphorylation, and plasma protein infiltration. The relative contributions of these proteinopathies and their interplay in the pathogenesis of both conditions remains unclear although important differences are emerging. This review synthesises emerging evidence for the critical role of the neurovascular unit in mediating protein accumulation and neurotoxicity in both TBI and AD. We propose a shared pathogenic cascade centred on a neurovascular unit, in which increased blood-brain barrier permeability induces a series of noxious mechanisms leading to neuronal loss, synaptic dysfunction and ultimately cognitive dysfunction in both conditions. We explore the application of this hypothesis to outstanding research questions and potential treatments for TBI and AD, as well as other neurodegenerative and neuroinflammatory conditions. Limitations of this hypothesis, including the challenges of establishing a causal relationship between neurovascular damage and proteinopathies, are also discussed.
Collapse
Affiliation(s)
- Gabrielle Cognacq
- John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, Oxfordshire, UK
| | - Jonathan E Attwood
- Nuffield Department of Clinical Neurosciences, Level 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, Oxfordshire, UK
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, Level 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, Oxfordshire, UK
| |
Collapse
|
6
|
Xu X, Wang J, Chen T, Wang S, Wang F, He J, Meng XY, Shen Y. Deciphering novel mitochondrial signatures: multi-omics analysis uncovers cross-disease markers and oligodendrocyte pathways in Alzheimer's disease and glioblastoma. Front Aging Neurosci 2025; 17:1536142. [PMID: 40018519 PMCID: PMC11865232 DOI: 10.3389/fnagi.2025.1536142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/28/2025] [Indexed: 03/01/2025] Open
Abstract
Introduction Alzheimer's disease (AD) and glioblastoma (GBM) are severe neurological disorders that pose significant global healthcare challenges. Despite extensive research, the molecular mechanisms, particularly those involving mitochondrial dysfunction, remain poorly understood. A major limitation in current studies is the lack of cell-specific markers that effectively represent mitochondrial dynamics in AD and GBM. Methods In this study, we analyzed single-cell transcriptomic data using 10 machine learning algorithms to identify mitochondria-associated cell-specific markers. We validated these markers through the integration of gene expression and methylation data across diverse cell types. Our dataset comprised single-nucleus RNA sequencing (snRNA-seq) from AD patients, single-cell RNA sequencing (scRNA-seq) from GBM patients, and additional DNA methylation and transcriptomic data from the ROSMAP, ADNI, TCGA, and CGGA cohorts. Results Our analysis identified four significant cross-disease mitochondrial markers: EFHD1, SASH1, FAM110B, and SLC25A18. These markers showed both shared and unique expression profiles in AD and GBM, suggesting a common mitochondrial mechanism contributing to both diseases. Additionally, oligodendrocytes and their interactions with astrocytes were implicated in disease progression, particularly through the APP signaling pathway. Key hub genes, such as HS6ST3 and TUBB2B, were identified across different cellular subpopulations, highlighting a cell-specific co-expression network linked to mitochondrial function.
Collapse
Affiliation(s)
- Xuan Xu
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Jiaqi Wang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Tong Chen
- School of Biomedical Engineering, Anhui Medical University, Hefei, Anhui, China
| | - Shuaibin Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, Anhui, China
| | - Fei Wang
- School of Basic Medical Science, Anhui Medical University, Hefei, Anhui, China
| | - Junwen He
- College of Informatics, Huazhong Agricultural University, Hubei, Wuhan, China
| | - Xiang-Yu Meng
- School of Basic Medical Sciences, Medical School, Hubei Minzu University, Enshi, Hubei, China
| | - Yin Shen
- School of Biomedical Engineering, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
7
|
Elkotamy MS, Elgohary MK, Alkabbani MA, Hefina MM, Tawfik HO, Fares M, Eldehna WM, Abdel-Aziz HA. Design, synthesis, and evaluation of novel benzofuran and pyrazole-based derivatives as dual AChE/BuChE inhibitors with antioxidant properties for Alzheimer's disease management. Eur J Med Chem 2025; 283:117158. [PMID: 39673864 DOI: 10.1016/j.ejmech.2024.117158] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/19/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
As a complicated neurodegenerative disorder with several clinical hallmarks, Alzheimer's disease (AD) requires multi-target treatment medicines to address multiple elements of disease progression. In this study, we reported two novel series of compounds: benzofuran-based donepezil analogs (9a-i) and their pyrazole-based counterparts (11a-i) as potential dual inhibitors of AChE and BuChE with additional antioxidant properties, aiming to address multiple pathological aspects of AD simultaneously. The design strategy employed bioisosteric replacement, substituting donepezil's indanone motif with a benzofuran ring in series (9a-i) to maintain crucial hydrogen bonding interactions with the Phe295 residue in the enzyme's active site. Subsequently, the benzofuran ring underwent cleavage, yielding pyrazole-tethered hydroxyphenyl derivatives (11a-i). The biological evaluation revealed that benzofuran-based derivative 9g exhibited exceptional efficacy against both AChE and BuChE, with IC50 values of 0.39 and 0.51 μg/ml, respectively, although it lacked antioxidant activity. Compound 11f demonstrated dual inhibition of AChE (IC50 = 1.24 μg/ml) and BuChE (IC50 = 1.85 μg/ml) while also displaying strong DPPH free radical scavenging activity (IC50 = 3.15 μg/ml). In vivo toxicity studies on compound 11f revealed a favorable safety profile, with no signs of toxicity or adverse events in acute oral toxicity tests in male Wistar rats. Chronic administration of 11f resulted in negligible differences in blood profiles, hepatic enzymes, urea, creatinine, and albumin levels compared to the control group. Histopathological examination of hepatic and kidney tissues from treated rats showed normal histology without damage. In silico molecular docking analysis was performed to rationalize the design approaches and support the experimental findings. This study provides valuable insights into the development of multi-target compounds for potential Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Mahmoud S Elkotamy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Cairo, 11829, Egypt.
| | - Mohamed K Elgohary
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Cairo, 11829, Egypt
| | - Mahmoud Abdelrahman Alkabbani
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Cairo, 11829, Egypt
| | - Mohamed M Hefina
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Mohamed Fares
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Cairo, 11829, Egypt; School of Pharmacy, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt.
| | - Hatem A Abdel-Aziz
- Applied Organic Chemistry Department, National Research Center, Dokki, Cairo, 12622, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia St., Alexandria, 21648, Egypt.
| |
Collapse
|
8
|
Hoshide S, Nishizawa M, Kanegae H, Kario K. Association of Longitudinal Change in Ambulatory Blood Pressure With Cognitive Decline in Older Adults. JACC. ADVANCES 2025; 4:101560. [PMID: 39898343 PMCID: PMC11782814 DOI: 10.1016/j.jacadv.2024.101560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 02/04/2025]
Abstract
Background There has been no study about the association of longitudinal change in ambulatory blood pressure (BP) variability and level with cognitive decline. Objectives The purpose of the study was to evaluate whether BP changes via ambulatory BP monitoring predict cognitive decline progression. Methods Twice-annual ambulatory BP readings were examined during 5 years and their relationship with changes in the Japanese version of the Montreal Cognitive Assessment (MoCA-J) scores. BP variability was assessed using SD, coefficient of variation, and average real variability (ARV). Cognitive decline, defined as a change in the MoCA-J score, was assessed, with the threshold set at the quartile showing the greatest decrease, which we categorized as cognitive dysfunction (-4 points or less). Results Among 206 participants (mean age 79.9 [± 7.5] years), baseline 24-hour systolic blood pressure (SBP)/diastolic blood pressure (DBP) averaged 115.2/67.0 mm Hg. Over 4.98 years (IQR: 4.94-5.04 years), MoCA-J scores showed a nonsignificant decline from 20.2 (± 0.4) to 19.9 (± 0.4). A generalized linear mixed model showed that increased SD of daytime SBP (-0.064 [95% CI: -0.121 to -0.007]; P < 0.029) and DBP (-0.125 [95% CI: -0.213 to -0.037]; P = 0.005) were significantly linked to MoCA-J score decline, with similar trends for most measures except nighttime ARV. Logistic regression revealed higher ORs for cognitive decline with increased SD of daytime SBP (1.52 [95% CI: 1.18-1.96]; P = 0.001) and DBP (1.36 [95% CI: 1.09-1.71]; P = 0.007), consistent across coefficient of variation and ARV. No association was found between changes in BP level and MoCA-J score decline. Conclusions In older adults with controlled BP, increased BP variability was linked to cognitive decline, warranting further study as a prevention target.
Collapse
Affiliation(s)
- Satoshi Hoshide
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Masafumi Nishizawa
- Department of Medicine, Minamisanriku Public Medical Clinic, Miyagi, Japan
| | - Hiroshi Kanegae
- Office of Research and Analysis, Genki Plaza Medical Center for Health Care, Tokyo, Japan
| | - Kazuomi Kario
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| |
Collapse
|
9
|
Wang X, Zhu K, Wu W, Zhou D, Lu H, Du J, Cai L, Yan X, Li W, Qian X, Wang X, Ma C, Hu Y, Tian C, Sun B, Fang Z, Wu J, Jiang P, Liu J, Liu C, Fan J, Cui H, Shen Y, Duan S, Bao A, Yang Y, Qiu W, Zhang J. Prevalence of mixed neuropathologies in age-related neurodegenerative diseases: A community-based autopsy study in China. Alzheimers Dement 2025; 21:e14369. [PMID: 39582417 PMCID: PMC11782840 DOI: 10.1002/alz.14369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 11/26/2024]
Abstract
INTRODUCTION Despite extensive studies on mixed neuropathologies, data from China are limited. This study aims to fill this gap by analyzing brain samples from Chinese brain banks. METHODS A total of 1142 brains from six Chinese brain banks were examined using standardized methods. Independent pathologists conducted evaluations with stringent quality control. Prevalence and correlations of neurological disorders were analyzed. RESULTS Significant proportions of brains displayed primary age-related tauopathy (PART, 35%), limbic-predominant age-related TDP-43 encephalopathy (LATE, 46%), and aging-related tau astrogliopathy (ARTAG, 12%). Alzheimer's disease neuropathological change (ADNC, 48%), Lewy body disease (LBD, 13%), and cerebrovascular disease (CVD, 63%) were also prevalent, often co-occurring with regional variations. CVD emerged as the potential most early contributor to neuropathological changes. DISCUSSION This analysis highlights the prevalence of PART, LATE, ARTAG, ADNC, LBD, and CVD, with regional differences. The findings suggest CVD may be the earliest contributing factor, potentially preceding other neuropathologies. Highlights The prevalence of primary age-related tauopathy (PART), limbic-predominant age-related TDP-43 encephalopathy (LATE), aging-related tau astrogliopathy (ARTAG), Alzheimer's disease neuropathologic change, Lewy body disease, and cerebrovascular disease (CVD) in China, increasing with age, is comparable to other countries. Significant regional differences in the prevalences of diseases are noted. CVD develops prior to any other disorders, including PART, LATE, and ARTAG.
Collapse
|
10
|
Zimmermann G, Joly L, Schoepfer P, Doyen M, Roch V, Grignon R, Salvi P, Marie PY, Benetos A, Verger A. Early wave reflection of carotid artery is associated with 18 F-FDG PET hypometabolism in Alzheimer's brain areas of cognitively normal adults. J Hypertens 2025; 43:145-151. [PMID: 39351888 DOI: 10.1097/hjh.0000000000003886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/26/2024] [Indexed: 01/19/2025]
Abstract
INTRODUCTION Arterial stiffening likely plays a role in Alzheimer disease (AD) pathogenesis. The current study investigated whether inter-individual variations in arterial stiffness and pressure wave parameters were associated with 18 F-FDG positron emission tomography (PET) metabolism in AD-associated brain areas throughout adulthood, independently of age and before the onset of any neuropsychological disorders. METHODS A prospective, large age-range population of 67 patients (17 young, 16 middle-aged, and 34 older adults; 37 women) underwent a: brain 18 F-FDG PET, blood pressure recording, and carotid/femoral pulse wave-based measurements, including the time-to-peak of the reflected backward carotid pulse wave (bT), on the same day. Multivariable and quantitative voxel-to-voxel analyses ( P -voxel < 0.005, corrected for cluster volumes) were conducted to assess associations between vascular parameters and 18 F-FDG PET metabolism in AD-associated brain areas. RESULTS In the multivariable analysis, only increased age and decreased bT were independently associated with the decline of metabolic activity in AD-associated brain areas ( P < 0.001). In the voxel-to-voxel analysis with age as a covariate, bT was strongly associated with the metabolic activity of 40 clusters in AD-associated brain areas (clusters cumulative volume: 63 cm 3 ; T score max: 5.7). CONCLUSION In a large age-range population of adult patients, who are still unaffected by neuropsychological disorders, an early reflected arterial pressure wave, as evidenced by a decreased bT value, is strongly associated with hypometabolic activity of AD-associated brain areas, independently of age.
