1
|
Choudhary G, Prajapat M, Kaur G, Singh H, Mahendiratta S, Prakash A, Medhi B. Integrated in-silico and in-vitro assessments of HDAC6 inhibitor efficacy in mitigating amyloid beta pathology in Alzheimer's disease. J Biomol Struct Dyn 2024; 42:9720-9730. [PMID: 37878051 DOI: 10.1080/07391102.2023.2274518] [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: 02/22/2023] [Accepted: 08/21/2023] [Indexed: 10/26/2023]
Abstract
Alzheimer's disease, marked by memory loss and cognitive decline, is associated with amyloid-beta (Aβ) peptide accumulation in the brain. The enzyme neprilysin (NEP), crucial for Aβ degradation, decreases with age and in sporadic Alzheimer's disease, leading to increased Aβ build-up. This study hypothesized the targeting of enzyme HDAC6, believed to influence NEP activity. An in-silico study was conducted using an FDA-approved drug database, with the focus on their interaction with the HDAC6 structure. Among tested ligands, Panobinostat showed the most favourable interaction with HDAC6. In-vitro experiments on the SH-SY5Y neuronal cell line confirmed these findings, with Panobinostat inhibiting HDAC6, enhancing NEP levels, and reducing Aβ load. The study suggests Panobinostat as a potential Alzheimer's therapeutic agent, mitigating Aβ accumulation via NEP upregulation. Further research is required for comprehensive understanding and validation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | | | - Gurjeet Kaur
- Department of Pharmacology, PGIMER, Chandigarh, India
| | | | | | - Ajay Prakash
- Department of Pharmacology, PGIMER, Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, PGIMER, Chandigarh, India
| |
Collapse
|
2
|
Chen F, Zhao J, Meng F, He F, Ni J, Fu Y. The vascular contribution of apolipoprotein E to Alzheimer's disease. Brain 2024; 147:2946-2965. [PMID: 38748848 DOI: 10.1093/brain/awae156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/23/2024] [Accepted: 04/21/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease, the most prevalent form of dementia, imposes a substantial societal burden. The persistent inadequacy of disease-modifying drugs targeting amyloid plaques and neurofibrillary tangles suggests the contribution of alternative pathogenic mechanisms. A frequently overlooked aspect is cerebrovascular dysfunction, which may manifest early in the progression of Alzheimer's disease pathology. Mounting evidence underscores the pivotal role of the apolipoprotein E gene, particularly the apolipoprotein ε4 allele as the strongest genetic risk factor for late-onset Alzheimer's disease, in the cerebrovascular pathology associated with Alzheimer's disease. In this review, we examine the evidence elucidating the cerebrovascular impact of both central and peripheral apolipoprotein E on the pathogenesis of Alzheimer's disease. We present a novel three-hit hypothesis, outlining potential mechanisms that shed light on the intricate relationship among different pathogenic events. Finally, we discuss prospective therapeutics targeting the cerebrovascular pathology associated with apolipoprotein E and explore their implications for future research endeavours.
Collapse
Affiliation(s)
- Feng Chen
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jing Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Fanxia Meng
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Fangping He
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jie Ni
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuan Fu
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
3
|
Zhao M, Wang Y, Shen Y, Wei C, Zhang G, Sun L. A review of the roles of pathogens in Alzheimer's disease. Front Neurosci 2024; 18:1439055. [PMID: 39224577 PMCID: PMC11366636 DOI: 10.3389/fnins.2024.1439055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease (AD) is one of the leading causes of dementia and is characterized by memory loss, mental and behavioral abnormalities, and impaired ability to perform daily activities. Even as a global disease that threatens human health, effective treatments to slow the progression of AD have not been found, despite intensive research and significant investment. In recent years, the role of infections in the etiology of AD has sparked intense debate. Pathogens invade the central nervous system through a damaged blood-brain barrier or nerve trunk and disrupt the neuronal structure and function as well as homeostasis of the brain microenvironment through a series of molecular biological events. In this review, we summarize the various pathogens involved in AD pathology, discuss potential interactions between pathogens and AD, and provide an overview of the promising future of anti-pathogenic therapies for AD.
Collapse
Affiliation(s)
| | | | | | | | | | - Li Sun
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
4
|
Ozdilek B, Agirbasli M. Soluble LRP-1 in Parkinson's disease: clues for paradoxical effects. Int J Neurosci 2024; 134:620-627. [PMID: 36184975 DOI: 10.1080/00207454.2022.2131552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 12/29/2021] [Accepted: 09/21/2022] [Indexed: 10/17/2022]
Abstract
Background: Low density lipoprotein receptor-related protein-1 (LRP-1) is highly expressed in the central nervous system and plays a role in neurodegenerative disorders. The available data on this subject-matter seem to support the presence of a correlation between LRP-1 levels and abnormal aggregation of a plurality of proteins, including tau, amyloid, and α‑synuclein. Understanding the molecular mechanisms underlying Parkinson's disease (PD) is critical for development of new therapies. Aim: To investigate serum soluble LRP-1 (sLRP-1) concentrations in patients with PD and explored their potential role as a biomarker in diagnosis and prognosis of disease. Methods: Based on well-defined inclusion and exclusion criteria, we have included 133 PD patients and 45 healthy controls. The clinical severity was assessed using Hoehn Yahr and Unified PD Rating Scale (UPDRS). Following a fasting period, venous blood samples were taken, and centrifuged. Serum samples were stored until analysis. sLRP-1 was measured by ELISA assay. Results: The median of serum sLRP-1 levels was higher in PD patients compared to that in healthy controls, but without reaching a statistical significance. There was a positive, but statistically insignificant, correlation between sLRP-1 levels and duration of disease. sLRP-1 levels had a significant correlation with UPDRS Parts I and IV. Patients with hypertension showed lower levels of sLRP-1. Conclusion: The present study suggests that serum sLRP-1 concentrations are associated with the factors influencing prognosis of PD and disease severity. Further studies are needed to definitively determine whether or not sLRP-1 can be utilized as a diagnostic and prognostic biomarker for PD.
Collapse
Affiliation(s)
- Betul Ozdilek
- Department of Neurology, Istanbul Medeniyet University Faculty of Medicine, Istanbul, Turkey
- Clinic of Neurology, Ministry of Health Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Mehmet Agirbasli
- Department of Cardiology, Istanbul Medeniyet University Faculty of Medicine, Istanbul, Turkey
- Clinic of Cardiology, Ministry of Health Goztepe Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
5
|
Hickey JP, Collins AE, Nelson ML, Chen H, Kalisch BE. Modulation of Oxidative Stress and Neuroinflammation by Cannabidiol (CBD): Promising Targets for the Treatment of Alzheimer's Disease. Curr Issues Mol Biol 2024; 46:4379-4402. [PMID: 38785534 PMCID: PMC11120237 DOI: 10.3390/cimb46050266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and the most common form of dementia globally. Although the direct cause of AD remains under debate, neuroinflammation and oxidative stress are critical components in its pathogenesis and progression. As a result, compounds like cannabidiol (CBD) are being increasingly investigated for their ability to provide antioxidant and anti-inflammatory neuroprotection. CBD is the primary non-psychotropic phytocannabinoid derived from Cannabis sativa. It has been found to provide beneficial outcomes in a variety of medical conditions and is gaining increasing attention for its potential therapeutic application in AD. CBD is not psychoactive and its lipophilic nature allows its rapid distribution throughout the body, including across the blood-brain barrier (BBB). CBD also possesses anti-inflammatory, antioxidant, and neuroprotective properties, making it a viable candidate for AD treatment. This review outlines CBD's mechanism of action, the role of oxidative stress and neuroinflammation in AD, and the effectiveness and limitations of CBD in preclinical models of AD.
Collapse
Affiliation(s)
| | | | | | | | - Bettina E. Kalisch
- Department of Biomedical Sciences and Collaborative Specialization in Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada; (J.P.H.); (A.E.C.); (M.L.N.); (H.C.)
| |
Collapse
|
6
|
Lu Y, Pike JR, Hoogeveen R, Walker K, Raffield L, Selvin E, Avery C, Engel S, Mielke MM, Garcia T, Heiss G, Palta P. Nonalcoholic Fatty Liver Disease and Longitudinal Change in Imaging and Plasma Biomarkers of Alzheimer Disease and Vascular Pathology. Neurology 2024; 102:e209203. [PMID: 38471046 PMCID: PMC11033987 DOI: 10.1212/wnl.0000000000209203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/23/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Prospective measures of plasma and cerebral MRI biomarkers of Alzheimer disease (AD) and vascular neuropathology provide an opportunity to investigate possible mechanisms linking liver disease and dementia. We aimed to quantify the association of midlife nonalcoholic fatty liver disease (NAFLD) with change in plasma and brain MRI biomarkers of AD and vascular neuropathology. METHODS We included participants from the Atherosclerosis Risk in Communities Study with brain MRI measurements of white matter hyperintensity (WMH) volume and temporal-parietal lobe cortical thickness meta region of interest (ROI) at up to 2 different visits, in 2011-13 and 2016-19, and plasma biomarkers of β-amyloid (Aβ)42:40, phosphorylated tau at threonine 181, and neurofilament light (NfL) were measured up to 3 times in 1993-95, 2011-13, and 2016-19. NAFLD was categorized using the fatty liver index in 1990-92. Multivariate linear regression was performed for associations between midlife NAFLD and change in plasma and brain MRI biomarkers of AD and vascular neuropathology. The primary models adjusted for demographics, Apolipoprotein E, alcohol use, and kidney function. RESULTS Among 1,706 participants (mean age 56 years, 62% female, 28% Black), midlife NAFLD vs no NAFLD was associated with greater late-life WMH volume (difference per SD 0.19, 95% CI 0.06-0.31) and faster late-life WMH increase over 6 years (difference in annual change, SD 0.28, 95% CI 0.05-0.51), suggesting accumulating vascular pathology. Midlife NAFLD vs no NAFLD was also associated with AD biomarkers in midlife (lower Aβ42:40 [SD -0.21, 95% CI -0.39 to -0.04] measured in 1993-95) and late life (lower Aβ42:40 [SD -0.13, 95% CI -0.23 to -0.03] and lower temporal-parietal lobe cortical thickness meta ROI [SD -0.16, 95% CI -0.28 to -0.05] measured in 2011-13). Although midlife NfL was lower in individuals with vs without midlife NAFLD, those with NAFLD exhibited a faster rate of NfL increase that accelerated over time. DISCUSSION Midlife NAFLD shows associations with AD and accumulating vascular pathology, revealing potential pathways linking liver function to dementia. Plasma biomarkers of neuropathology and neuronal injury may serve as easily measurable and dynamic indicators for monitoring the impacts of impaired liver function on brain health.
Collapse
Affiliation(s)
- Yifei Lu
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - James R Pike
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Ron Hoogeveen
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Keenan Walker
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Laura Raffield
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Elizabeth Selvin
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Christy Avery
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Stephanie Engel
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Michelle M Mielke
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Tanya Garcia
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Gerardo Heiss
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| | - Priya Palta
- From the Departments of Epidemiology (Y.L., C.A., S.E., G.H.) and Biostatistics (T.G.), Gillings School of Global Public Health and Departments of Genetics (L.R.) and Neurology (P.P.), School of Medicine, University of North Carolina at Chapel Hill, NC; Department of Epidemiology (J.R.P., E.S.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine (R.H.), Baylor College of Medicine, Houston, TX; Laboratory of Behavioral Neuroscience (K.W.), National Institute on Aging, Bethesda, MD; and Department of Epidemiology and Prevention (M.M.M.), Wake Forest University School of Medicine, Winston-Salem, NC
| |
Collapse
|
7
|
Sun R, Feng J, Wang J. Underlying Mechanisms and Treatment of Cellular Senescence-Induced Biological Barrier Interruption and Related Diseases. Aging Dis 2024; 15:612-639. [PMID: 37450933 PMCID: PMC10917536 DOI: 10.14336/ad.2023.0621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
Given its increasing prevalence, aging is of great concern to researchers worldwide. Cellular senescence is a physiological or pathological cellular state caused by aging and a prominent risk factor for the interruption of the integrity and functionality of human biological barriers. Health barriers play an important role in maintaining microenvironmental homeostasis within the body. The senescence of barrier cells leads to barrier dysfunction and age-related diseases. Cellular senescence has been reported to be a key target for the prevention of age-related barrier diseases, including Alzheimer's disease, Parkinson's disease, age-related macular degeneration, diabetic retinopathy, and preeclampsia. Drugs such as metformin, dasatinib, quercetin, BCL-2 inhibitors, and rapamycin have been shown to intervene in cellular senescence and age-related diseases. In this review, we conclude that cellular senescence is involved in age-related biological barrier impairment. We further outline the cellular pathways and mechanisms underlying barrier impairment caused by cellular senescence and describe age-related barrier diseases associated with senescent cells. Finally, we summarize the currently used anti-senescence pharmacological interventions and discuss their therapeutic potential for preventing age-related barrier diseases.
Collapse
Affiliation(s)
- Ruize Sun
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang, China
| | - Jue Wang
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
8
|
Balczon R, Lin MT, Voth S, Nelson AR, Schupp JC, Wagener BM, Pittet JF, Stevens T. Lung endothelium, tau, and amyloids in health and disease. Physiol Rev 2024; 104:533-587. [PMID: 37561137 PMCID: PMC11281824 DOI: 10.1152/physrev.00006.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/26/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023] Open
Abstract
Lung endothelia in the arteries, capillaries, and veins are heterogeneous in structure and function. Lung capillaries in particular represent a unique vascular niche, with a thin yet highly restrictive alveolar-capillary barrier that optimizes gas exchange. Capillary endothelium surveys the blood while simultaneously interpreting cues initiated within the alveolus and communicated via immediately adjacent type I and type II epithelial cells, fibroblasts, and pericytes. This cell-cell communication is necessary to coordinate the immune response to lower respiratory tract infection. Recent discoveries identify an important role for the microtubule-associated protein tau that is expressed in lung capillary endothelia in the host-pathogen interaction. This endothelial tau stabilizes microtubules necessary for barrier integrity, yet infection drives production of cytotoxic tau variants that are released into the airways and circulation, where they contribute to end-organ dysfunction. Similarly, beta-amyloid is produced during infection. Beta-amyloid has antimicrobial activity, but during infection it can acquire cytotoxic activity that is deleterious to the host. The production and function of these cytotoxic tau and amyloid variants are the subject of this review. Lung-derived cytotoxic tau and amyloid variants are a recently discovered mechanism of end-organ dysfunction, including neurocognitive dysfunction, during and in the aftermath of infection.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Mike T Lin
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Sarah Voth
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States
| | - Amy R Nelson
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Jonas C Schupp
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University, New Haven, Connecticut, United States
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Hannover, Germany
| | - Brant M Wagener
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| |
Collapse
|
9
|
Li X, Chen X, Gao X. Copper and cuproptosis: new therapeutic approaches for Alzheimer's disease. Front Aging Neurosci 2023; 15:1300405. [PMID: 38178962 PMCID: PMC10766373 DOI: 10.3389/fnagi.2023.1300405] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/13/2023] [Indexed: 01/06/2024] Open
Abstract
Copper (Cu) plays a crucial role as a trace element in various physiological processes in humans. Nonetheless, free copper ions accumulate in the brain over time, resulting in a range of pathological changes. Compelling evidence indicates that excessive free copper deposition contributes to cognitive decline in individuals with Alzheimer's disease (AD). Free copper levels in the serum and brain of AD patients are notably elevated, leading to reduced antioxidant defenses and mitochondrial dysfunction. Moreover, free copper accumulation triggers a specific form of cell death, namely copper-dependent cell death (cuproptosis). This article aimed to review the correlation between copper dysregulation and the pathogenesis of AD, along with the primary pathways regulating copper homoeostasis and copper-induced death in AD. Additionally, the efficacy and safety of natural and synthetic agents, including copper chelators, lipid peroxidation inhibitors, and antioxidants, were examined. These treatments can restore copper equilibrium and prevent copper-induced cell death in AD cases. Another aim of this review was to highlight the significance of copper dysregulation and promote the development of pharmaceutical interventions to address it.