Collapse
Affiliation(s)
- Gaétan Zimmermann
- Université de Lorraine, CHRU Nancy, Department of Nuclear Medicine and Nancyclotep Imaging Platform
| | - Laure Joly
- Université de Lorraine, CHRU-Nancy, Geriatric Department, Nancy
- Université de Lorraine, INSERM, DCAC, Vandoeuvre Les Nancy
| | | | - Matthieu Doyen
- Université de Lorraine, CHRU Nancy, Department of Nuclear Medicine and Nancyclotep Imaging Platform
- Université de Lorraine, IADI, INSERM U1254, Nancy, France
| | - Veronique Roch
- Université de Lorraine, CHRU Nancy, Department of Nuclear Medicine and Nancyclotep Imaging Platform
| | - Rachel Grignon
- Université de Lorraine, CHRU Nancy, Department of Nuclear Medicine and Nancyclotep Imaging Platform
| | - Paolo Salvi
- Cardiology Unit, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Pierre-Yves Marie
- Université de Lorraine, CHRU Nancy, Department of Nuclear Medicine and Nancyclotep Imaging Platform
- Université de Lorraine, INSERM, DCAC, Vandoeuvre Les Nancy
| | - Athanase Benetos
- Université de Lorraine, CHRU-Nancy, Geriatric Department, Nancy
- Université de Lorraine, INSERM, DCAC, Vandoeuvre Les Nancy
| | - Antoine Verger
- Université de Lorraine, CHRU Nancy, Department of Nuclear Medicine and Nancyclotep Imaging Platform
- Université de Lorraine, IADI, INSERM U1254, Nancy, France
| |
Collapse
|
11
|
Cummings TH, Magagnoli J, Sikirzhytskaya S, Tyagin I, Safro I, Wyatt MD, Shtutman M, Sutton SS. Exposure to angiotensin-converting enzyme inhibitors that cross the blood-brain barrier and the risk of dementia among people with HIV. AIDS 2024; 38:1999-2009. [PMID: 39250700 PMCID: PMC11666406 DOI: 10.1097/qad.0000000000004009] [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: 02/25/2024] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND The decreased mortality of people with HIV (PWH) has revealed non-HIV-associated comorbidities such as neurocognitive disorders (e.g., dementia). There is an urgency to discover therapeutics to prevent or delay neurocognitive decline among PWH. METHODS The artificial intelligence platform Automatic Graph-mining And Transformer based Hypothesis Generation Approach (AGATHA) was utilized to seek potential drugs to be repurposed for the management of non-HIV-associated dementia. AGATHA revealed angiotensin-converting enzyme inhibitors that cross the blood-brain barrier (BBB ACEi) as a target for decreasing dementia. Subsequently, we conducted a retrospective study evaluating incident dementia using the Veterans Affairs Informatics and Computing Infrastructure (VINCI) evaluating ACE inhibitors. Cox proportional hazards models were fit and hazard ratios (HRs) with corresponding 95% confidence intervals (CIs) are presented. FINDINGS A total 9419 PWH exposed to an BBB ACE inhibitor (ACEi) and 8831 PWH unexposed demonstrated that PWH exposed to BBB ACEi had a 21.4% (univariate) and 15.2% (multivariate) lower hazard of dementia. The propensity score matched analysis demonstrated a 14.3% lower hazard of incident dementia compared to BBB ACEi unexposed (HR 0.857, 95% CI 0.747-0.984). INTERPRETATION An artificial intelligence-based literature mining system (AGATHA) was utilized to uncover a medication with potential to be repurposed. AGATHA demonstrated that BBB ACEi as a target for decreasing dementia among PWH. Additionally, we conducted a retrospective study demonstrating a decrease in incident dementia among PWH exposed to BBB ACEi. Future research is needed to explore further and understand the relationship of dementia among PWH exposed to ACEi.
Collapse
Affiliation(s)
- Tammy H. Cummings
- Dorn Research Institute, Columbia Veterans Affairs Health Care System, Columbia, South Carolina
- College of Pharmacy, University of South Carolina, Department of Clinical Pharmacy & Outcomes Sciences
| | - Joseph Magagnoli
- Dorn Research Institute, Columbia Veterans Affairs Health Care System, Columbia, South Carolina
- College of Pharmacy, University of South Carolina, Department of Clinical Pharmacy & Outcomes Sciences
| | - Sasha Sikirzhytskaya
- College of Pharmacy, University of South Carolina, Department of Drug Discovery & Biomedical Sciences
| | - Ilya Tyagin
- Department of Computer and Information Sciences, University of Delaware
| | - Ilya Safro
- Department of Computer and Information Sciences, University of Delaware
| | - Michael D. Wyatt
- College of Pharmacy, University of South Carolina, Department of Drug Discovery & Biomedical Sciences
| | - Michael Shtutman
- College of Pharmacy, University of South Carolina, Department of Drug Discovery & Biomedical Sciences
| | - S. Scott Sutton
- Dorn Research Institute, Columbia Veterans Affairs Health Care System, Columbia, South Carolina
- College of Pharmacy, University of South Carolina, Department of Clinical Pharmacy & Outcomes Sciences
| |
Collapse
|
12
|
de Havenon A, Gottesman RF, Willamson JD, Rost N, Sharma R, Li V, Littig L, Stulberg E, Falcone GJ, Prabhakaran S, Schneider ALC, Sheth KN, Pajewski NM, Brickman AM. White matter hyperintensity on MRI and plasma Aβ42/40 ratio additively increase the risk of cognitive impairment in hypertensive adults. Alzheimers Dement 2024; 20:6810-6819. [PMID: 39229896 PMCID: PMC11485393 DOI: 10.1002/alz.14126] [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: 04/11/2024] [Revised: 06/04/2024] [Accepted: 06/18/2024] [Indexed: 09/05/2024]
Abstract
INTRODUCTION Dementia often involves comorbid Alzheimer's and vascular pathology, but their combined impact warrants additional study. METHODS We analyzed the Systolic Blood Pressure Intervention Trial and categorized white matter hyperintensity (WMH) volume into highest versus lowest/mid tertile and the amyloid beta (Aβ)42/40 ratio into lowest versus mid/highest ratio tertile. Using these binary variables, we created four exposure categories: (1) combined low risk, (2) Aβ risk, (3) WMH risk, and (4) combined high risk. RESULTS In the cohort of 467 participants (mean age 69.7 ± 7.1, 41.8% female, 31.9% nonwhite or Hispanic) during 4.8 years of follow-up and across the four exposure categories the rates of cognitive impairment were 5.3%, 7.8%, 11.8%, and 22.6%. Compared to the combined low-risk category, the adjusted hazard ratio for cognitive impairment was 4.12 (95% confidence interval, 1.71 to 9.94) in the combined high-risk category. DISCUSSION This study emphasizes the potential impact of therapeutic approaches to dementia prevention that target both vascular and amyloid pathology. HIGHLIGHTS White matter hyperintensity (WMH) and plasma amyloid (Aβ42/40) are additive risk factors for the development of cognitive impairment in the SPRINT MIND trial. Individuals in the high-risk categories of both WMH and Aβ42/40 had a near fivefold increase in risk of cognitive impairment during 4.8 years of follow-up on average. These findings suggest that treatment strategies targeting both vascular health and amyloid burden warrant further research.
Collapse
Affiliation(s)
- Adam de Havenon
- Department of NeurologyCenter for Brain and Mind HealthYale University School of MedicineNew HavenConnecticutUSA
| | | | - Jeff D. Willamson
- Department of Internal MedicineWake Forrest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Natalia Rost
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Richa Sharma
- Department of NeurologyCenter for Brain and Mind HealthYale University School of MedicineNew HavenConnecticutUSA
| | - Vivian Li
- Department of NeurologyCenter for Brain and Mind HealthYale University School of MedicineNew HavenConnecticutUSA
| | - Lauren Littig
- Department of NeurologyCenter for Brain and Mind HealthYale University School of MedicineNew HavenConnecticutUSA
| | - Eric Stulberg
- Department of NeurologyUniversity of UtahSalt Lake CityUtahUSA
| | - Guido J. Falcone
- Department of NeurologyCenter for Brain and Mind HealthYale University School of MedicineNew HavenConnecticutUSA
| | | | - Andrea L. C. Schneider
- Department of NeurologyDepartment of BiostatisticsEpidemiology, and InformaticsUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Kevin N. Sheth
- Department of NeurologyCenter for Brain and Mind HealthYale University School of MedicineNew HavenConnecticutUSA
| | - Nicholas M. Pajewski
- Department of Biostatistics and Data ScienceWake Forrest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brainand the Department of NeurologyColumbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
13
|
Pan X, Lei Z, Chen J, Jia C, Deng J, Liu Y, Luo X, Wang L, Zi D, Wang Z, Li S, Tan J. Blocking α 1 Adrenergic Receptor as a Novel Target for Treating Alzheimer's Disease. ACS Chem Neurosci 2024. [PMID: 39325017 DOI: 10.1021/acschemneuro.4c00411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024] Open
Abstract
While amyloidopathy and tauopathy have been recognized as hallmarks in Alzheimer's disease (AD) brain, recently, increasing lines of evidence have supported the pathological roles of cerebrovascular changes in the pathogenesis and progression of AD. Restoring or ameliorating the impaired cerebrovascular function during the early phase of the disease may yield benefits against the cognitive decline in AD. In the present study, we evaluated the potential therapeutic effects of nicergoline [NG, a well-known α1 adrenergic receptor (ADR) blocker and vasodilator] against AD through ameliorating vascular abnormalities. Our in vitro data revealed that NG could reverse β-amyloid1-42 (Aβ1-42)-induced PKC/ERK1/2 activation, the downstream pathway of α1-ADR activation, in α1-ADR-overexpressed N2a cells. NG also blocked Aβ1-42- or phenylephrine-induced constrictions in isolated rat arteries. All these in vitro data may suggest ADR-dependent impacts of Aβ on vascular function and the reversal effect of NG. In addition, the ameliorating impacts of NG treatment on cerebral vasoconstriction, vasoremodeling, and cognitive decline were investigated in vivo in a PSAPP transgenic AD mouse model. Consistent with in vitro findings, the chronic treatment of NG significantly ameliorated the cerebrovascular dysfunctions and Aβ plaque depositions in the brain. Moreover, an improved cognitive performance was also observed. Taken together, our findings supported the beneficial effects of NG on AD through adrenergic-related mechanisms and highlighted the therapeutic potential of α1-adrenergic vasomodulators against AD pathologies.
Collapse
Affiliation(s)
- Xidong Pan
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
- Emergency Department, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Zhifeng Lei
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
| | - Jiang Chen
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
| | - Congcong Jia
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Institute of Brain Science and Brain-Inspired Research, Jinan 271016, China
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - Jie Deng
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
| | - Ying Liu
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
| | - Xingmei Luo
- Comprehensive Ward, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Likun Wang
- Emergency Department, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Dan Zi
- Department of Obstetrics and Gynecology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550004, China
| | - Zhen Wang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Song Li
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - Jun Tan
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang 550004, China
- Institute of Translational Medicine; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China
| |
Collapse
|
14
|
Khan S, Hussain R, Iqbal T, Rahim F, Khan Y. Recent development and strategies towards target interactions: Synthesis, characterization and in silico analysis of benzimidazole based thiadiazole as potential anti-Alzheimer agents. Biochem Biophys Res Commun 2024; 726:150201. [PMID: 38924881 DOI: 10.1016/j.bbrc.2024.150201] [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: 02/26/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024]
Abstract
In the current research study, we aim to design and synthesize highly potent hybrid analogs of benzimidazole derived thiadiazole based Schiff base derivatives which can combat the cholinesterase enzymes (acetylcholinesterase and butyrylcholinesterase) accountable for developing Alzheimer's disease. In this context, we have synthesized 15 analogs of benzimidazole based thiadiazole derivatives, which were subsequently confirmed through spectroscopic techniques including 1H NMR, 13C NMR and HREI-MS. Biological investigation of all the analogs revealed their varied acetylcholinesterase inhibitory potency covering a range between 3.20 ± 0.10 μM to 20.50 ± 0.20 μM as well as butyrylcholinesterase inhibitory potential with a range of 4.30 ± 0.50 μM to 20.70 ± 0.50 μM when compared with the standard drug Donepezil having IC50 = 6.70 ± 0.20 μM for AChE and 7.90 ± 0.10 μM for BuChE. The promising inhibition by the analogs was evaluated in SAR analysis, where analog-1 (IC50 = 3.20 ± 0.10 μM for AChE and 4.30 ± 0.50 μM for BuChE), analog-4 (IC50 = 4.30 ± 0.30 μM for AChE and 5.50 ± 0.20 μM for BuChE) and analog-5 (IC50 = 4.10 ± 0.30 μM for AChE and 4.60 ± 0.40 μM for BuChE) were found as the lead candidates. Moreover, molecular docking and ADME analysis were conducted to explore the better binding interactions and drugs likeness respectively.
Collapse
Affiliation(s)
- Shoaib Khan
- Department of Chemistry, Abbottabad University of Science and Technology, Abbottabad, 22500, Pakistan.
| | - Rafaqat Hussain
- Department of Chemistry, Hazara University, Mansehra, 21120, Pakistan.
| | - Tayyiaba Iqbal
- Department of Chemistry, Abbottabad University of Science and Technology, Abbottabad, 22500, Pakistan
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra, 21120, Pakistan
| | - Yousaf Khan
- Department of Chemistry, COMSATS University Islamabad, 45550, Islamabad, Pakistan
| |
Collapse
|
15
|
Khan AF, Iturria-Medina Y. Beyond the usual suspects: multi-factorial computational models in the search for neurodegenerative disease mechanisms. Transl Psychiatry 2024; 14:386. [PMID: 39313512 PMCID: PMC11420368 DOI: 10.1038/s41398-024-03073-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 08/20/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
From Alzheimer's disease to amyotrophic lateral sclerosis, the molecular cascades underlying neurodegenerative disorders remain poorly understood. The clinical view of neurodegeneration is confounded by symptomatic heterogeneity and mixed pathology in almost every patient. While the underlying physiological alterations originate, proliferate, and propagate potentially decades before symptomatic onset, the complexity and inaccessibility of the living brain limit direct observation over a patient's lifespan. Consequently, there is a critical need for robust computational methods to support the search for causal mechanisms of neurodegeneration by distinguishing pathogenic processes from consequential alterations, and inter-individual variability from intra-individual progression. Recently, promising advances have been made by data-driven spatiotemporal modeling of the brain, based on in vivo neuroimaging and biospecimen markers. These methods include disease progression models comparing the temporal evolution of various biomarkers, causal models linking interacting biological processes, network propagation models reproducing the spatial spreading of pathology, and biophysical models spanning cellular- to network-scale phenomena. In this review, we discuss various computational approaches for integrating cross-sectional, longitudinal, and multi-modal data, primarily from large observational neuroimaging studies, to understand (i) the temporal ordering of physiological alterations, i(i) their spatial relationships to the brain's molecular and cellular architecture, (iii) mechanistic interactions between biological processes, and (iv) the macroscopic effects of microscopic factors. We consider the extents to which computational models can evaluate mechanistic hypotheses, explore applications such as improving treatment selection, and discuss how model-informed insights can lay the groundwork for a pathobiological redefinition of neurodegenerative disorders.
Collapse
Affiliation(s)
- Ahmed Faraz Khan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada
| | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
- McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada.
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada.
| |
Collapse
|
16
|
Yi F, Wang J, Lin M, Li B, Han S, Wang S, Jin Y, Hu N, Chen Y, Shang X. Correspondence between white matter hyperintensities and regional grey matter volumes in Alzheimer's disease. Front Aging Neurosci 2024; 16:1429098. [PMID: 39351014 PMCID: PMC11439820 DOI: 10.3389/fnagi.2024.1429098] [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: 05/07/2024] [Accepted: 08/30/2024] [Indexed: 10/04/2024] Open
Abstract
Objective White matter hyperintensities (WMH) are the most common neuroimaging manifestation of cerebral small vessel disease, and is frequently observed in Alzheimer's disease (AD). This study aimed to investigate the relationship between WMH and cognition and to verify the mediation of grey matter atrophy in this relationship. Methods The diffusion tensor imaging (DTI) technique analyses white matter fiber tract to assess white matter integrity. Voxel-based morphometry was applied to measure the grey matter volume (GMV). A linear regression model was applied to examine the associations between WMH and GMV, and mediation analyses was performed to determine the mediating role of regional GMV in the effect of WMH on cognitive function. Results Compared to the HC group, AD group have 8 fiber tract fractional anisotropy (FA) decreased and 16 fiber tract mean diffusivity (MD) increased. Compared to AD without WMH, AD with high WMH had 9 fiber tracts FA decreased and 13 fiber tracts MD increased. High WMH volume was negatively correlated with GMV in the frontal-parietal region. Low WMH volume was also negatively correlated with GMV except for the three regions (right angular gyrus, right superior frontal gyrus and right middle/inferior parietal gyrus), where GMV was positively correlated. Mediation analysis showed that the association between WMH and executive function or episodic memory were mediated by GMV in the frontal-parietal region. Conclusion Damage to white matter integrity was more severe in AD with WMH. Differential changes in DTI metrics may be caused by progressive myelin and axonal damage. There was a negative correlation between WMH and grey matter atrophy in frontal-parietal regions in a volume-dependent manner. This study indicates the correspondence between WMH volume and GMV in cognition, and GMV being a key modulator between WMH and cognition in AD. This result will contribute to understanding the progression of the disease process and applying targeted therapeutic intervention in the earlier stage to delay neurodegenerative changes in frontal-parietal regions to achieve better treatment outcomes and affordability.
Collapse
Affiliation(s)
- Fangyuan Yi
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Neurology, Jin Qiu Hospital of Liaoning Province (Geriatric Hospital of Liaoning Province), Shenyang, China
| | - Jirui Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Meiqing Lin
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Baizhu Li
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shiyu Han
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shan Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yingbin Jin
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ning Hu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yutong Chen
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiuli Shang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
17
|
Othman MS, Hussain R, Rahim F, Ullah H, Khan S, Taha M, Fareid MA, Altaleb AT, Aboelnaga SM, Shah SAA. Synthesis, biological and computational evaluation of benzoxazole hybrid analogs as potential anti-Alzheimer's agents. Future Med Chem 2024; 16:2013-2023. [PMID: 39269160 PMCID: PMC11485858 DOI: 10.1080/17568919.2024.2393569] [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: 04/23/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Aim: Current study aims exploration of bis-benzoxazole bearing bis-Schiff base scaffolds (1-16) as anti-Alzheimer's agents.Materials & methods: 2-aminophenol is used as starting materials which react with different reagents in different step to give us bis-benzoxazole bearing bis-Schiff base analogs. NMR and HREI-MS techniques were used for characterization. All derivatives demonstrated varied range of activities with IC50 values 1.10 ± 0.40-24.50 ± 0.90 μM against acetylcholinesterase (AChE) and 1.90 ± 0.70-28.60 ± 0.60 μM against butyrylcholinesterase (BuChE) in contrast to donepezil. In both cases, analog-3 was found most potent. Molecular docking explored modes of interactions between scaffolds and receptor sites of targeted enzymes.Conclusion: This study offering promising approach for optimization and development of potent inhibitors of cholinesterase enzymes.
Collapse
Affiliation(s)
- Mohamed S Othman
- Biochemistry Department, College of Medicine, University of Ha'il, Hail, 2440, Saudi Arabia
- Faculty of Biotechnology, October University for Modern Science & Arts (MSA), Giza, Egypt
| | - Rafaqat Hussain
- Department of Chemistry, Hazara University, Mansehra, 21120, Pakistan
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra, 21120, Pakistan
| | - Hayat Ullah
- Institute of Chemistry, University of Okara, Okara-56300, Pakistan
| | - Shoaib Khan
- Department of Chemistry, Abbottabad University of Science & Technology (AUST), Abbottabad, Pakistan
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research & Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Mohamed A Fareid
- Department of Medical Laboratories, Faculty of Applied Medical Science, University of Ha'il, Hail, 2440, Saudi Arabia
- Botany & Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, 11651, Egypt
| | - Anas T Altaleb
- Basic Sciences Department, Deanship of Preparatory Year, University of Ha'il, Hail, 2440, Saudi Arabia
| | - Shimaa M Aboelnaga
- Basic Sciences Department, Deanship of Preparatory Year, University of Ha'il, Hail, 2440, Saudi Arabia
| | - Syed Adnan Ali Shah
- Faculty of Pharmacy, Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam, Selangor, 42300, Malaysia
- Atta-ur-Rahman Institute for Natural Product Discovery (AuRIns), Universityi Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam, Selangor, 42300, Malaysia
| |
Collapse
|
18
|
Pandit M, Frishman WH. The Association Between Cardiovascular Disease and Dementia: A Review of Trends in Epidemiology, Risk Factors, Pathophysiologic Mechanisms, and Clinical Implications. Cardiol Rev 2024; 32:463-467. [PMID: 36946920 DOI: 10.1097/crd.0000000000000523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
With increases in life expectancy and the size of the aging population, cognitive decline and neurodegenerative pathologies are expected to increase in the next few decades. Age-related increases in risk for dementia and cardiovascular disease have been researched widely. Epidemiology trends reveal a predicted increase of neurodegenerative disease to more than 65 million by 2030 in the United States. There are several risk factors for the development of cardiovascular disease that have been widely studied for their impact on dementia; such as: diabetes, hypertension, and hyperlipidemia. Several pathophysiologic mechanisms exist by which cardiovascular disease could impact dementia including cerebral hypoperfusion, reactive oxidative species, and increased cleavage of amyloid precursor protein into amyloid beta plaques and accumulation of neurofibrillary tangles. Emerging evidence also suggests that treatment of cardiovascular disease risk factors could reduce the risk of dementia development. In this review, we seek to examine the relationship between cardiovascular disease and dementia by examining epidemiologic trends, common risk factors, pathophysiologic mechanisms and implications for clinical management.
Collapse
Affiliation(s)
- Maya Pandit
- From the New York Medical College, Valhalla, NY
| | - William H Frishman
- Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| |
Collapse
|
19
|
Li P, Kianmehr H, Guan D, Kulshreshtha A, Narayan KV, Ali MK, Umpierrez GE, Hu X, Fonseca VA, Shi L, Shao H. Renal function as an effect modifier of intensive glucose control in delaying cognitive function decline among individuals with type 2 diabetes: A revisit to the ACCORD MIND trial. Diabetes Obes Metab 2024; 26:3958-3968. [PMID: 38962812 PMCID: PMC11300148 DOI: 10.1111/dom.15744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/04/2024] [Accepted: 06/11/2024] [Indexed: 07/05/2024]
Abstract
AIM Dysglycaemia accelerates cognitive decline. Intensive glucose control may help delay or prevent cognitive function decline (CFD). We aimed to determine how patient characteristics influence the effect of intensive glucose control [glycated haemoglobin (HbA1c) <6.0%] on delaying CFD in people with type 2 diabetes. RESEARCH DESIGN AND METHODS In this post-hoc analysis of 2977 type 2 diabetes participants from the ACCORD MIND trial, we applied the causal forest and causal tree algorithms to identify the effect modifier of intensive glucose control in delaying CFD from 68 variables (demographics, disease history, medications, vitals and baseline biomarkers). The exposure was intensive versus standard glucose control (HbA1c <6.0% vs. 7.0%-7.9%). The main outcome was cognitive function changes from baseline to the 40th month follow-up, which were evaluated using the digit symbol substitution test, Rey auditory verbal learning test, mini-mental state examination and Stroop test. We used Cohen's d, a measure of standardized difference, to quantify the effect size of intensive glucose control on delaying CFD. RESULTS Among all the baseline characteristics, renal function was the most significant effect modifier. Participants with urinary albumin levels <0.4 mg/dl [absolute function change (AFC): 0.51 in mini-mental state examination, 95% confidence interval (CI): 0.04, 0.98, Cohen's d: 0.25] had slower CFD with intensive glucose control. Patients with preserved renal function (estimated glomerular filtration rate between 60 and 90 ml/min/1.73 m2) were associated with small benefits (AFC: 1.28 in Stroop, 95% CI: 0.28, 2.27, Cohen's d: 0.12) when undergoing intensive glucose control. Conversely, participants with an estimated glomerular filtration rate <60 ml/min/1.73 m2 (AFC: -0.57 in the Rey auditory verbal learning test, 95% CI: -1.09, -0.05, Cohen's d: -0.30) exhibited faster CFD when undergoing intensive glucose control. Participants who were <60 years old showed a significant benefit from intensive glucose control in delaying CFD (AFC: 1.08 in the digit symbol substitution test, 95% CI: 0.06, 2.10, Cohen's d: 0.13). All p < .05. CONCLUSIONS Our findings linked renal function with the benefits of intensive glucose control in delaying CFD, informing personalized HbA1c goals for those with diabetes and at risk of CFD.
Collapse
Affiliation(s)
- Piaopiao Li
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Hamed Kianmehr
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Dawei Guan
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Ambar Kulshreshtha
- Department of Family and Preventive Medicine, School of Medicine, Emory University, Atlanta, GA
| | - K.M. Venkat Narayan
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Mohammed K Ali
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA
- Department of Family and Preventive Medicine, School of Medicine, Emory University, Atlanta, GA
| | | | - Xin Hu
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlotteville, VA
| | - Vivian A. Fonseca
- Department of Medicine and Pharmacology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Lizheng Shi
- Department of Health Policy and Management, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Hui Shao
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, FL, USA
- Department of Family and Preventive Medicine, School of Medicine, Emory University, Atlanta, GA
| |
Collapse
|
20
|
Jakubowski H. Homocysteine Thiolactone Detoxifying Enzymes and Alzheimer's Disease. Int J Mol Sci 2024; 25:8095. [PMID: 39125665 PMCID: PMC11312131 DOI: 10.3390/ijms25158095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Elevated levels of homocysteine (Hcy) and related metabolites are associated with Alzheimer's disease (AD). Severe hyperhomocysteinemia causes neurological deficits and worsens behavioral and biochemical traits associated with AD. Although Hcy is precluded from entering the Genetic Code by proofreading mechanisms of aminoacyl-tRNA synthetases, and thus is a non-protein amino acid, it can be attached to proteins via an N-homocysteinylation reaction mediated by Hcy-thiolactone. Because N-homocysteinylation is detrimental to a protein's function and biological integrity, Hcy-thiolactone-detoxifying enzymes-PON1, BLMH, BPHL-have evolved. This narrative review provides an account of the biological function of these enzymes and of the consequences of their impairments, leading to the phenotype characteristic of AD. Overall, accumulating evidence discussed in this review supports a hypothesis that Hcy-thiolactone contributes to neurodegeneration associated with a dysregulated Hcy metabolism.