Collapse
Affiliation(s)
- Xiao Li
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xinwang Chen
- College of Acupuncture-Moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Acupuncture Clinic of the Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xiyan Gao
- College of Acupuncture-Moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Acupuncture Clinic of the Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
10
|
Alkhalifa AE, Al-Ghraiybah NF, Odum J, Shunnarah JG, Austin N, Kaddoumi A. Blood-Brain Barrier Breakdown in Alzheimer's Disease: Mechanisms and Targeted Strategies. Int J Mol Sci 2023; 24:16288. [PMID: 38003477 PMCID: PMC10671257 DOI: 10.3390/ijms242216288] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
The blood-brain barrier (BBB) is a unique and selective feature of the central nervous system's vasculature. BBB dysfunction has been observed as an early sign of Alzheimer's Disease (AD) before the onset of dementia or neurodegeneration. The intricate relationship between the BBB and the pathogenesis of AD, especially in the context of neurovascular coupling and the overlap of pathophysiology in neurodegenerative and cerebrovascular diseases, underscores the urgency to understand the BBB's role more deeply. Preserving or restoring the BBB function emerges as a potentially promising strategy for mitigating the progression and severity of AD. Molecular and genetic changes, such as the isoform ε4 of apolipoprotein E (ApoEε4), a significant genetic risk factor and a promoter of the BBB dysfunction, have been shown to mediate the BBB disruption. Additionally, receptors and transporters like the low-density lipoprotein receptor-related protein 1 (LRP1), P-glycoprotein (P-gp), and the receptor for advanced glycation end products (RAGEs) have been implicated in AD's pathogenesis. In this comprehensive review, we endeavor to shed light on the intricate pathogenic and therapeutic connections between AD and the BBB. We also delve into the latest developments and pioneering strategies targeting the BBB for therapeutic interventions, addressing its potential as a barrier and a carrier. By providing an integrative perspective, we anticipate paving the way for future research and treatments focused on exploiting the BBB's role in AD pathogenesis and therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S. Donahue Dr., Auburn, AL 36849, USA; (A.E.A.); (N.F.A.-G.); (J.O.); (J.G.S.); (N.A.)
| |
Collapse
|
11
|
De Marchi F, Munitic I, Vidatic L, Papić E, Rački V, Nimac J, Jurak I, Novotni G, Rogelj B, Vuletic V, Liscic RM, Cannon JR, Buratti E, Mazzini L, Hecimovic S. Overlapping Neuroimmune Mechanisms and Therapeutic Targets in Neurodegenerative Disorders. Biomedicines 2023; 11:2793. [PMID: 37893165 PMCID: PMC10604382 DOI: 10.3390/biomedicines11102793] [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: 09/11/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Many potential immune therapeutic targets are similarly affected in adult-onset neurodegenerative diseases, such as Alzheimer's (AD) disease, Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and frontotemporal dementia (FTD), as well as in a seemingly distinct Niemann-Pick type C disease with primarily juvenile onset. This strongly argues for an overlap in pathogenic mechanisms. The commonly researched immune targets include various immune cell subsets, such as microglia, peripheral macrophages, and regulatory T cells (Tregs); the complement system; and other soluble factors. In this review, we compare these neurodegenerative diseases from a clinical point of view and highlight common pathways and mechanisms of protein aggregation, neurodegeneration, and/or neuroinflammation that could potentially lead to shared treatment strategies for overlapping immune dysfunctions in these diseases. These approaches include but are not limited to immunisation, complement cascade blockade, microbiome regulation, inhibition of signal transduction, Treg boosting, and stem cell transplantation.
Collapse
Affiliation(s)
- Fabiola De Marchi
- Department of Neurology and ALS Centre, University of Piemonte Orientale, Maggiore Della Carità Hospital, Corso Mazzini 18, 28100 Novara, Italy;
| | - Ivana Munitic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000 Rijeka, Croatia;
| | - Lea Vidatic
- Laboratory for Neurodegenerative Disease Research, Division of Molecular Medicine, Ruder Boskovic Institute, 10000 Zagreb, Croatia;
| | - Eliša Papić
- Department of Neurology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia; (E.P.); (V.R.); (V.V.)
- Department of Neurology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Valentino Rački
- Department of Neurology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia; (E.P.); (V.R.); (V.V.)
- Department of Neurology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Jerneja Nimac
- Department of Biotechnology, Jozef Stefan Institute, SI-1000 Ljubljana, Slovenia; (J.N.); (B.R.)
- Graduate School of Biomedicine, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Igor Jurak
- Molecular Virology Laboratory, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000 Rijeka, Croatia;
| | - Gabriela Novotni
- Department of Cognitive Neurology and Neurodegenerative Diseases, University Clinic of Neurology, Medical Faculty, University Ss. Cyril and Methodius, 91701 Skoplje, North Macedonia;
| | - Boris Rogelj
- Department of Biotechnology, Jozef Stefan Institute, SI-1000 Ljubljana, Slovenia; (J.N.); (B.R.)
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Vladimira Vuletic
- Department of Neurology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia; (E.P.); (V.R.); (V.V.)
- Department of Neurology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Rajka M. Liscic
- Department of Neurology, Sachsenklinik GmbH, Muldentalweg 1, 04828 Bennewitz, Germany;
| | - Jason R. Cannon
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA;
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34149 Trieste, Italy;
| | - Letizia Mazzini
- Department of Neurology and ALS Centre, University of Piemonte Orientale, Maggiore Della Carità Hospital, Corso Mazzini 18, 28100 Novara, Italy;
| | - Silva Hecimovic
- Laboratory for Neurodegenerative Disease Research, Division of Molecular Medicine, Ruder Boskovic Institute, 10000 Zagreb, Croatia;
| |
Collapse
|
12
|
Shan M, Bai Y, Fang X, Lan X, Zhang Y, Cao Y, Zhu D, Luo H. American Ginseng for the Treatment of Alzheimer's Disease: A Review. Molecules 2023; 28:5716. [PMID: 37570686 PMCID: PMC10420665 DOI: 10.3390/molecules28155716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent degenerative condition that is increasingly affecting populations globally. American ginseng (AG) has anti-AD bioactivity, and ginsenosides, as the main active components of AG, have shown strong anti-AD effects in both in vitro and in vivo studies. It has been reported that ginsenosides can inhibit amyloid β-protein (Aβ) production and deposition, tau phosphorylation, apoptosis and cytotoxicity, as well as possess anti-oxidant and anti-inflammatory properties, thus suppressing the progression of AD. In this review, we aim to provide a comprehensive overview of the pathogenesis of AD, the potential anti-AD effects of ginsenosides found in AG, and the underlying molecular mechanisms associated with these effects. Additionally, we will discuss the potential use of AG in the treatment of AD, and how ginsenosides in AG may exert more potent anti-AD effects in vivo may be a direction for further research.
Collapse
Affiliation(s)
- Mengyao Shan
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Pharmaceutical Chemistry and Traditional Chinese Medicine Chemistry, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yunfan Bai
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Pharmaceutical Chemistry and Traditional Chinese Medicine Chemistry, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Xiaoxue Fang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Pharmaceutical Chemistry and Traditional Chinese Medicine Chemistry, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Xintian Lan
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Pharmaceutical Chemistry and Traditional Chinese Medicine Chemistry, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yegang Zhang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Pharmaceutical Chemistry and Traditional Chinese Medicine Chemistry, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yiming Cao
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Pharmaceutical Chemistry and Traditional Chinese Medicine Chemistry, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Difu Zhu
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Biopharmaceutical and Health Food, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Haoming Luo
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.S.); (Y.B.); (X.F.); (X.L.); (Y.Z.); (Y.C.)
- Department of Pharmaceutical Chemistry and Traditional Chinese Medicine Chemistry, Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|
13
|
Kilian JG, Mejias-Ortega M, Hsu HW, Herman DA, Vidal J, Arechavala RJ, Renusch S, Dalal H, Hasen I, Ting A, Rodriguez-Ortiz CJ, Lim SL, Lin X, Vu J, Saito T, Saido TC, Kleinman MT, Kitazawa M. Exposure to quasi-ultrafine particulate matter accelerates memory impairment and Alzheimer's disease-like neuropathology in the AppNL-G-F knock-in mouse model. Toxicol Sci 2023; 193:175-191. [PMID: 37074955 PMCID: PMC10230292 DOI: 10.1093/toxsci/kfad036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023] Open
Abstract
Exposure to traffic-related air pollution consisting of particulate matter (PM) is associated with cognitive decline leading to Alzheimer's disease (AD). In this study, we sought to examine the neurotoxic effects of exposure to ultrafine PM and how it exacerbates neuronal loss and AD-like neuropathology in wildtype (WT) mice and a knock-in mouse model of AD (AppNL-G-F/+-KI) when the exposure occurs at a prepathologic stage or at a later age with the presence of neuropathology. AppNL-G-F/+-KI and WT mice were exposed to concentrated ultrafine PM from local ambient air in Irvine, California, for 12 weeks, starting at 3 or 9 months of age. Particulate matter-exposed animals received concentrated ultrafine PM up to 8 times above the ambient levels, whereas control animals were exposed to purified air. Particulate matter exposure resulted in a marked impairment of memory tasks in prepathologic AppNL-G-F/+-KI mice without measurable changes in amyloid-β pathology, synaptic degeneration, and neuroinflammation. At aged, both WT and AppNL-G-F/+-KI mice exposed to PM showed a significant memory impairment along with neuronal loss. In AppNL-G-F/+-KI mice, we also detected an increased amyloid-β buildup and potentially harmful glial activation including ferritin-positive microglia and C3-positive astrocytes. Such glial activation could promote the cascade of degenerative consequences in the brain. Our results suggest that exposure to PM impairs cognitive function at both ages while exacerbation of AD-related pathology and neuronal loss may depend on the stage of pathology, aging, and/or state of glial activation. Further studies will be required to unveil the neurotoxic role of glial activation activated by PM exposure.
Collapse
Affiliation(s)
- Jason G Kilian
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
- Institute for Memory Impairmants and Neurological Disorders (UCI MIND), University of California, Irvine, California 92697, USA
| | - Marina Mejias-Ortega
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
- Department of Cell Biology, Genetics and Physiology, Facultad de Ciencias, Instituto de Investigacion Biomedica de Malaga-IBIMA, Universidad de Malaga, Malaga, Spain
- Centro de Investigación Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Heng-Wei Hsu
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
- Institute for Memory Impairmants and Neurological Disorders (UCI MIND), University of California, Irvine, California 92697, USA
| | - David A Herman
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
| | - Janielle Vidal
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
- Institute for Memory Impairmants and Neurological Disorders (UCI MIND), University of California, Irvine, California 92697, USA
| | - Rebecca J Arechavala
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
| | - Samantha Renusch
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
| | - Hansal Dalal
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
| | - Irene Hasen
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
| | - Amanda Ting
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
| | - Carlos J Rodriguez-Ortiz
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
- Institute for Memory Impairmants and Neurological Disorders (UCI MIND), University of California, Irvine, California 92697, USA
| | - Siok-Lam Lim
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
- Institute for Memory Impairmants and Neurological Disorders (UCI MIND), University of California, Irvine, California 92697, USA
| | - Xiaomeng Lin
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
| | - Joan Vu
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
- Institute for Memory Impairmants and Neurological Disorders (UCI MIND), University of California, Irvine, California 92697, USA
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University, Nagoya, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Michael T Kleinman
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
| | - Masashi Kitazawa
- Department of Environmental and Occupational Health, Center for Occupational and Environmental Health (COEH), University of California, Irvine, California 92697-1830, USA
- Institute for Memory Impairmants and Neurological Disorders (UCI MIND), University of California, Irvine, California 92697, USA
| |
Collapse
|
14
|
Endothelial Dysfunction in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:ijms24032909. [PMID: 36769234 PMCID: PMC9918222 DOI: 10.3390/ijms24032909] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
The cerebral vascular system stringently regulates cerebral blood flow (CBF). The components of the blood-brain barrier (BBB) protect the brain from pathogenic infections and harmful substances, efflux waste, and exchange substances; however, diseases develop in cases of blood vessel injuries and BBB dysregulation. Vascular pathology is concurrent with the mechanisms underlying aging, Alzheimer's disease (AD), and vascular dementia (VaD), which suggests its involvement in these mechanisms. Therefore, in the present study, we reviewed the role of vascular dysfunction in aging and neurodegenerative diseases, particularly AD and VaD. During the development of the aforementioned diseases, changes occur in the cerebral blood vessel morphology and local cells, which, in turn, alter CBF, fluid dynamics, and vascular integrity. Chronic vascular inflammation and blood vessel dysregulation further exacerbate vascular dysfunction. Multitudinous pathogenic processes affect the cerebrovascular system, whose dysfunction causes cognitive impairment. Knowledge regarding the pathophysiology of vascular dysfunction in neurodegenerative diseases and the underlying molecular mechanisms may lead to the discovery of clinically relevant vascular biomarkers, which may facilitate vascular imaging for disease prevention and treatment.
Collapse
|
15
|
Brook ES, D'Alonzo ZJ, Lam V, Chan DC, Dhaliwal SS, Watts GF, Mamo JCL, Takechi R. Plasma Amyloid-β Homeostasis Is Associated with Body Mass Index and Weight Loss in People with Overweight and Obesity. J Alzheimers Dis 2023; 93:653-664. [PMID: 37066906 DOI: 10.3233/jad-220529] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
BACKGROUND Obesity is linked to a higher incidence of Alzheimer's disease (AD). Studies show that plasma amyloid-β (Aβ) dyshomeostasis, particularly low 42/40 ratio indicates a heightened risk for developing AD. However, the relationship between body mass index (BMI) and circulating plasma Aβ has not been extensively studied. OBJECTIVE We hypothesized that people with a high BMI have altered plasma Aβ homeostasis compared with people with a lower BMI. We also tested whether reducing BMI by calorie-restriction could normalize plasma concentrations of Aβ. METHODS Plasma concentrations of Aβ40, Aβ42, and Aβ42/40 ratio were measured in 106 participants with BMIs classified as lean, overweight, or obese. From this cohort, twelve participants with overweight or obese BMIs entered a 12-week calorie-restriction weight loss program. We then tested whether decreasing BMI affected plasma Aβ concentrations. RESULTS Plasma Aβ42/40 ratio was 17.54% lower in participants with an obese BMI compared to lean participants (p < 0.0001), and 11.76% lower compared to participants with an overweight BMI (p < 0.0001). The weight loss regimen decreased BMI by an average of 4.02% (p = 0.0005) and was associated with a 6.5% decrease in plasma Aβ40 (p = 0.0425). However, weight loss showed negligible correlations with plasma Aβ40, Aβ42, and Aβ42/40 ratio. CONCLUSION Obesity is associated with aberrant plasma Aβ homeostasis which may be associated with an increased risk for AD. Weight loss appears to lower Aβ40, but large-scale longitudinal studies in addition to molecular studies are required to elucidate the underlying mechanisms of how obesity and weight loss influence plasma Aβ homeostasis.
Collapse
Affiliation(s)
- Emily S Brook
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Bentley, Australia
| | - Zachary J D'Alonzo
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Bentley, Australia
| | - Virginie Lam
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia
- School of Population Health, Faculty of Health Sciences, Curtin University, Bentley, Australia
| | - Dick C Chan
- Medical School, University of Western Australia, Perth, Australia
| | - Satvinder S Dhaliwal
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia
- Duke-NUS Medical School, National University of Singapore, Singapore
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Pulau Pinang, Malaysia
- Singapore University of Social Sciences, Singapore
| | - Geraldb F Watts
- Medical School, University of Western Australia, Perth, Australia
- Cardiometabolic Service, Department of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Australia
| | - John C L Mamo
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia
- School of Population Health, Faculty of Health Sciences, Curtin University, Bentley, Australia
- Perron Institute of Neurological and Translational Sciences, Nedlands, Australia
| | - Ryusuke Takechi
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia
- School of Population Health, Faculty of Health Sciences, Curtin University, Bentley, Australia
| |
Collapse
|
16
|
Apolipoprotein E in Cardiometabolic and Neurological Health and Diseases. Int J Mol Sci 2022; 23:ijms23179892. [PMID: 36077289 PMCID: PMC9456500 DOI: 10.3390/ijms23179892] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 11/17/2022] Open
Abstract
A preponderance of evidence obtained from genetically modified mice and human population studies reveals the association of apolipoprotein E (apoE) deficiency and polymorphisms with pathogenesis of numerous chronic diseases, including atherosclerosis, obesity/diabetes, and Alzheimer’s disease. The human APOE gene is polymorphic with three major alleles, ε2, ε3 and ε4, encoding apoE2, apoE3, and apoE4, respectively. The APOE gene is expressed in many cell types, including hepatocytes, adipocytes, immune cells of the myeloid lineage, vascular smooth muscle cells, and in the brain. ApoE is present in subclasses of plasma lipoproteins, and it mediates the clearance of atherogenic lipoproteins from plasma circulation via its interaction with LDL receptor family proteins and heparan sulfate proteoglycans. Extracellular apoE also interacts with cell surface receptors and confers signaling events for cell regulation, while apoE expressed endogenously in various cell types regulates cell functions via autocrine and paracrine mechanisms. This review article focuses on lipoprotein transport-dependent and -independent mechanisms by which apoE deficiency or polymorphisms contribute to cardiovascular disease, metabolic disease, and neurological disorders.