Collapse
Affiliation(s)
- Hieronim Jakubowski
- Department of Biochemistry and Biotechnology, University of Life Sciences, 60-637 Poznań, Poland; ; Tel.: +48-973-972-8733; Fax: +48-973-972-8981
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, International Center for Public Health, Newark, NJ 07103, USA
| |
Collapse
|
21
|
O’Hare N, Millican K, Ebong EE. Unraveling neurovascular mysteries: the role of endothelial glycocalyx dysfunction in Alzheimer's disease pathogenesis. Front Physiol 2024; 15:1394725. [PMID: 39027900 PMCID: PMC11254711 DOI: 10.3389/fphys.2024.1394725] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/27/2024] [Indexed: 07/20/2024] Open
Abstract
While cardiovascular disease, cancer, and human immunodeficiency virus (HIV) mortality rates have decreased over the past 20 years, Alzheimer's Disease (AD) deaths have risen by 145% since 2010. Despite significant research efforts, effective AD treatments remain elusive due to a poorly defined etiology and difficulty in targeting events that occur too downstream of disease onset. In hopes of elucidating alternative treatment pathways, now, AD is commonly being more broadly defined not only as a neurological disorder but also as a progression of a variety of cerebrovascular pathologies highlighted by the breakdown of the blood-brain barrier. The endothelial glycocalyx (GCX), which is an essential regulator of vascular physiology, plays a crucial role in the function of the neurovascular system, acting as an essential vascular mechanotransducer to facilitate ultimate blood-brain homeostasis. Shedding of the cerebrovascular GCX could be an early indication of neurovascular dysfunction and may subsequently progress neurodegenerative diseases like AD. Recent advances in in vitro modeling, gene/protein silencing, and imaging techniques offer new avenues of scrutinizing the GCX's effects on AD-related neurovascular pathology. Initial studies indicate GCX degradation in AD and other neurodegenerative diseases and have begun to demonstrate a possible link to GCX loss and cerebrovascular dysfunction. This review will scrutinize the GCX's contribution to known vascular etiologies of AD and propose future work aimed at continuing to uncover the relationship between GCX dysfunction and eventual AD-associated neurological deterioration.
Collapse
Affiliation(s)
- Nicholas O’Hare
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Karina Millican
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Eno E. Ebong
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
- Department of Bioengineering, Northeastern University, Boston, MA, United States
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
22
|
Burtscher J, Millet GP, Fresa M, Lanzi S, Mazzolai L, Pellegrin M. The link between impaired oxygen supply and cognitive decline in peripheral artery disease. Prog Cardiovasc Dis 2024; 85:63-73. [PMID: 38061613 DOI: 10.1016/j.pcad.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 12/26/2023]
Abstract
Although peripheral artery disease (PAD) primarily affects large arteries outside the brain, PAD is also associated with elevated cerebral vulnerabilities, including greater risks for brain injury (such as stroke), cognitive decline and dementia. In the present review, we aim to evaluate recent literature and extract information on potential mechanisms linking PAD and consequences on the brain. Furthermore, we suggest novel therapeutic avenues to mitigate cognitive decline and reduce risk of brain injury in patients with PAD. Various interventions, notably exercise, directly or indirectly improve systemic blood flow and oxygen supply and are effective strategies in patients with PAD or cognitive decline. Moreover, triggering protective cellular and systemic mechanisms by modulating inspired oxygen concentrations are emerging as potential novel treatment strategies. While several genetic and pharmacological approaches to modulate adaptations to hypoxia showed promising results in preclinical models of PAD, no clear benefits have yet been clinically demonstrated. We argue that genetic/pharmacological regulation of the involved adaptive systems remains challenging but that therapeutic variation of inspired oxygen levels (e.g., hypoxia conditioning) are promising future interventions to mitigate associated cognitive decline in patients with PAD.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland.
| | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Marco Fresa
- Angiology Department, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Stefano Lanzi
- Angiology Department, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Lucia Mazzolai
- Angiology Department, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Maxime Pellegrin
- Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland; Angiology Department, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland.
| |
Collapse
|
23
|
Gaire BP, Koronyo Y, Fuchs DT, Shi H, Rentsendorj A, Danziger R, Vit JP, Mirzaei N, Doustar J, Sheyn J, Hampel H, Vergallo A, Davis MR, Jallow O, Baldacci F, Verdooner SR, Barron E, Mirzaei M, Gupta VK, Graham SL, Tayebi M, Carare RO, Sadun AA, Miller CA, Dumitrascu OM, Lahiri S, Gao L, Black KL, Koronyo-Hamaoui M. Alzheimer's disease pathophysiology in the Retina. Prog Retin Eye Res 2024; 101:101273. [PMID: 38759947 PMCID: PMC11285518 DOI: 10.1016/j.preteyeres.2024.101273] [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: 02/11/2023] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
The retina is an emerging CNS target for potential noninvasive diagnosis and tracking of Alzheimer's disease (AD). Studies have identified the pathological hallmarks of AD, including amyloid β-protein (Aβ) deposits and abnormal tau protein isoforms, in the retinas of AD patients and animal models. Moreover, structural and functional vascular abnormalities such as reduced blood flow, vascular Aβ deposition, and blood-retinal barrier damage, along with inflammation and neurodegeneration, have been described in retinas of patients with mild cognitive impairment and AD dementia. Histological, biochemical, and clinical studies have demonstrated that the nature and severity of AD pathologies in the retina and brain correspond. Proteomics analysis revealed a similar pattern of dysregulated proteins and biological pathways in the retina and brain of AD patients, with enhanced inflammatory and neurodegenerative processes, impaired oxidative-phosphorylation, and mitochondrial dysfunction. Notably, investigational imaging technologies can now detect AD-specific amyloid deposits, as well as vasculopathy and neurodegeneration in the retina of living AD patients, suggesting alterations at different disease stages and links to brain pathology. Current and exploratory ophthalmic imaging modalities, such as optical coherence tomography (OCT), OCT-angiography, confocal scanning laser ophthalmoscopy, and hyperspectral imaging, may offer promise in the clinical assessment of AD. However, further research is needed to deepen our understanding of AD's impact on the retina and its progression. To advance this field, future studies require replication in larger and diverse cohorts with confirmed AD biomarkers and standardized retinal imaging techniques. This will validate potential retinal biomarkers for AD, aiding in early screening and monitoring.
Collapse
Affiliation(s)
- Bhakta Prasad Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ron Danziger
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jean-Philippe Vit
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jonah Doustar
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Miyah R Davis
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ousman Jallow
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Filippo Baldacci
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | | | - Ernesto Barron
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Vivek K Gupta
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Stuart L Graham
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia; Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Roxana O Carare
- Department of Clinical Neuroanatomy, University of Southampton, Southampton, UK
| | - Alfredo A Sadun
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Carol A Miller
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Shouri Lahiri
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Liang Gao
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Keith L Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Moazzami K, Kulshreshtha A, Gold M, Rahbar A, Goldstein F, Shah AJ, Bremner JD, Vaccarino V, Quyyumi AA. Hemodynamic Reactivity to Mental Stress and Cognitive Function in Coronary Artery Disease. Psychosom Med 2024; 86:498-506. [PMID: 38648028 PMCID: PMC11270642 DOI: 10.1097/psy.0000000000001314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
OBJECTIVE People with coronary artery disease (CAD) are at higher risk of cognitive impairment than those without CAD. Psychological stress is a risk factor for both conditions, and assessing the hemodynamic reactivity to mental stress could explain the link between stress and cognitive function. METHODS A total of 779 individuals with stable CAD from two prospective cohort studies were included. All individuals underwent acute mental stress testing, as well as conventional stress testing. Cognitive function was assessed both at baseline and at a 2-year follow-up. The rate-pressure product (RPP) was calculated as the mean systolic blood pressure times the mean heart rate at rest. RPP reactivity was defined as the maximum RPP during standardized mental stress test minus the RPP at rest. RESULTS After multivariable adjustment, every standard deviation decrease in RPP reactivity with mental stress was associated with slower completion of Trail-A and Trail-B in both cohorts (13% and 11% in cohort 1, and 15% and 16% in cohort 2, respectively; p for all <.01). After a 2-year follow-up period, every standard deviation decrease in RPP reactivity with mental stress was associated with a 8% and 9% slower completion of Trail-A and Trail-B, respectively ( p for all <.01). There was no significant association between RPP reactivity with conventional stress testing and any of the cognitive tests. CONCLUSION In the CAD population, a blunted hemodynamic response to mental stress is associated with slower visuomotor processing and worse executive function at baseline and with greater decline in these abilities over time.
Collapse
Affiliation(s)
- Kasra Moazzami
- From the Division of Cardiology, Department of Medicine (Moazzami, Gold, Rahbar, Quyyumi), Emory Clinical Cardiovascular Research Institute; Grady Health System (Moazzami); Department of Epidemiology, Rollins School of Public Health (Kulshreshtha, Shah, Vaccarino), Emory University; Departments of Family and Preventive Medicine (Kulshreshtha) and Neurology (Goldstein), and Goizuetta Alzheimer's Disease Research Center (Goldstein), Emory University School of Medicine, Emory University, Atlanta; Atlanta VA Medical Center (Shah, Bremner), Decatur; Departments of Radiology and Imaging Sciences (Bremner) and Psychiatry and Behavioral Sciences (Bremner), Emory University School of Medicine, Atlanta, Georgia
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Beinlich FR, Asiminas A, Untiet V, Bojarowska Z, Plá V, Sigurdsson B, Timmel V, Gehrig L, Graber MH, Hirase H, Nedergaard M. Oxygen imaging of hypoxic pockets in the mouse cerebral cortex. Science 2024; 383:1471-1478. [PMID: 38547288 PMCID: PMC11251491 DOI: 10.1126/science.adn1011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/23/2024] [Indexed: 04/02/2024]
Abstract
Consciousness is lost within seconds upon cessation of cerebral blood flow. The brain cannot store oxygen, and interruption of oxidative phosphorylation is fatal within minutes. Yet only rudimentary knowledge exists regarding cortical partial oxygen tension (Po2) dynamics under physiological conditions. Here we introduce Green enhanced Nano-lantern (GeNL), a genetically encoded bioluminescent oxygen indicator for Po2 imaging. In awake behaving mice, we uncover the existence of spontaneous, spatially defined "hypoxic pockets" and demonstrate their linkage to the abrogation of local capillary flow. Exercise reduced the burden of hypoxic pockets by 52% compared with rest. The study provides insight into cortical oxygen dynamics in awake behaving animals and concurrently establishes a tool to delineate the importance of oxygen tension in physiological processes and neurological diseases.
Collapse
Affiliation(s)
- Felix R.M. Beinlich
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
| | - Antonios Asiminas
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
| | - Verena Untiet
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
| | - Zuzanna Bojarowska
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
| | - Virginia Plá
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
| | - Björn Sigurdsson
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
| | - Vincenzo Timmel
- School of Engineering, FHNW University of Applied Sciences and Arts Northwestern Switzerland; 5210 Windisch, Switzerland
| | - Lukas Gehrig
- School of Engineering, FHNW University of Applied Sciences and Arts Northwestern Switzerland; 5210 Windisch, Switzerland
| | - Michael H. Graber
- School of Engineering, FHNW University of Applied Sciences and Arts Northwestern Switzerland; 5210 Windisch, Switzerland
| | - Hajime Hirase
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Rochester Medical Center; Rochester, NY 14642, USA
| | - Maiken Nedergaard
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen; 2200 Copenhagen, Denmark
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Rochester Medical Center; Rochester, NY 14642, USA
| |
Collapse
|
26
|
Ho K, Bodi NE, Sharma TP. Normal-Tension Glaucoma and Potential Clinical Links to Alzheimer's Disease. J Clin Med 2024; 13:1948. [PMID: 38610712 PMCID: PMC11012506 DOI: 10.3390/jcm13071948] [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: 02/19/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Glaucoma is a group of optic neuropathies and the world's leading cause of irreversible blindness. Normal-tension glaucoma (NTG) is a subtype of glaucoma that is characterized by a typical pattern of peripheral retinal loss, in which the patient's intraocular pressure (IOP) is considered within the normal range (<21 mmHg). Currently, the only targetable risk factor for glaucoma is lowering IOP, and patients with NTG continue to experience visual field loss after IOP-lowering treatments. This demonstrates the need for a better understanding of the pathogenesis of NTG and underlying mechanisms leading to neurodegeneration. Recent studies have found significant connections between NTG and cerebral manifestations, suggesting NTG as a neurodegenerative disease beyond the eye. Gaining a better understanding of NTG can potentially provide new Alzheimer's Disease diagnostics capabilities. This review identifies the epidemiology, current biomarkers, altered fluid dynamics, and cerebral and ocular manifestations to examine connections and discrepancies between the mechanisms of NTG and Alzheimer's Disease.