Collapse
|
17
|
Gong M, Jia J. Contribution of blood-brain barrier-related blood-borne factors for Alzheimer’s disease vs. vascular dementia diagnosis: A pilot study. Front Neurosci 2022; 16:949129. [PMID: 36003963 PMCID: PMC9393528 DOI: 10.3389/fnins.2022.949129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022] Open
Abstract
Background Alzheimer’s disease (AD) and vascular dementia (VaD) are the two most common types of neurodegenerative dementia among the elderly with similar symptoms of cognitive decline and overlapping neuropsychological profiles. Biological markers to distinguish patients with VaD from AD would be very useful. We aimed to investigate the expression of blood-brain barrier (BBB)-related blood-borne factors of soluble low-density lipoprotein receptor-related protein 1 (sLRP1), cyclophilin A (CyPA), and matrix metalloproteinase 9 (MMP9) and its correlation with cognitive function between patients with AD and VaD. Materials and methods Plasma levels of sLRP1, CyPA, and MMP9 were analyzed in 26 patients with AD, 27 patients with VaD, and 27 normal controls (NCs). Spearman’s rank correlation analysis was used to explore the relationships among biomarker levels, cognitive function, and imaging references. Receiver operating characteristic (ROC) curve analysis was used to discriminate the diagnosis of AD and VaD. Results Among these BBB-related factors, plasma CyPA levels in the VaD group were significantly higher than that in the AD group (p < 0.05). Plasma sLRP1 levels presented an increasing trend in VaD while maintaining slightly low levels in patients with AD (p > 0.05). Plasma MMP9 in different diagnostic groups displayed the following trend: VaD group > AD group > NC group, but the difference was not statistically significant (p > 0.05). Furthermore, plasma sLRP1 levels were positively related to MoCA scores, and plasma CyPA levels were significantly correlated with MTA scores (p < 0.05) in the AD group. Plasma MMP9 levels were negatively correlated with MoCA scores (p < 0.05) in the VaD groups. No significant correlation was detected between the other factors and different cognitive scores (p > 0.05). ROC analysis showed a good preference of plasma CyPA [AUC = 0.725, 95% CI (0.586–0.865); p = 0.0064] in diagnosis. Conclusion The plasma CyPA level is a reference index when distinguishing between an AD and subcortical ischemic vascular dementia (SIVD) diagnosis. Blood-derived factors associated with the BBB may provide new insights into the differential diagnosis of neurodegenerative dementia and warrant further investigation.
Collapse
Affiliation(s)
- Min Gong
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
- Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, China
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, China
- Center of Alzheimer’s Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- *Correspondence: Jianping Jia,
| |
Collapse
|
18
|
Ahmed S, Jing Y, Mockett BG, Zhang H, Abraham WC, Liu P. Partial Endothelial Nitric Oxide Synthase Deficiency Exacerbates Cognitive Deficit and Amyloid Pathology in the APPswe/PS1ΔE9 Mouse Model of Alzheimer’s Disease. Int J Mol Sci 2022; 23:ijms23137316. [PMID: 35806318 PMCID: PMC9266765 DOI: 10.3390/ijms23137316] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 01/25/2023] Open
Abstract
Increasing evidence implicates endothelial dysfunction in the pathogenesis of Alzheimer’s disease (AD). Nitric oxide (NO) derived from endothelial NO synthase (eNOS) is essential in maintaining cerebrovascular function and can modulate the production and clearance of amyloid beta (Aβ). APPswe/PSdE1 (APP/PS1) mice display age-related Aβ accumulation and memory deficits. In order to make the model more clinically relevant with an element of endothelial dysfunction, we generated APP/PS1/eNOS+/− mice by crossing complete eNOS deficient (eNOS−/−) mice and APP/PS1 mice. APP/PS1/eNOS+/− mice at 8 months of age displayed a more severe spatial working memory deficit relative to age-matched APP/PS1 mice. Moreover, immunohistochemistry and immunoblotting revealed significantly increased Aβ plaque load in the brains of APP/PS1/eNOS+/− mice, concomitant with upregulated BACE-1 (hence increased Aβ production), downregulated insulin-degrading enzyme (hence reduced Aβ clearance) and increased immunoreactivity and expression of microglia. The present study, for the first time, demonstrated that partial eNOS deficiency exacerbated behavioral dysfunction, Aβ brain deposition, and microglial pathology in APP/PS1 mice, further implicating endothelial dysfunction in the pathogenesis of AD. The present findings also provide the scientific basis for developing preventive and/or therapeutic strategies by targeting endothelial dysfunction.
Collapse
Affiliation(s)
- Sara Ahmed
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand; (S.A.); (Y.J.)
| | - Yu Jing
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand; (S.A.); (Y.J.)
| | - Bruce G. Mockett
- Department of Psychology, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand; (B.G.M.); (W.C.A.)
| | - Hu Zhang
- School of Pharmacy, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand;
| | - Wickliffe C. Abraham
- Department of Psychology, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand; (B.G.M.); (W.C.A.)
| | - Ping Liu
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand; (S.A.); (Y.J.)
- Correspondence:
| |
Collapse
|
19
|
N-terminally truncated Aβ4-x proteoforms and their relevance for Alzheimer's pathophysiology. Transl Neurodegener 2022; 11:30. [PMID: 35641972 PMCID: PMC9158284 DOI: 10.1186/s40035-022-00303-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 04/21/2022] [Indexed: 01/22/2023] Open
Abstract
Background The molecular heterogeneity of Alzheimer’s amyloid-β (Aβ) deposits extends well beyond the classic Aβ1-40/Aβ1-42 dichotomy, substantially expanded by multiple post-translational modifications that increase the proteome diversity. Numerous truncated fragments consistently populate the brain Aβ peptidome, and their homeostatic regulation and potential contribution to disease pathogenesis are largely unknown. Aβ4-x peptides have been reported as major components of plaque cores and the limited studies available indicate their relative abundance in Alzheimer’s disease (AD). Methods Immunohistochemistry was used to assess the topographic distribution of Aβ4-x species in well-characterized AD cases using custom-generated monoclonal antibody 18H6—specific for Aβ4-x species and blind for full-length Aβ1-40/Aβ1-42—in conjunction with thioflavin-S and antibodies recognizing Aβx-40 and Aβx-42 proteoforms. Circular dichroism, thioflavin-T binding, and electron microscopy evaluated the biophysical and aggregation/oligomerization properties of full-length and truncated synthetic homologues, whereas stereotaxic intracerebral injections of monomeric and oligomeric radiolabeled homologues in wild-type mice were used to evaluate their brain clearance characteristics. Results All types of amyloid deposits contained the probed Aβ epitopes, albeit expressed in different proportions. Aβ4-x species showed preferential localization within thioflavin-S-positive cerebral amyloid angiopathy and cored plaques, strongly suggesting poor clearance characteristics and consistent with the reduced solubility and enhanced oligomerization of their synthetic homologues. In vivo clearance studies demonstrated a fast brain efflux of N-terminally truncated and full-length monomeric forms whereas their oligomeric counterparts—particularly of Aβ4-40 and Aβ4-42—consistently exhibited enhanced brain retention. Conclusions The persistence of aggregation-prone Aβ4-x proteoforms likely contributes to the process of amyloid formation, self-perpetuating the amyloidogenic loop and exacerbating amyloid-mediated pathogenic pathways.
Collapse
|
20
|
Vargas-George S, Dave KR. Models of cerebral amyloid angiopathy-related intracerebral hemorrhage. BRAIN HEMORRHAGES 2022. [DOI: 10.1016/j.hest.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
21
|
Collins AE, Saleh TM, Kalisch BE. Naturally Occurring Antioxidant Therapy in Alzheimer's Disease. Antioxidants (Basel) 2022; 11:213. [PMID: 35204096 PMCID: PMC8868221 DOI: 10.3390/antiox11020213] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 02/06/2023] Open
Abstract
It is estimated that the prevalence rate of Alzheimer's disease (AD) will double by the year 2040. Although currently available treatments help with symptom management, they do not prevent, delay the progression of, or cure the disease. Interestingly, a shared characteristic of AD and other neurodegenerative diseases and disorders is oxidative stress. Despite profound evidence supporting the role of oxidative stress in the pathogenesis and progression of AD, none of the currently available treatment options address oxidative stress. Recently, attention has been placed on the use of antioxidants to mitigate the effects of oxidative stress in the central nervous system. In preclinical studies utilizing cellular and animal models, natural antioxidants showed therapeutic promise when administered alone or in combination with other compounds. More recently, the concept of combination antioxidant therapy has been explored as a novel approach to preventing and treating neurodegenerative conditions that present with oxidative stress as a contributing factor. In this review, the relationship between oxidative stress and AD pathology and the neuroprotective role of natural antioxidants from natural sources are discussed. Additionally, the therapeutic potential of natural antioxidants as preventatives and/or treatment for AD is examined, with special attention paid to natural antioxidant combinations and conjugates that are currently being investigated in human clinical trials.