Collapse
Affiliation(s)
- Kathleen Ho
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Nicole E. Bodi
- Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Tasneem P. Sharma
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| |
Collapse
|
27
|
Shang J, Dong J, Zhu S, Chen Q, Hua J. Trends in cognitive function before and after myocardial infarction: findings from the China Health and Retirement Longitudinal Study. Front Aging Neurosci 2024; 16:1283997. [PMID: 38455665 PMCID: PMC10917921 DOI: 10.3389/fnagi.2024.1283997] [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: 08/27/2023] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
Objectives Incident stroke was associated with cognitive dysfunction after stroke and even before stroke. However, cognitive trends prior to myocardial infarction (MI) and the timeline of cognitive decline in a few years following incident MI remain unclear, especially among the Chinese population. We aimed to evaluate whether MI was associated with cognitive change both before and after MI in China. Methods This cohort study included 11,287 participants without baseline heart problems or stroke from the China Health and Retirement Longitudinal Study. The exposure was self-reported MI. The outcomes were scores of cognitive functions in five domains, which reflected abilities of episodic memory, visuospatial abilities, orientation, attention and calculation, and global cognition as a summary measure. A Linear mixed model was constructed to explore cognitive function before and after incident MI among the MI participants and the cognitive trends of participants free of MI. Results During the 7-year follow-up, 421 individuals [3.7% of 11,287, mean (SD) age, 60.0 (9.0) years; 59.1% female] experienced MI events. The cognitive scores of participants of both the MI group and the control group without MI declined gradually as time went by. The annual decline rate of the MI group before incident MI was similar to that of the control group during the whole follow-up period. Incident MI was not associated with acute cognitive decline in all five cognitive domains. Moreover, MI did not accelerate the cognitive decline rate after MI compared with the pre-MI cognitive trends. The decline rate of cognitive function after MI was similar to the rate before MI. Conclusions Different from stroke, participants who had an MI did not show steeper cognitive decline before MI. MI was not associated with acute cognitive decline and accelerated decline in several years after MI. Future studies are needed to learn the mechanisms behind the different patterns of cognitive decline between MI and stroke.
Collapse
Affiliation(s)
- Jing Shang
- Department of Psychiatry, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jianye Dong
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Sijia Zhu
- Department of Neurology, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, China
| | - Qingmei Chen
- Department of Physical Medicine and Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jianian Hua
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
28
|
Jiang Z, Wang J, Qin Y, Liu S, Luo B, Bai F, Wei H, Zhang S, Wei J, Ding G, Ma L, He S, Chen R, Sun Y, Chen Y, Wang L, Xu H, Wang X, Chen G, Lei W. A nonhuman primate model with Alzheimer's disease-like pathology induced by hippocampal overexpression of human tau. Alzheimers Res Ther 2024; 16:22. [PMID: 38281031 PMCID: PMC10821564 DOI: 10.1186/s13195-024-01392-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/15/2024] [Indexed: 01/29/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is one of the most burdening diseases of the century with no disease-modifying treatment at this time. Nonhuman primates (NHPs) share genetic, anatomical, and physiological similarities with humans, making them ideal model animals for investigating the pathogenesis of AD and potential therapies. However, the use of NHPs in AD research has been hindered by the paucity of AD monkey models due to their long generation time, ethical considerations, and technical challenges in genetically modifying monkeys. METHODS Here, we developed an AD-like NHP model by overexpressing human tau in the bilateral hippocampi of adult rhesus macaque monkeys. We evaluated the pathological features of these monkeys with immunostaining, Nissl staining, cerebrospinal fluid (CSF) analysis, magnetic resonance imaging (MRI), positron emission tomography (PET), and behavioural tests. RESULTS We demonstrated that after hippocampal overexpression of tau protein, these monkeys displayed multiple pathological features of AD, including 3-repeat (3R)/4-repeat (4R) tau accumulation, tau hyperphosphorylation, tau propagation, neuronal loss, hippocampal atrophy, neuroinflammation, Aβ clearance deficits, blood vessel damage, and cognitive decline. More interestingly, the accumulation of both 3R and 4R tau is specific to NHPs but not found in adult rodents. CONCLUSIONS This work establishes a tau-induced AD-like NHP model with many key pathological and behavioural features of AD. In addition, our model may potentially become one of the AD NHP models adopted by researchers worldwide since it can be generated within 2 ~ 3 months through a single injection of AAVs into the monkey brains. Hence, our model NHPs may facilitate mechanistic studies and therapeutic treatments for AD.
Collapse
Affiliation(s)
- Zhouquan Jiang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Jing Wang
- Department of Neurosurgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Yongpeng Qin
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Shanggong Liu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Bin Luo
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Fan Bai
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Huiyi Wei
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Shaojuan Zhang
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Junjie Wei
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Guoyu Ding
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Long Ma
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Shu He
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Rongjie Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Ying Sun
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Yi Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Lu Wang
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Hao Xu
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Xiangyu Wang
- Department of Neurosurgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Gong Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China.
| | - Wenliang Lei
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
29
|
Cummings TH, Magagnoli J, Sikirzhytskaya A, Tyagin I, Safro I, Wyatt MD, Shtutman M, Sutton SS. Exposure to angiotensin-converting enzyme inhibitors that cross the blood-brain barrier and the risk of dementia among patients with human immunodeficiency virus. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.16.24301275. [PMID: 38293017 PMCID: PMC10827249 DOI: 10.1101/2024.01.16.24301275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
More than one million people in the United States and over 38 million people worldwide are living with human immunodeficiency virus (HIV) infection. Antiretroviral therapy (ART) greatly improves the health of people living with HIV (PLWH); however, the increased life longevity of PLWH has revealed consequences of HIV-associated comorbidities. HIV can enter the brain and cause inflammation even in individuals with well-controlled HIV infection. The quality of life for PLWH can be compromised by cognitive deficits and memory loss, termed HIV-associated neurological disorders (HAND). HIV-associated dementia is a related but distinct diagnosis. Common causes of dementia in PLWH are similar to the general population and can affect cognition. There is an urgent need to identify treatments for the aging PWLH population. We previously developed AI-based biomedical literature mining systems to uncover a potential novel connection between HAND the renin-angiotensin system (RAAS), which is a pharmacological target for hypertension. RAAS-targeting anti-hypertensives are gaining attention for their protective benefits in several neurocognitive disorders. To our knowledge, the effect of RAAS-targeting drugs on the cognition of PLWH development of dementia has not previously been analyzed. We hypothesized that exposure to angiotensin-converting enzyme inhibitors (ACEi) that cross the blood brain barrier (BBB) reduces the risk/occurrence of dementia in PLWH. We report a retrospective cohort study of electronic health records (EHRs) to examine the proposed hypothesis using data from the United States Department of Veterans Affairs, in which a primary outcome of dementia was measured in controlled cohorts of patients exposed to BBB-penetrant ACEi versus those unexposed to BBB-penetrant ACEi. The results reveal a statistically significant reduction in dementia diagnosis for PLWH exposed to BBB-penetrant ACEi. These results suggest there is a potential protective effect of BBB ACE inhibitor exposure against dementia in PLWH that warrants further investigation.
Collapse
Affiliation(s)
- Tammy H Cummings
- Dorn Research Institute, Columbia Veterans Affairs Health Care System, Columbia, South Carolina
- College of Pharmacy, University of South Carolina, Department of Clinical Pharmacy & Outcomes Sciences
| | - Joseph Magagnoli
- Dorn Research Institute, Columbia Veterans Affairs Health Care System, Columbia, South Carolina
- College of Pharmacy, University of South Carolina, Department of Clinical Pharmacy & Outcomes Sciences
| | | | - Ilya Tyagin
- Department of Computer and Information Sciences, University of Delaware
| | - Ilya Safro
- Department of Computer and Information Sciences, University of Delaware
| | - Michael D Wyatt
- College of Pharmacy, University of South Carolina, Department of Drug Discovery & Biomedical Sciences
| | - Michael Shtutman
- College of Pharmacy, University of South Carolina, Department of Drug Discovery & Biomedical Sciences
| | - S Scott Sutton
- Dorn Research Institute, Columbia Veterans Affairs Health Care System, Columbia, South Carolina
- College of Pharmacy, University of South Carolina, Department of Clinical Pharmacy & Outcomes Sciences
| |
Collapse
|
30
|
Irwin C, Tjandra D, Hu C, Aggarwal V, Lienau A, Giordani B, Wiens J, Migrino RQ. Predicting 5-year dementia conversion in veterans with mild cognitive impairment. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12572. [PMID: 38545542 PMCID: PMC10965752 DOI: 10.1002/dad2.12572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/29/2024] [Accepted: 02/18/2024] [Indexed: 11/11/2024]
Abstract
INTRODUCTION Identifying mild cognitive impairment (MCI) patients at risk for dementia could facilitate early interventions. Using electronic health records (EHRs), we developed a model to predict MCI to all-cause dementia (ACD) conversion at 5 years. METHODS Cox proportional hazards model was used to identify predictors of ACD conversion from EHR data in veterans with MCI. Model performance (area under the receiver operating characteristic curve [AUC] and Brier score) was evaluated on a held-out data subset. RESULTS Of 59,782 MCI patients, 15,420 (25.8%) converted to ACD. The model had good discriminative performance (AUC 0.73 [95% confidence interval (CI) 0.72-0.74]), and calibration (Brier score 0.18 [95% CI 0.17-0.18]). Age, stroke, cerebrovascular disease, myocardial infarction, hypertension, and diabetes were risk factors, while body mass index, alcohol abuse, and sleep apnea were protective factors. DISCUSSION EHR-based prediction model had good performance in identifying 5-year MCI to ACD conversion and has potential to assist triaging of at-risk patients. Highlights Of 59,782 veterans with mild cognitive impairment (MCI), 15,420 (25.8%) converted to all-cause dementia within 5 years.Electronic health record prediction models demonstrated good performance (area under the receiver operating characteristic curve 0.73; Brier 0.18).Age and vascular-related morbidities were predictors of dementia conversion.Synthetic data was comparable to real data in modeling MCI to dementia conversion. Key Points An electronic health record-based model using demographic and co-morbidity data had good performance in identifying veterans who convert from mild cognitive impairment (MCI) to all-cause dementia (ACD) within 5 years.Increased age, stroke, cerebrovascular disease, myocardial infarction, hypertension, and diabetes were risk factors for 5-year conversion from MCI to ACD.High body mass index, alcohol abuse, and sleep apnea were protective factors for 5-year conversion from MCI to ACD.Models using synthetic data, analogs of real patient data that retain the distribution, density, and covariance between variables of real patient data but are not attributable to any specific patient, performed just as well as models using real patient data. This could have significant implications in facilitating widely distributed computing of health-care data with minimized patient privacy concern that could accelerate scientific discoveries.
Collapse
Affiliation(s)
- Chase Irwin
- Phoenix Veterans Affairs Health Care SystemPhoenixArizonaUSA
- University of Arizona College of Medicine‐PhoenixPhoenixArizonaUSA
| | - Donna Tjandra
- Phoenix Veterans Affairs Health Care SystemPhoenixArizonaUSA
- University of MichiganAnn ArborMichiganUSA
| | - Chengcheng Hu
- University of Arizona College of Medicine‐PhoenixPhoenixArizonaUSA
- College of Public HealthUniversity of ArizonaTucsonArizonaUSA
| | - Vinod Aggarwal
- MDClone LimitedBe'er ShevaIsrael
- VHA Office of Healthcare Innovation and LearningVA Central OfficeWashington, District of ColumbiaUSA
| | - Amanda Lienau
- VHA Office of Healthcare Innovation and LearningVA Central OfficeWashington, District of ColumbiaUSA
| | | | | | - Raymond Q. Migrino
- Phoenix Veterans Affairs Health Care SystemPhoenixArizonaUSA
- University of Arizona College of Medicine‐PhoenixPhoenixArizonaUSA
| |
Collapse
|
31
|
Bergman H, Borson S, Jessen F, Krolak-Salmon P, Pirani A, Rasmussen J, Rodrigo J, Taddeo D. Dementia and comorbidities in primary care: a scoping review. BMC PRIMARY CARE 2023; 24:277. [PMID: 38097969 PMCID: PMC10720181 DOI: 10.1186/s12875-023-02229-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND People with dementia (PwD) are known to have more chronic conditions compared to those without dementia, which can impact the clinical presentation of dementia, complicate clinical management and reduce overall quality of life. While primary care providers (PCPs) are integral to dementia care, it is currently unclear how PCPs adapt dementia care practices to account for comorbidities. This scoping review maps recent literature that describes the role for PCPs in the prevention, detection/diagnosis and management of dementia in the context of comorbidities, identifies critical knowledge gaps and proposes potential avenues for future research. METHODS We searched for peer-reviewed literature published between 2017-2022 in MEDLINE, Cochrane Library, and Scopus using key terms related to dementia, primary care, and comorbidity. The literature was screened for relevance by title-abstract screening and subsequent full-text screening. The prioritized papers were categorized as either 'Risk Assessment and Prevention', 'Screening, Detection, and Diagnosis' or 'Management' and were further labelled as either 'Tools and Technologies', 'Recommendations for Clinical Practice' or 'Programs and Initiatives'. RESULTS We identified 1,058 unique records in our search and respectively excluded 800 and 230 publications during title-abstract and full-text screening. Twenty-eight articles were included in our review, where ~ 50% describe the development and testing of tools and technologies that use pre-existing conditions to assess dementia risk. Only one publication provides official dementia screening guidelines for PCPs in people with pre-existing conditions. About 30% of the articles discuss managing the care of PwD, where most were anchored around models of multidisciplinary care and mitigating potentially inappropriate prescribing. CONCLUSION To our knowledge, this is the first scoping review that examines the role for PCPs in the prevention, detection/diagnosis and management of dementia in the context of comorbidities. Given our findings, we recommend that future studies: 1) further validate tools for risk assessment, timely detection and diagnosis that incorporate other health conditions; 2) provide additional guidance into how comorbidities could impact dementia care (including prescribing medication) in primary care settings; 3) incorporate comorbidities into primary care quality indicators for dementia; and 4) explore how to best incorporate dementia and comorbidities into models/frameworks of holistic, person-centred care.