Collapse
Affiliation(s)
| | | | - Bettina E. Kalisch
- Department of Biomedical Sciences and Collaborative Specialization in Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.E.C.); (T.M.S.)
| |
Collapse
|
22
|
Chasovskikh NY, Chizhik EE, Bobrysheva AA. Bioinformatic Annotation of Genes for Alzheimer’s Disease and Coronary Heart Disease. RUSS J GENET+ 2021. [DOI: 10.1134/s102279542111003x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
23
|
Zhou XB, Zhang YX, Zhou CX, Ma JJ. Chinese Herbal Medicine Adjusting Brain Microenvironment via Mediating Central Nervous System Lymphatic Drainage in Alzheimer's Disease. Chin J Integr Med 2021; 28:176-184. [PMID: 34731433 DOI: 10.1007/s11655-021-3342-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 02/05/2023]
Abstract
Due to its complex pathogenesis and lack of effective therapeutic methods, Alzheimer's disease (AD) has become a severe public health problem worldwide. Recent studies have discovered the function of central nervous system lymphatic drainage, which provides a new strategy for the treatment of AD. Chinese herbal medicine (CHM) has been considered as a cure for AD for hundreds of years in China, and its effect on scavenging β-amyloid protein in the brain of AD patients has been confirmed. In this review, the mechanism of central nervous system lymphatic drainage and the regulatory functions of CHM on correlation factors were briefly summarized. The advances in our understanding regarding the treatment of AD via regulating the central lymphatic system with CHM will promote the clinical application of CHM in AD patients and the discovery of new therapeutic drugs.
Collapse
Affiliation(s)
- Xi-Bin Zhou
- Department of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Yu-Xing Zhang
- Department of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Chun-Xiang Zhou
- Department of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China.,Department of Traditional Chinese Medicine, Nanjing BenQ Hospital, Nanjing, 210036, China
| | - Jun-Jie Ma
- Department of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China.
| |
Collapse
|
24
|
Park S, Kim Y. Bias-generating factors in biofluid amyloid-β measurements for Alzheimer's disease diagnosis. Biomed Eng Lett 2021; 11:287-295. [PMID: 34616582 DOI: 10.1007/s13534-021-00201-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 01/03/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia worldwide, yet the dearth of readily accessible diagnostic biomarkers is a substantial hindrance towards progressing to effective preventive and therapeutic approaches. Due to a long delay between cerebral amyloid-β (Aβ) accumulation and the onset of cognitive impairments, biomarkers that reflect Aβ pathology and enable routine screening for disease progression are of urgent need for application in the clinical diagnosis of AD. According to accumulating evidences, cerebrospinal fluid (CSF) and plasma offer windows to the brain as they allow monitoring of biochemical changes in the brain. Considering the high availability and accuracy in depicting Aβ deposition in the brain, Aβ levels in CSF and plasma are regarded as promising fluid biomarkers for the diagnosis of AD patients at an early stage. However, clinical data with intra- and interindividual variations in the concentrations of CSF and plasma Aβ implicate the need to reevaluate current Aβ detection methods and establish a standardized operating procedure. Therefore, this review introduces three bias-generating factors in biofluid Aβ measurement that may hamper the accurate Aβ quantification and how such complications can be overcome for the widespread implementation of fluid Aβ detection in clinical practice.
Collapse
Affiliation(s)
- Sohui Park
- Department of Pharmacy, Department of Integrative Biotechnology and Translational Medicine, and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983 Republic of Korea
| | - YoungSoo Kim
- Department of Pharmacy, Department of Integrative Biotechnology and Translational Medicine, and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983 Republic of Korea
| |
Collapse
|
25
|
Quintela T, Furtado A, Duarte AC, Gonçalves I, Myung J, Santos CRA. The role of circadian rhythm in choroid plexus functions. Prog Neurobiol 2021; 205:102129. [PMID: 34343629 DOI: 10.1016/j.pneurobio.2021.102129] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 12/17/2022]
Abstract
For several years, a great effort has been devoted to understand how circadian oscillations in physiological processes are determined by the circadian clock system. This system is composed by the master clock at the suprachiasmatic nucleus which sets the pace and tunes peripheral clocks in several organs. It was recently demonstrated that the choroid plexus epithelial cells that compose the blood-cerebrospinal fluid barrier hold a circadian clock which might control their multiple functions with implications for the maintenance of brain homeostasis. However, the choroid plexus activities regulated by its inner clock are still largely unknown. In this review, we propose that several choroid plexus functions might be regulated by the circadian clock, alike in other tissues. We provide evidences that the timing of cerebrospinal fluid secretion, clearance of amyloid-beta peptides and xenobiotics, and the barrier function of the blood-cerebrospinal fluid barrier are regulated by the circadian clock. These data, highlight that the circadian regulation of the blood-cerebrospinal fluid barrier must be taken into consideration for enhancing drug delivery to central nervous system disorders.
Collapse
Affiliation(s)
- Telma Quintela
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| | - André Furtado
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana C Duarte
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Isabel Gonçalves
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Jihwan Myung
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, No. 172-1 Sec. 2 Keelung Road, Da'an District, Taipei 106, Taiwan; Brain and Consciousness Research Centre, Shuang Ho Hospital, Ministry of Health and Welfare, No. 291 Zhongzheng Road, Zhonghe District, New Taipei City 235, Taiwan
| | - Cecília R A Santos
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
26
|
Abstract
BACKGROUND SARS-CoV-2, a coronavirus (CoV), is known to cause acute respiratory distress syndrome, and a number of non-respiratory complications, particularly in older male patients with prior health conditions, such as obesity, diabetes and hypertension. These prior health conditions are associated with vascular dysfunction, and the CoV disease 2019 (COVID-19) complications include multiorgan failure and neurological problems. While the main route of entry into the body is inhalation, this virus has been found in many tissues, including the choroid plexus and meningeal vessels, and in neurons and CSF. MAIN BODY We reviewed SARS-CoV-2/COVID-19, ACE2 distribution and beneficial effects, the CNS vascular barriers, possible mechanisms by which the virus enters the brain, outlined prior health conditions (obesity, hypertension and diabetes), neurological COVID-19 manifestation and the aging cerebrovascualture. The overall aim is to provide the general reader with a breadth of information on this type of virus and the wide distribution of its main receptor so as to better understand the significance of neurological complications, uniqueness of the brain, and the pre-existing medical conditions that affect brain. The main issue is that there is no sound evidence for large flux of SARS-CoV-2 into brain, at present, compared to its invasion of the inhalation pathways. CONCLUSIONS While SARS-CoV-2 is detected in brains from severely infected patients, it is unclear on how it gets there. There is no sound evidence of SARS-CoV-2 flux into brain to significantly contribute to the overall outcomes once the respiratory system is invaded by the virus. The consensus, based on the normal route of infection and presence of SARS-CoV-2 in severely infected patients, is that the olfactory mucosa is a possible route into brain. Studies are needed to demonstrate flux of SARS-CoV-2 into brain, and its replication in the parenchyma to demonstrate neuroinvasion. It is possible that the neurological manifestations of COVID-19 are a consequence of mainly cardio-respiratory distress and multiorgan failure. Understanding potential SARS-CoV-2 neuroinvasion pathways could help to better define the non-respiratory neurological manifestation of COVID-19.