Collapse
Affiliation(s)
- Howard Bergman
- Department of Family Medicine, McGill University, 5858 Ch. de La Côte-Des-Neiges, Suite 300, Montreal, QC, H3S 1Z1, Canada.
| | - Soo Borson
- Department of Family Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Frank Jessen
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
| | | | | | | | - Jesus Rodrigo
- Spanish Alzheimer's Confederation (Confederación Española de Alzheimer), Pamplona, Spain
| | - Daiana Taddeo
- Italian College of General Practitioners and Primary Care (SIMG - Società Italiana Di Medicina Genrale E Cure Primarie), Florence, Italy
| |
Collapse
|
32
|
Khan Y, Khan S, Hussain R, Rehman W, Maalik A, Gulshan U, Attwa MW, Darwish HW, Ghabbour HA, Ali N. Identification of Indazole-Based Thiadiazole-Bearing Thiazolidinone Hybrid Derivatives: Theoretical and Computational Approaches to Develop Promising Anti-Alzheimer's Candidates. Pharmaceuticals (Basel) 2023; 16:1667. [PMID: 38139795 PMCID: PMC10747300 DOI: 10.3390/ph16121667] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/18/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
A hybrid library of compounds based on indazole-based thiadiazole containing thiazolidinone moieties (1-17) was synthesized. The synthesized compounds were screened in vitro for their inhibition profile against targetedacetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) activities. All the derivatives demonstrated a varied range of inhibitory activities having IC50 values ranging from 0.86 ± 0.33 μM to 26.73 ± 0.84 μM (AChE) and 0.89 ± 0.12 μM to 27.08 ± 0.19 μM (BuChE), respectively. The results obtained were compared with standard Donepezil drugs (IC50 = 1.26 ± 0.18 μM for AChE) and (1.35 ± 0.37 μM for BuChE), respectively. Specifically, the derivatives 1-17, 1, 9, and 14 were found to be significantly active, with IC50 values of 0.86 ± 0.30, 0.92 ± 0.10, and 1.10 ± 0.37 μM (against AChE) and 0.89 ± 0.12, 0.98 ± 0.48 and 1.19 ± 0.42 μM (against BuChE), respectively.The structure-activity relationship (SAR) studies revealed that derivatives bearing para-CF3, ortho-OH, and para-F substitutions on the phenyl ring attached to the thiadiazole skeleton, as well as meta-Cl, -NO2, and para-chloro substitutions on the phenyl ring, having a significant effect on inhibitory potential. The synthesized scaffolds have been further characterized by using 1H-NMR, 13C-NMR, and (HR-MS) to confirm the precise structures of the synthesized compounds. Additionally, the molecular docking approach was carried out for most active compounds to explore the binding interactions established by most active compounds, with the active sites of targeted enzymes and obtained results supporting the experimental data.
Collapse
Affiliation(s)
- Yousaf Khan
- Department of Chemistry, COMSATS University Islamabad, Islamabad 45550, Pakistan; (Y.K.); (U.G.)
| | - Shoaib Khan
- Department of Chemistry, Abbottabad University of Science and Technology (AUST), Abbottabad 22500, Pakistan;
| | - Rafaqat Hussain
- Department of Chemistry, Hazara University, Mansehra 21120, Pakistan;
| | - Wajid Rehman
- Department of Chemistry, Hazara University, Mansehra 21120, Pakistan;
| | - Aneela Maalik
- Department of Chemistry, COMSATS University Islamabad, Islamabad 45550, Pakistan; (Y.K.); (U.G.)
| | - Urooba Gulshan
- Department of Chemistry, COMSATS University Islamabad, Islamabad 45550, Pakistan; (Y.K.); (U.G.)
| | - Mohamed W. Attwa
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.W.A.); (H.W.D.)
| | - Hany W. Darwish
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.W.A.); (H.W.D.)
| | - Hazem A. Ghabbour
- School of Health and Biomedical Sciences, RMIT University, Melbourne 3083, Australia;
| | - Nawab Ali
- Shangai Key Laboratory of Functional Material Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Meilong Road 130, Shanghai 200237, China;
| |
Collapse
|
33
|
Devi G. A how-to guide for a precision medicine approach to the diagnosis and treatment of Alzheimer's disease. Front Aging Neurosci 2023; 15:1213968. [PMID: 37662550 PMCID: PMC10469885 DOI: 10.3389/fnagi.2023.1213968] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/24/2023] [Indexed: 09/05/2023] Open
Abstract
Article purpose The clinical approach to Alzheimer's disease (AD) is challenging, particularly in high-functioning individuals. Accurate diagnosis is crucial, especially given the significant side effects, including brain hemorrhage, of newer monoclonal antibodies approved for treating earlier stages of Alzheimer's. Although early treatment is more effective, early diagnosis is also more difficult. Several clinical mimickers of AD exist either separately, or in conjunction with AD pathology, adding to the diagnostic complexity. To illustrate the clinical decision-making process, this study includes de-identified cases and reviews of the underlying etiology and pathology of Alzheimer's and available therapies to exemplify diagnostic and treatment subtleties. Problem The clinical presentation of Alzheimer's is complex and varied. Multiple other primary brain pathologies present with clinical phenotypes that can be difficult to distinguish from AD. Furthermore, Alzheimer's rarely exists in isolation, as almost all patients also show evidence of other primary brain pathologies, including Lewy body disease and argyrophilic grain disease. The phenotype and progression of AD can vary based on the brain regions affected by pathology, the coexistence and severity of other brain pathologies, the presence and severity of systemic comorbidities such as cardiac disease, the common co-occurrence with psychiatric diagnoses, and genetic risk factors. Additionally, symptoms and progression are influenced by an individual's brain reserve and cognitive reserve, as well as the timing of the diagnosis, which depends on the demographics of both the patient and the diagnosing physician, as well as the availability of biomarkers. Methods The optimal clinical and biomarker strategy for accurately diagnosing AD, common neuropathologic co-morbidities and mimickers, and available medication and non-medication-based treatments are discussed. Real-life examples of cognitive loss illustrate the diagnostic and treatment decision-making process as well as illustrative treatment responses. Implications AD is best considered a syndromic disorder, influenced by a multitude of patient and environmental characteristics. Additionally, AD existing alone is a unicorn, as there are nearly always coexisting other brain pathologies. Accurate diagnosis with biomarkers is essential. Treatment response is affected by the variables involved, and the effective treatment of Alzheimer's disease, as well as its prevention, requires an individualized, precision medicine strategy.
Collapse
Affiliation(s)
- Gayatri Devi
- Neurology and Psychiatry, Zucker School of Medicine, Hempstead, NY, United States
- Neurology and Psychiatry, Lenox Hill Hospital, New York City, NY, United States
- Park Avenue Neurology, New York City, NY, United States
| |
Collapse
|
34
|
Tarawneh R. Microvascular Contributions to Alzheimer Disease Pathogenesis: Is Alzheimer Disease Primarily an Endotheliopathy? Biomolecules 2023; 13:830. [PMID: 37238700 PMCID: PMC10216678 DOI: 10.3390/biom13050830] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/07/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer disease (AD) models are based on the notion that abnormal protein aggregation is the primary event in AD, which begins a decade or longer prior to symptom onset, and culminates in neurodegeneration; however, emerging evidence from animal and clinical studies suggests that reduced blood flow due to capillary loss and endothelial dysfunction are early and primary events in AD pathogenesis, which may precede amyloid and tau aggregation, and contribute to neuronal and synaptic injury via direct and indirect mechanisms. Recent data from clinical studies suggests that endothelial dysfunction is closely associated with cognitive outcomes in AD and that therapeutic strategies which promote endothelial repair in early AD may offer a potential opportunity to prevent or slow disease progression. This review examines evidence from clinical, imaging, neuropathological, and animal studies supporting vascular contributions to the onset and progression of AD pathology. Together, these observations support the notion that the onset of AD may be primarily influenced by vascular, rather than neurodegenerative, mechanisms and emphasize the importance of further investigations into the vascular hypothesis of AD.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Department of Neurology, Center for Memory and Aging, University of New Mexico, Albuquerque, NM 87106, USA
| |
Collapse
|
35
|
Wu CY, Ho CY, Yang YH. Developing Biomarkers for the Skin: Biomarkers for the Diagnosis and Prediction of Treatment Outcomes of Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24108478. [PMID: 37239825 DOI: 10.3390/ijms24108478] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/02/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by memory decline and cognitive impairment. Research on biomarkers can aid in early diagnosis, monitoring disease progression, evaluating treatment efficacy, and advancing fundamental research. We conducted a cross-sectional longitudinal study to see if there is an association between AD patients and age-matched healthy controls for their physiologic skin characteristics, such as pH, hydration, transepidermal water loss (TEWL), elasticity, microcirculation, and ApoE genotyping. The study used the Mini-Mental State Examination (MMSE) and Clinical Dementia Rating-Sum of the Boxes (CDR-SB) scales as references to quantify the presence of disease, if any. Our findings demonstrate that AD patients have a dominantly neutral pH, greater skin hydration, and less elasticity compared to the control subjects. At baseline, the tortuous capillary percentage negatively correlated with MMSE scores in AD patients. However, AD patients who carry the ApoE E4 allele and exhibit a high percentage of tortuous capillaries and capillary tortuous numbers have shown better treatment outcomes at six months. Therefore, we believe that physiologic skin testing is a rapid and effective way to screen, monitor progression, and ultimately guide the most appropriate treatment for AD patients.
Collapse
Affiliation(s)
- Ching-Ying Wu
- Department of Dermatology, College of Medicine and Post Baccalaureat Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Cosmetic Science, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
| | - Chih-Yi Ho
- Department of Dermatology, College of Medicine and Post Baccalaureat Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yuan-Han Yang
- Department of Neurology, College of Post Baccalaureat Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
36
|
Jakubowski H. Proteomic Exploration of Paraoxonase 1 Function in Health and Disease. Int J Mol Sci 2023; 24:7764. [PMID: 37175471 PMCID: PMC10178420 DOI: 10.3390/ijms24097764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/08/2023] [Accepted: 04/14/2023] [Indexed: 05/15/2023] Open
Abstract
High-density lipoprotein (HDL) exhibits cardio- and neuro-protective properties, which are thought to be promoted by paraoxonase 1 (PON1), a hydrolytic enzyme associated with an HDL subfraction also enriched with an anticoagulant protein (PROS1) and amyloid beta-transport protein clusterin (CLU, APOJ). Reduced levels of PON1 activity, characterized biochemically by elevated levels of homocysteine (Hcy)-thiolactone, oxidized lipids, and proteins modified by these metabolites in humans and mice, are associated with pathological abnormalities affecting the cardiovascular system (atherothrombosis) and the central nervous system (cognitive impairment, Alzheimer's disease). The molecular bases of these abnormalities have been largely unknown. Proteomic and metabolic studies over the past decade have significantly contributed to our understanding of PON1 function and the mechanisms by which PON1 deficiency can lead to disease. Recent studies discussed in this review highlight the involvement of dysregulated proteostasis in the pro-oxidative, pro-atherothrombotic, and pro-amyloidogenic phenotypes associated with low PON1 activity.
Collapse
Affiliation(s)
- Hieronim Jakubowski
- Department of Biochemistry and Biotechnology, University of Life Sciences, 60-637 Poznań, Poland; ; Tel.: +48-973-972-8733; Fax: +48-973-972-8981
- Department of Microbiology, Biochemistry and Molecular Genetics, International Center for Public Health, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| |
Collapse
|
37
|
Cho MH, Han K, Lee S, Jeong SM, Yoo JE, Kim S, Lee J, Chun S, Shin DW. Blood pressure and dementia risk by physical frailty in the elderly: a nationwide cohort study. Alzheimers Res Ther 2023; 15:56. [PMID: 36941727 PMCID: PMC10026431 DOI: 10.1186/s13195-023-01211-y] [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: 02/11/2022] [Accepted: 03/13/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND Midlife hypertension has been recognized as a modifiable risk factor for dementia, but association between blood pressure (BP) in late life and dementia has been inconclusive. In addition, few studies have investigated effects of BP control on dementia incidence in the frail elderly. Thus, this study aimed to investigate the association of BP and dementia incidence with concomitant consideration of physical frailty in the young elderly population. METHODS Using the Korean National Health Information Database, we identified 804,024 subjects without history of dementia at age 66. Dementia diagnosis was defined with prescription records of anti-dementia drugs and dementia-related diagnostic codes. Physical frailty was measured using the Timed Up and Go test. Association of BP and dementia incidence with concomitant consideration of physical frailty was investigated using Cox hazards analyses. RESULTS The risks of Alzheimer's and vascular dementia increased from systolic BP ≥ 160 and 130-139 mmHg, respectively; a significant association of dementia incidence with low BP was not observed. In the analyses stratified by the physical frailty status, low BP was not associated with increased risks of dementia within the groups both with and without physical frailty. CONCLUSIONS High BP was associated with increased risks of dementia, especially for vascular dementia, while low BP was not associated with increased risks of any type of dementia in young elderly people, even in those with physical frailty. This study suggests the need for tight BP control in young elderly people, irrespective of frailty status, to prevent dementia and supports the current clinical guidelines of hypertension treatment.