Collapse
Affiliation(s)
- Conor McQuaid
- Department of Neuroscience, University of Rochester, URMC, 601 Elmwood Avenue, Rochester, NY 14642 USA
| | - Molly Brady
- Department of Neuroscience, University of Rochester, URMC, 601 Elmwood Avenue, Rochester, NY 14642 USA
| | - Rashid Deane
- Department of Neuroscience, University of Rochester, URMC, 601 Elmwood Avenue, Rochester, NY 14642 USA
| |
Collapse
|
27
|
Ouyang Q, Meng Y, Zhou W, Tong J, Cheng Z, Zhu Q. New advances in brain-targeting nano-drug delivery systems for Alzheimer's disease. J Drug Target 2021; 30:61-81. [PMID: 33983096 DOI: 10.1080/1061186x.2021.1927055] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide and its incidence is increasing due to the ageing population. Currently, the main limitations of AD treatment are low blood-brain barrier permeability, severe off-target of drugs, and immune abnormality. In this review, four hypotheses for Alzheimer's pathogenesis and three challenges for Alzheimer's drug delivery are discussed. In addition, this article summarises the different strategies of brain targeting nano-drug delivery systems (NDDSs) developed in the last 10 years. These strategies include receptor-mediated (transferrin receptor, low-density lipoprotein receptor-related protein, lactoferrin receptor, etc.), adsorption-mediated (cationic, alkaline polypeptide, cell-penetrating peptides, etc.), and transporter-mediated (P-gp, GLUT1, etc.). Moreover, it provides insights into novel strategies used in AD, such as exosomes, virus-like particles, and cell membrane coating particles. Hence, this review will help researchers to understand the current progress in the field of NDDSs for the central nervous system and find new directions for AD therapy.HighlightsCharacteristics and challenges based on the pathogenesis of AD were discussed.Recent advances in novel brain-targeting NDDSs for AD over the past 10 years were summarised.
Collapse
Affiliation(s)
- Qin Ouyang
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Yingcai Meng
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Jianbin Tong
- Department of Anaesthesiology, Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China.,Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Zeneng Cheng
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Qubo Zhu
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| |
Collapse
|
28
|
Olsen I. Possible effects of Porphyromonas gingivalis on the blood-brain barrier in Alzheimer's disease. Expert Rev Anti Infect Ther 2021; 19:1367-1371. [PMID: 33938372 DOI: 10.1080/14787210.2021.1925540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Ingar Olsen
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway,
| |
Collapse
|
29
|
Soluble Receptors Affecting Stroke Outcomes: Potential Biomarkers and Therapeutic Tools. Int J Mol Sci 2021; 22:ijms22031108. [PMID: 33498620 PMCID: PMC7865279 DOI: 10.3390/ijms22031108] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/16/2021] [Accepted: 01/19/2021] [Indexed: 02/06/2023] Open
Abstract
Soluble receptors are widely understood to be freestanding moieties formed via cleavage from their membrane-bound counterparts. They have unique structures, are found among various receptor families, and have intriguing mechanisms of generation and release. Soluble receptors’ ability to exhibit pleiotropic action by receptor modulation or by exhibiting a dual role in cytoprotection and neuroinflammation is concentration dependent and has continually mystified researchers. Here, we have compiled findings from preclinical and clinical studies to provide insights into the role of soluble/decoy receptors, focusing on the soluble cluster of differentiation 36, the soluble cluster of differentiation 163, and soluble lipoprotein-related protein 1 (sCD36, sCD163, and sLRP1, respectively) and the functions they could likely serve in the management of stroke, as they would notably regulate the bioavailability of the hemoglobin and heme after red blood cell lysis. The key roles that these soluble receptors play in inflammation, oxidative stress, and the related pharmacotherapeutic potential in improving stroke outcomes are described. The precise pleiotropic physiological functions of soluble receptors remain unclear, and further scientific investigation/validation is required to establish their respective role in diagnosis and therapy.
Collapse
|
30
|
Zhu F, Wang W, Zhang F, Dhinakaran MK, Wang Y, Wang R, Cheng J, Toimil-Molares ME, Trautmann C, Li H. Selective transmembrane transport of Aβ protein regulated by tryptophan enantiomers. Chem Commun (Camb) 2021; 57:215-218. [PMID: 33300917 DOI: 10.1039/d0cc06104b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Tryptophan enantiomers (d/l-Trp) were introduced into artificial nanochannels to regulate the chiral selective transport of Aβ proteins. The l-Trp channel performs effectively selectivity for the transport of Aβ protein, which would provide a new perspective for the pathological studies of Alzheimer's disease.
Collapse
Affiliation(s)
- Fei Zhu
- Key Laboratory of Pesticide and Chemical Biology (CCNU), Ministry of Education, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Edler MK, Johnson CT, Ahmed HS, Richardson JR. Age, sex, and regional differences in scavenger receptor CD36 in the mouse brain: Potential relevance to cerebral amyloid angiopathy and Alzheimer's disease. J Comp Neurol 2020; 529:2209-2226. [PMID: 33319367 DOI: 10.1002/cne.25089] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 12/29/2022]
Abstract
Scavenger receptor CD36 contributes significantly to lipid homeostasis, inflammation, and amyloid deposition, while CD36 deficiency is associated with restored cerebrovascular function in an Alzheimer's disease (AD) mouse model. Yet the distribution of CD36 has not been examined in the brain. Here, we characterized CD36 gene and protein expression in the brains of young, middle aged, aged, and elderly male and female C57BL/6J mice. Age-related increases in CD36 mRNA expression were observed in the male hippocampus and female midbrain. Additionally, male mice had greater CD36 mRNA expression than females in the striatum, hippocampus, and midbrain. CD36 protein was primarily expressed intravascularly, and this expression differed by region, age, and sex in the mouse brain. Although male mice brains demonstrated an increase in CD36 protein with age in several cortices, basal ganglia, hippocampus, and midbrain, a decrease with age was observed in female mice in the same regions. These data suggest that distinctive age, region, and sex expression of CD36 in the brain may contribute to Aβ deposition and neuroinflammation in AD.
Collapse
Affiliation(s)
- Melissa K Edler
- Department of Anthropology, Kent State University, Kent, Ohio, USA.,School of Biomedical Sciences, Kent State University, Kent, Ohio, USA.,Brain Health Research Institute, Kent State University, Kent, Ohio, USA.,Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Cooper T Johnson
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Hashim S Ahmed
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Jason R Richardson
- Robert Stempel School of Public Health and Social Work, Florida International University, Miami, Florida, USA
| |
Collapse
|
32
|
Shimokawa T, Nabeka H, Khan SI, Yamamiya K, Doihara T, Kobayashi N, Wakisaka H, Matsuda S. Prosaposin in the rat oviductal epithelial cells. Cell Tissue Res 2020; 383:1191-1202. [PMID: 33242172 DOI: 10.1007/s00441-020-03339-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 11/05/2020] [Indexed: 10/22/2022]
Abstract
Prosaposin (PSAP) has two forms: a precursor and a secreted form. The secreted form has neurotrophic, myelinotrophic, and myotrophic properties. The precursor form is a precursor protein of saposins A-D. Although the distribution of PSAP in male reproductive organs is well known, its distribution in female reproductive organs, especially in the oviduct, is unclear. Immunoblots and immunohistochemistry of oviducts showed that oviductal tissues contain PSAP proteins, and a significant increase in PSAP was observed in the estrus-metestrus phase compared to the diestrus-proestrus phase in the ampulla. To identify PSAP trafficking in cells, double-immunostaining was performed with antibodies against PSAP in combination with sortilin, mannose 6 phosphate receptor (M6PR), or low-density lipoprotein receptor-related protein 1 (LRP1). PSAP and sortilin double-positive reactions were observed near the nuclei, as well as in the apical portion of microvillous epithelial cells, whereas these reactions were only observed near the nuclei of ciliated epithelial cells. PSAP and M6PR double-positive reactions were observed near the nuclei of microvillous and ciliated epithelial cells. PSAP and M6PR double-positive reactions were also observed in the apical portion of microvillous epithelial cells. PSAP and LRP1 double-positive reactions were observed in the plasma membrane and apical portion of both microvillous and ciliated epithelial cells. Immunoelectron staining revealed PSAP immunoreactive small vesicles with exocytotic features at the apical portion of microvillous epithelial cells. These findings suggest that PSAP is present in the oviductal epithelium and has a pivotal role during pregnancy in providing an optimal environment for gametes and/or sperm in the ampulla.