Collapse
Affiliation(s)
- Mi Hee Cho
- Samsung C&T Medical Clinic, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Seungwoo Lee
- Department of Medical Statistics, College of Medicine, Catholic University of Korea, Seoul, Republic of Korea
| | - Su-Min Jeong
- Department of Medicine, Seoul National University College of Medicine, Seoul , Republic of Korea
| | - Jung Eun Yoo
- Department of Family Medicine, Health System Gangnam Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University Bundang Hospital & Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Jinkook Lee
- Department of Economics & Center for Economic & Social Research, University of Southern California, Los Angeles, CA, USA
| | - Sohyun Chun
- International Healthcare Center, Samsung Medical Center, 81 Irwon-Ro, Gangnam-Gu, Seoul, Republic of Korea.
| | - Dong Wook Shin
- Department of Family Medicine/Supportive Care Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
- Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea.
| |
Collapse
|
38
|
Kosenko E, Tikhonova L, Alilova G, Montoliu C. Erythrocytes Functionality in SARS-CoV-2 Infection: Potential Link with Alzheimer's Disease. Int J Mol Sci 2023; 24:5739. [PMID: 36982809 PMCID: PMC10051442 DOI: 10.3390/ijms24065739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a rapidly spreading acute respiratory infection caused by SARS-CoV-2. The pathogenesis of the disease remains unclear. Recently, several hypotheses have emerged to explain the mechanism of interaction between SARS-CoV-2 and erythrocytes, and its negative effect on the oxygen-transport function that depends on erythrocyte metabolism, which is responsible for hemoglobin-oxygen affinity (Hb-O2 affinity). In clinical settings, the modulators of the Hb-O2 affinity are not currently measured to assess tissue oxygenation, thereby providing inadequate evaluation of erythrocyte dysfunction in the integrated oxygen-transport system. To discover more about hypoxemia/hypoxia in COVID-19 patients, this review highlights the need for further investigation of the relationship between biochemical aberrations in erythrocytes and oxygen-transport efficiency. Furthermore, patients with severe COVID-19 experience symptoms similar to Alzheimer's, suggesting that their brains have been altered in ways that increase the likelihood of Alzheimer's. Mindful of the partly assessed role of structural, metabolic abnormalities that underlie erythrocyte dysfunction in the pathophysiology of Alzheimer's disease (AD), we further summarize the available data showing that COVID-19 neurocognitive impairments most probably share similar patterns with known mechanisms of brain dysfunctions in AD. Identification of parameters responsible for erythrocyte function that vary under SARS-CoV-2 may contribute to the search for additional components of progressive and irreversible failure in the integrated oxygen-transport system leading to tissue hypoperfusion. This is particularly relevant for the older generation who experience age-related disorders of erythrocyte metabolism and are prone to AD, and provide an opportunity for new personalized therapies to control this deadly infection.
Collapse
Affiliation(s)
- Elena Kosenko
- Institute of Theoretical and Experimental Biophysics of Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Lyudmila Tikhonova
- Institute of Theoretical and Experimental Biophysics of Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Gubidat Alilova
- Institute of Theoretical and Experimental Biophysics of Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Carmina Montoliu
- Hospital Clinico Research Foundation, INCLIVA Health Research Institute, 46010 Valencia, Spain
- Pathology Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
39
|
Berk-Rauch HE, Choudhury A, Richards AT, Singh PK, Chen ZL, Norris EH, Strickland S, Ahn HJ. Striatal fibrinogen extravasation and vascular degeneration correlate with motor dysfunction in an aging mouse model of Alzheimer’s disease. Front Aging Neurosci 2023; 15:1064178. [PMID: 36967821 PMCID: PMC10034037 DOI: 10.3389/fnagi.2023.1064178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/13/2023] [Indexed: 03/11/2023] Open
Abstract
Introduction: Alzheimer’s Disease (AD) patients exhibit signs of motor dysfunction, including gait, locomotion, and balance deficits. Changes in motor function often precede other symptoms of AD as well as correlate with increased severity and mortality. Despite the frequent occurrence of motor dysfunction in AD patients, little is known about the mechanisms by which this behavior is altered.Methods and Results: In the present study, we investigated the relationship between cerebrovascular impairment and motor dysfunction in a mouse model of AD (Tg6799). We found an age-dependent increase of extravasated fibrinogen deposits in the cortex and striatum of AD mice. Interestingly, there was significantly decreased cerebrovascular density in the striatum of the 15-month-old as compared to 7-month-old AD mice. We also found significant demyelination and axonal damage in the striatum of aged AD mice. We analyzed striatum-related motor function and anxiety levels of AD mice at both ages and found that aged AD mice exhibited significant impairment of motor function but not in the younger AD mice.Discussion: Our finding suggests an enticing correlation between extravasated fibrinogen, cerebrovascular damage of the striatum, and motor dysfunction in an AD mouse model, suggesting a possible mechanism underlying motor dysfunction in AD.
Collapse
Affiliation(s)
- Hanna E. Berk-Rauch
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Arnab Choudhury
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Allison T. Richards
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Pradeep K. Singh
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Zu-Lin Chen
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Hyung Jin Ahn
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, United States
- Brain Health Institute, Rutgers University, Piscataway, NJ, United States
- *Correspondence: Hyung Jin Ahn,
| |
Collapse
|
40
|
Madsen LS, Parbo P, Ismail R, Gottrup H, Østergaard L, Brooks DJ, Eskildsen SF. Capillary dysfunction correlates with cortical amyloid load in early Alzheimer's disease. Neurobiol Aging 2023; 123:1-9. [PMID: 36610198 DOI: 10.1016/j.neurobiolaging.2022.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Alterations in cerebral perfusion is increasingly considered to play a crucial role in Alzheimer's disease (AD) and together with accumulated amyloid-β, deficiencies in the brain microvascular circulation may result in local hypoxia. Here, we studied alterations in cerebral circulation and the correlation between amyloid-β load and cerebral perfusion in prodromal AD (pAD). Using dynamic susceptibility contrast MRI and PET, we evaluated cerebral perfusion and amyloid-β levels in 19 individuals with mild cognitive impairment (MCI) and high amyloid-β load (pAD-MCI), 13 MCI individuals without AD pathology and 21 healthy controls. The pAD-MCI group showed significantly lower microvascular blood flow and significantly higher heterogeneity of microvascular blood transit times (p < 0.01) compared with the other 2 groups. Additionally, in the pAD-MCI group raised amyloid-β levels correlated with decreased microvascular blood flow and increased heterogeneity of microvascular blood flow in frontal and temporal areas (p < 0.01). These results indicate a close connection between levels of amyloid-β deposition and brain microvascular perfusion in pAD. A vicious cycle may be established where amyloid-β load and deficiencies in brain perfusion may reinforce each other.
Collapse
Affiliation(s)
- Lasse S Madsen
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark; Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Peter Parbo
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Rola Ismail
- Department of Nuclear Medicine, Sygehus Lillebaelt, Kolding, Denmark
| | - Hanne Gottrup
- Dementia Clinic, Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark
| | - David J Brooks
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark; Institute of Translational and Clinical Research, University of Newcastle upon Tyne, Newcastle, UK
| | - Simon F Eskildsen
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
41
|
Zhang Y, Liu W, Wang X, Liu Y, Wei H. Nanozyme-Enabled Treatment of Cardio- and Cerebrovascular Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204809. [PMID: 36192166 DOI: 10.1002/smll.202204809] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/18/2022] [Indexed: 06/16/2023]
Abstract
Cardio- and cerebrovascular diseases are two major vascular-related diseases that lead to death worldwide. Reactive oxygen species (ROS) play a vital role in the occurrence and exacerbation of diseases. Excessive ROS induce cellular context damage and lead to tissue dysfunction. Nanozymes, as emerging enzyme mimics, offer a unique perspective for therapy through multifunctional activities, achieving essential results in the treatment of ROS-related cardio- and cerebrovascular diseases by directly scavenging excess ROS or regulating pathologically related molecules. This review first introduces nanozyme-enabled therapeutic mechanisms at the cellular level. Then, the therapies for several typical cardio- and cerebrovascular diseases with nanozymes are discussed, mainly including cardiovascular diseases, ischemia reperfusion injury, and neurological disorders. Finally, the challenges and outlooks for the application of nanozymes are also presented. This review will provide some instructive perspectives on nanozymes and promote the development of enzyme-mimicking strategies in cardio- and cerebrovascular disease therapy.
Collapse
Affiliation(s)
- Yihong Zhang
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Wanling Liu
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Xiaoyu Wang
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
- Department of Chemistry and Material Science, College of Science, Nanjing Forestry University, Nanjing, Jiangsu, 210037, China
| | - Yufeng Liu
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Hui Wei
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, Jiangsu, 210023, China
| |
Collapse
|
42
|
Zuliani G, Marsillach J, Trentini A, Rosta V, Cervellati C. Lipoprotein-Associated Phospholipase A2 Activity as Potential Biomarker of Vascular Dementia. Antioxidants (Basel) 2023; 12:597. [PMID: 36978845 PMCID: PMC10045550 DOI: 10.3390/antiox12030597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023] Open
Abstract
A wealth of evidence suggests that Lipoprotein-associated phospholipase A2 (Lp-PLA2) plays a relevant role in atherogenesis and inflammation, which in turn are associated with the risk of developing dementia. The aim of this study was to evaluate whether serum Lp-PLA2 activity might be an early and/or late biomarker for different forms of dementia. Serum Lp-PLA2 activity was assessed in older patients with mild cognitive impairment (MCI, n = 166; median clinical follow-up = 29 months), Late-Onset Alzheimer's disease (LOAD, n = 176), vascular dementia (VAD, n = 43), dementia characterized by an overlap between LOAD and VAD (AD-VAD MIXED dementia) (n = 136), other dementia subtypes (n = 45), and cognitively normal controls (n = 151). We found a significant trend towards higher levels of Lp-PLA2 activity in VAD compared with the other groups (ANOVA, p = 0.028). Similarly, Lp-PLA2 activity was greater in MCI converting to VAD compared with those that did not or did convert to the other types of dementia (ANOVA, p = 0.011). After adjusting for potential confounders, high levels of Lp-PLA2 activity were associated with the diagnosis of VAD (O.R. = 2.38, 95% C.I. = 1.06-5.10), but not with other types of dementia. Our data suggest that increased serum Lp-PLA2 activity may represent a potential biomarker for the diagnosis of VAD.
Collapse
Affiliation(s)
- Giovanni Zuliani
- Department of Translational Medicine and for Romagna, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Judit Marsillach
- Department of Environmental & Occupational Health Sciences, University of Washington, 4225 NE Roosevelt Way, Seattle, WA 98105, USA
| | - Alessandro Trentini
- Department of Environmental and Prevention Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Valentina Rosta
- Department of Translational Medicine and for Romagna, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Carlo Cervellati
- Department of Translational Medicine and for Romagna, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| |
Collapse
|
43
|
Witucki Ł, Jakubowski H. Depletion of Paraoxonase 1 (Pon1) Dysregulates mTOR, Autophagy, and Accelerates Amyloid Beta Accumulation in Mice. Cells 2023; 12:746. [PMID: 36899882 PMCID: PMC10001133 DOI: 10.3390/cells12050746] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/16/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Paraoxonase 1 (PON1), a homocysteine (Hcy)-thiolactone detoxifying enzyme, has been associated with Alzheimer's disease (AD), suggesting that PON1 plays an important protective role in the brain. To study the involvement of PON1 in the development of AD and to elucidate the mechanism involved, we generated a new mouse model of AD, the Pon1-/-xFAD mouse, and examined how Pon1 depletion affects mTOR signaling, autophagy, and amyloid beta (Aβ) accumulation. To elucidate the mechanism involved, we examined these processes in N2a-APPswe cells. We found that Pon1 depletion significantly downregulated Phf8 and upregulated H4K20me1; mTOR, phospho-mTOR, and App were upregulated while autophagy markers Bcln1, Atg5, and Atg7 were downregulated at the protein and mRNA levels in the brains of Pon1─/─5xFAD vs. Pon1+/+5xFAD mice. Pon1 depletion in N2a-APPswe cells by RNA interference led to downregulation of Phf8 and upregulation of mTOR due to increased H4K20me1-mTOR promoter binding. This led to autophagy downregulation and significantly increased APP and Aβ levels. Phf8 depletion by RNA interference or treatments with Hcy-thiolactone or N-Hcy-protein metabolites similarly increased Aβ levels in N2a-APPswe cells. Taken together, our findings define a neuroprotective mechanism by which Pon1 prevents Aβ generation.
Collapse
Affiliation(s)
- Łukasz Witucki
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, 60-637 Poznań, Poland
- Department of Microbiology, Biochemistry and Molecular Genetics, International Center for Public Health, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Hieronim Jakubowski
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, 60-637 Poznań, Poland
- Department of Microbiology, Biochemistry and Molecular Genetics, International Center for Public Health, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| |
Collapse
|
44
|
Sergi D, Zauli E, Tisato V, Secchiero P, Zauli G, Cervellati C. Lipids at the Nexus between Cerebrovascular Disease and Vascular Dementia: The Impact of HDL-Cholesterol and Ceramides. Int J Mol Sci 2023; 24:ijms24054403. [PMID: 36901834 PMCID: PMC10002119 DOI: 10.3390/ijms24054403] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Cerebrovascular diseases and the subsequent brain hypoperfusion are at the basis of vascular dementia. Dyslipidemia, marked by an increase in circulating levels of triglycerides and LDL-cholesterol and a parallel decrease in HDL-cholesterol, in turn, is pivotal in promoting atherosclerosis which represents a common feature of cardiovascular and cerebrovascular diseases. In this regard, HDL-cholesterol has traditionally been considered as being protective from a cardiovascular and a cerebrovascular prospective. However, emerging evidence suggests that their quality and functionality play a more prominent role than their circulating levels in shaping cardiovascular health and possibly cognitive function. Furthermore, the quality of lipids embedded in circulating lipoproteins represents another key discriminant in modulating cardiovascular disease, with ceramides being proposed as a novel risk factor for atherosclerosis. This review highlights the role of HDL lipoprotein and ceramides in cerebrovascular diseases and the repercussion on vascular dementia. Additionally, the manuscript provides an up-to-date picture of the impact of saturated and omega-3 fatty acids on HDL circulating levels, functionality and ceramide metabolism.
Collapse
Affiliation(s)
- Domenico Sergi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Correspondence:
| | - Enrico Zauli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Veronica Tisato
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Paola Secchiero
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Giorgio Zauli
- King Khaled Eye Specialistic Hospital, Riyadh 11462, Saudi Arabia
| | - Carlo Cervellati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
45
|
Bazala R, Zoppellaro G, Kletetschka G. Iron Level Changes in the Brain with Neurodegenerative Disease. BRAIN MULTIPHYSICS 2023. [DOI: 10.1016/j.brain.2023.100063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
46
|
Obrenovich M, Singh SK, Li Y, Perry G, Siddiqui B, Haq W, Reddy VP. Natural Product Co-Metabolism and the Microbiota-Gut-Brain Axis in Age-Related Diseases. Life (Basel) 2022; 13:41. [PMID: 36675988 PMCID: PMC9865576 DOI: 10.3390/life13010041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Complementary alternative medicine approaches are growing treatments of diseases to standard medicine practice. Many of these concepts are being adopted into standard practice and orthomolecular medicine. Age-related diseases, in particular neurodegenerative disorders, are particularly difficult to treat and a cure is likely a distant expectation for many of them. Shifting attention from pharmaceuticals to phytoceuticals and "bugs as drugs" represents a paradigm shift and novel approaches to intervention and management of age-related diseases and downstream effects of aging. Although they have their own unique pathologies, a growing body of evidence suggests Alzheimer's disease (AD) and vascular dementia (VaD) share common pathology and features. Moreover, normal metabolic processes contribute to detrimental aging and age-related diseases such as AD. Recognizing the role that the cerebral and cardiovascular pathways play in AD and age-related diseases represents a common denominator in their pathobiology. Understanding how prosaic foods and medications are co-metabolized with the gut microbiota (GMB) would advance personalized medicine and represents a paradigm shift in our view of human physiology and biochemistry. Extending that advance to include a new physiology for the advanced age-related diseases would provide new treatment targets for mild cognitive impairment, dementia, and neurodegeneration and may speed up medical advancements for these particularly devastating and debilitating diseases. Here, we explore selected foods and their derivatives and suggest new dementia treatment approaches for age-related diseases that focus on reexamining the role of the GMB.
Collapse
Affiliation(s)
- Mark Obrenovich
- Research Service, Department of Veteran's Affairs Medical Center, Cleveland, OH 44106, USA
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
- The Gilgamesh Foundation for Medical Science and Research, Cleveland, OH 44116, USA
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
- Departments of Chemistry and Biological and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Sandeep Kumar Singh
- Indian Scientific Education and Technology (ISET) Foundation, Lucknow 226002, India
| | - Yi Li
- Department of Nutrition and Dietetics, Saint Louis University, Saint Louis, MO 63103, USA
| | - George Perry
- Department of Neuroscience Developmental and Regenerative Biology, University of Texas, San Antonio, TX 78249, USA
| | - Bushra Siddiqui
- School of Medicine, Northeast Ohio College of Medicine, Rootstown, OH 44272, USA
| | - Waqas Haq
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - V Prakash Reddy
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO 65409, USA
| |
Collapse
|
47
|
Cheon SY, Song J. Novel insights into non-alcoholic fatty liver disease and dementia: insulin resistance, hyperammonemia, gut dysbiosis, vascular impairment, and inflammation. Cell Biosci 2022; 12:99. [PMID: 35765060 PMCID: PMC9237975 DOI: 10.1186/s13578-022-00836-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/20/2022] [Indexed: 02/08/2023] Open
Abstract
AbstractNon-alcoholic fatty liver disease (NAFLD) is a metabolic disease characterized by multiple pathologies. The progression of dementia with NAFLD may be affected by various risk factors, including brain insulin resistance, cerebrovascular dysfunction, gut dysbiosis, and neuroinflammation. Many recent studies have focused on the increasing prevalence of dementia in patients with NAFLD. Dementia is characterized by cognitive and memory deficits and has diverse subtypes, including vascular dementia, Alzheimer’s dementia, and diabetes mellitus-induced dementia. Considering the common pathological features of NAFLD and dementia, further studies on the association between them are needed to find appropriate therapeutic solutions for diseases. This review summarizes the common pathological characteristics and mechanisms of NAFLD and dementia. Additionally, it describes recent evidence on association between NAFLD and dementia progression and provides novel perspectives with regard to the treatment of patients with dementia secondary to NAFLD.
Collapse
|
48
|
Greene AN, Solomon MB, Privette Vinnedge LM. Novel molecular mechanisms in Alzheimer's disease: The potential role of DEK in disease pathogenesis. Front Aging Neurosci 2022; 14:1018180. [PMID: 36275000 PMCID: PMC9582447 DOI: 10.3389/fnagi.2022.1018180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease and age-related dementias (AD/ADRD) are debilitating diseases that exact a significant physical, emotional, cognitive, and financial toll on the individual and their social network. While genetic risk factors for early-onset AD have been identified, the molecular and genetic drivers of late-onset AD, the most common subtype, remain a mystery. Current treatment options are limited for the 35 million people in the United States with AD/ADRD. Thus, it is critically important to identify novel molecular mechanisms of dementia-related pathology that may be targets for the development of new interventions. Here, we summarize the overarching concepts regarding AD/ADRD pathogenesis. Then, we highlight one potential molecular driver of AD/ADRD, the chromatin remodeling protein DEK. We discuss in vitro, in vivo, and ex vivo findings, from our group and others, that link DEK loss with the cellular, molecular, and behavioral signatures of AD/ADRD. These include associations between DEK loss and cellular and molecular hallmarks of AD/ADRD, including apoptosis, Tau expression, and Tau hyperphosphorylation. We also briefly discuss work that suggests sex-specific differences in the role of DEK in AD/ADRD pathogenesis. Finally, we discuss future directions for exploiting the DEK protein as a novel player and potential therapeutic target for the treatment of AD/ADRD.
Collapse
Affiliation(s)
- Allie N. Greene
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Matia B. Solomon
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Department of Psychology, University of Cincinnati, Cincinnati, OH, United States
| | - Lisa M. Privette Vinnedge
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
49
|
Canna A, Esposito F, Tedeschi G, Trojsi F, Passaniti C, di Meo I, Polito R, Maiorino MI, Paolisso G, Cirillo M, Rizzo MR. Neurovascular coupling in patients with type 2 diabetes mellitus. Front Aging Neurosci 2022; 14:976340. [PMID: 36118711 PMCID: PMC9476313 DOI: 10.3389/fnagi.2022.976340] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022] Open
Abstract
Functional and metabolic neural changes in Type 2 diabetes mellitus (T2DM) can be associated with poor cognitive performances. Here we analyzed the functional-metabolic neurovascular coupling (NVC) in the brain of T2DM patients. Thirty-three patients (70 ± 6 years, 15 males) with recent T2DM diagnosis and 18 healthy control (HC) subjects (65 ± 9 years, 9 males) were enrolled in a brain MRI study to identify the potential effects of T2DM on NVC. T2DM patients were either drug-naive (n = 19) or under treatment with metformin (n = 14) since less than 6 months. Arterial spin labeling and blood oxygen level dependent resting-state functional MRI (RS-fMRI) images were combined to derive NVC measures in brain regions and large-scale networks in a standard brain parcelation. Altered NVC values in T2DM patients were correlated with cognitive performances spanning several neurological domains using Spearman correlation coefficients. Compared to HC, T2DM patients had reduced NVC in the default mode network (DMN) and increased NVC in three regions of the dorsal (DAN) and salience-ventral (SVAN) attention networks. NVC abnormalities in DAN and SVAN were associated with reduced visuo-spatial cognitive performances. A spatial pattern of NVC reduction in the DMN, accompanied by isolated regional NVC increases in DAN and SVAN, could reflect the emergence of (defective) compensatory processes in T2DM patients in response to altered neurovascular conditions. Overall, this pattern is reminiscent of neural abnormalities previously observed in Alzheimer’s disease, suggesting that similar neurobiological mechanisms, secondary to insulin resistance and manifesting as NVC alterations, might be developing in T2DM pathology.
Collapse
|
50
|
Liu CC, Zhao J, Fu Y, Inoue Y, Ren Y, Chen Y, Doss SV, Shue F, Jeevaratnam S, Bastea L, Wang N, Martens YA, Qiao W, Wang M, Zhao N, Jia L, Yamazaki Y, Yamazaki A, Rosenberg CL, Wang Z, Kong D, Li Z, Kuchenbecker LA, Trottier ZA, Felton L, Rogers J, Quicksall ZS, Linares C, Knight J, Chen Y, Kurti A, Kanekiyo T, Fryer JD, Asmann YW, Storz P, Wang X, Peng J, Zhang B, Kim BYS, Bu G. Peripheral apoE4 enhances Alzheimer's pathology and impairs cognition by compromising cerebrovascular function. Nat Neurosci 2022; 25:1020-1033. [PMID: 35915180 PMCID: PMC10009873 DOI: 10.1038/s41593-022-01127-0] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 06/22/2022] [Indexed: 12/21/2022]
Abstract
The ε4 allele of the apolipoprotein E (APOE) gene, a genetic risk factor for Alzheimer's disease, is abundantly expressed in both the brain and periphery. Here, we present evidence that peripheral apoE isoforms, separated from those in the brain by the blood-brain barrier, differentially impact Alzheimer's disease pathogenesis and cognition. To evaluate the function of peripheral apoE, we developed conditional mouse models expressing human APOE3 or APOE4 in the liver with no detectable apoE in the brain. Liver-expressed apoE4 compromised synaptic plasticity and cognition by impairing cerebrovascular functions. Plasma proteome profiling revealed apoE isoform-dependent functional pathways highlighting cell adhesion, lipoprotein metabolism and complement activation. ApoE3 plasma from young mice improved cognition and reduced vessel-associated gliosis when transfused into aged mice, whereas apoE4 compromised the beneficial effects of young plasma. A human induced pluripotent stem cell-derived endothelial cell model recapitulated the plasma apoE isoform-specific effect on endothelial integrity, further supporting a vascular-related mechanism. Upon breeding with amyloid model mice, liver-expressed apoE4 exacerbated brain amyloid pathology, whereas apoE3 reduced it. Our findings demonstrate pathogenic effects of peripheral apoE4, providing a strong rationale for targeting peripheral apoE to treat Alzheimer's disease.
Collapse
Affiliation(s)
- Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
| | - Jing Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yuan Fu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yasuteru Inoue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yingxue Ren
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Yuanxin Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Sydney V Doss
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Ligia Bastea
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Na Wang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Wenhui Qiao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Minghui Wang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Lin Jia
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yu Yamazaki
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Akari Yamazaki
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Zhen Wang
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Dehui Kong
- Department of Biology, University of North Dakota, Grand Forks, ND, USA
| | - Zonghua Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Lindsey Felton
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Justin Rogers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Cynthia Linares
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Joshua Knight
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yixing Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Aishe Kurti
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - John D Fryer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yan W Asmann
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Xusheng Wang
- Department of Biology, University of North Dakota, Grand Forks, ND, USA
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|