Collapse
Affiliation(s)
- Tetsuya Shimokawa
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0212, Japan.
| | - Hiroaki Nabeka
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0212, Japan
| | - Sakirul Islam Khan
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0212, Japan
| | - Kimiko Yamamiya
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0212, Japan
| | - Takuya Doihara
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0212, Japan
| | - Naoto Kobayashi
- Medical Education Center, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, Japan
| | - Hiroyuki Wakisaka
- Department of Liberal Arts, Ehime Prefectural University of Health Sciences, 543 Takaoda, Tobe, Ehime, 791-2101, Japan
| | - Seiji Matsuda
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0212, Japan
| |
Collapse
|
33
|
Schartz ND, Tenner AJ. The good, the bad, and the opportunities of the complement system in neurodegenerative disease. J Neuroinflammation 2020; 17:354. [PMID: 33239010 PMCID: PMC7690210 DOI: 10.1186/s12974-020-02024-8] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
The complement cascade is a critical effector mechanism of the innate immune system that contributes to the rapid clearance of pathogens and dead or dying cells, as well as contributing to the extent and limit of the inflammatory immune response. In addition, some of the early components of this cascade have been clearly shown to play a beneficial role in synapse elimination during the development of the nervous system, although excessive complement-mediated synaptic pruning in the adult or injured brain may be detrimental in multiple neurogenerative disorders. While many of these later studies have been in mouse models, observations consistent with this notion have been reported in human postmortem examination of brain tissue. Increasing awareness of distinct roles of C1q, the initial recognition component of the classical complement pathway, that are independent of the rest of the complement cascade, as well as the relationship with other signaling pathways of inflammation (in the periphery as well as the central nervous system), highlights the need for a thorough understanding of these molecular entities and pathways to facilitate successful therapeutic design, including target identification, disease stage for treatment, and delivery in specific neurologic disorders. Here, we review the evidence for both beneficial and detrimental effects of complement components and activation products in multiple neurodegenerative disorders. Evidence for requisite co-factors for the diverse consequences are reviewed, as well as the recent studies that support the possibility of successful pharmacological approaches to suppress excessive and detrimental complement-mediated chronic inflammation, while preserving beneficial effects of complement components, to slow the progression of neurodegenerative disease.
Collapse
Affiliation(s)
- Nicole D. Schartz
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
| | - Andrea J. Tenner
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
- Department of Neurobiology and Behavior, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
| |
Collapse
|
34
|
Uddin MS, Kabir MT, Rahman MS, Behl T, Jeandet P, Ashraf GM, Najda A, Bin-Jumah MN, El-Seedi HR, Abdel-Daim MM. Revisiting the Amyloid Cascade Hypothesis: From Anti-Aβ Therapeutics to Auspicious New Ways for Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21165858. [PMID: 32824102 PMCID: PMC7461598 DOI: 10.3390/ijms21165858] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is the most prevalent neurodegenerative disorder related to age, characterized by the cerebral deposition of fibrils, which are made from the amyloid-β (Aβ), a peptide of 40–42 amino acids. The conversion of Aβ into neurotoxic oligomeric, fibrillar, and protofibrillar assemblies is supposed to be the main pathological event in AD. After Aβ accumulation, the clinical symptoms fall out predominantly due to the deficient brain clearance of the peptide. For several years, researchers have attempted to decline the Aβ monomer, oligomer, and aggregate levels, as well as plaques, employing agents that facilitate the reduction of Aβ and antagonize Aβ aggregation, or raise Aβ clearance from brain. Unluckily, broad clinical trials with mild to moderate AD participants have shown that these approaches were unsuccessful. Several clinical trials are running involving patients whose disease is at an early stage, but the preliminary outcomes are not clinically impressive. Many studies have been conducted against oligomers of Aβ which are the utmost neurotoxic molecular species. Trials with monoclonal antibodies directed against Aβ oligomers have exhibited exciting findings. Nevertheless, Aβ oligomers maintain equivalent states in both monomeric and aggregation forms; so, previously administered drugs that precisely decrease Aβ monomer or Aβ plaques ought to have displayed valuable clinical benefits. In this article, Aβ-based therapeutic strategies are discussed and several promising new ways to fight against AD are appraised.
Collapse
Affiliation(s)
- Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka 1213, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka 1207, Bangladesh
- Correspondence: ; Tel.: +880-171-022-0110
| | - Md. Tanvir Kabir
- Department of Pharmacy, BRAC University, Dhaka 1212, Bangladesh;
| | - Md. Sohanur Rahman
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi 6205, Bangladesh;
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687 Reims CEDEX 2, France;
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Agnieszka Najda
- Laboratory of Quality of Vegetables and Medicinal Plants, Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland;
| | - May N. Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia;
| | - Hesham R. El-Seedi
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China;
- Pharmacognosy Group, Department of Pharmaceutical Biosciences, Uppsala University, SE-751 23 Uppsala, Sweden
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Koom 32512, Egypt
| | - Mohamed M. Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
35
|
Kong Y, Liu C, Zhou Y, Qi J, Zhang C, Sun B, Wang J, Guan Y. Progress of RAGE Molecular Imaging in Alzheimer's Disease. Front Aging Neurosci 2020; 12:227. [PMID: 32848706 PMCID: PMC7417350 DOI: 10.3389/fnagi.2020.00227] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/29/2020] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease characterized by senile plaques (SPs), which are caused by amyloid beta (Aβ) deposition and neurofibrillary tangles (NFTs) of abnormal hyperphosphorylated tau protein. The receptor for advanced glycation end products (RAGE) binds to advanced glycation end products deposited during vascular dysfunction. Alzheimer’s disease may occur when RAGE binds to Aβ and releases reactive oxygen species, further exacerbating Aβ deposition and eventually leading to SPs and NFTs. As it is involved in early AD, RAGE may be considered as a more potent biomarker than Aβ. Positron emission tomography provides valuable information regarding the underlying pathological processes of AD many years before the appearance of clinical symptoms. Thus, to further reveal the role of RAGE in AD pathology and for early diagnosis of AD, a tracer that targets RAGE is needed. In this review, we first describe the early diagnosis of AD and then summarize the interaction between RAGE and Aβ and Tau that is required to induce AD pathology, and finally focus on RAGE-targeting probes, highlighting the potential of RAGE to be used as an effective target. The development of RAGE probes is expected to aid in AD diagnosis and treatment.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Cuiping Liu
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yinping Zhou
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Jingxuan Qi
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Chencheng Zhang
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bomin Sun
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
36
|
Kong Y, Wang F, Wang J, Liu C, Zhou Y, Xu Z, Zhang C, Sun B, Guan Y. Pathological Mechanisms Linking Diabetes Mellitus and Alzheimer's Disease: the Receptor for Advanced Glycation End Products (RAGE). Front Aging Neurosci 2020; 12:217. [PMID: 32774301 PMCID: PMC7388912 DOI: 10.3389/fnagi.2020.00217] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/19/2020] [Indexed: 12/13/2022] Open
Abstract
Diabetes and Alzheimer’s disease (AD) place a significant burden on health care systems in the world and its aging populations. These diseases have long been regarded as separate entities; however, advanced glycation end products (AGEs) and the receptors for AGEs (RAGE) may be a link between diabetes and AD. In our study, mice injected with AGEs through stereotaxic surgery showed significant AD-like features: behavior showed decreased memory; immunofluorescence showed increased phosphorylated tau and APP. These results suggest links between diabetes and AD. Patients with diabetes are at a higher risk of developing AD, and the possible underlying molecular components of this association are now beginning to emerge.
Collapse
Affiliation(s)
- Yanyan Kong
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Fushuai Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Cuiping Liu
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yinping Zhou
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Zhengqin Xu
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Chencheng Zhang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bomin Sun
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Kumthekar P, Tang SC, Brenner AJ, Kesari S, Piccioni DE, Anders C, Carrillo J, Chalasani P, Kabos P, Puhalla S, Tkaczuk K, Garcia AA, Ahluwalia MS, Wefel JS, Lakhani N, Ibrahim N. ANG1005, a Brain-Penetrating Peptide-Drug Conjugate, Shows Activity in Patients with Breast Cancer with Leptomeningeal Carcinomatosis and Recurrent Brain Metastases. Clin Cancer Res 2020; 26:2789-2799. [PMID: 31969331 DOI: 10.1158/1078-0432.ccr-19-3258] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/12/2019] [Accepted: 01/17/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE ANG1005, a novel taxane derivative, consists of three paclitaxel molecules covalently linked to Angiopep-2, designed to cross the blood-brain and blood-cerebrospinal barriers and to penetrate malignant cells via LRP1 transport system. Preclinical and clinical evidence of efficacy with ANG1005 has been previously shown. PATIENTS AND METHODS A multicenter, open-label phase II study in adult patients with measurable recurrent brain metastases from breast cancer (BCBM), with or without leptomeningeal carcinomatosis was conducted (n = 72 BCBM; n = 28 leptomeningeal carcinomatosis subset). ANG1005 was administered intravenously at 600 mg/m2 every 3 weeks. Tumor assessment was based on central nervous system (CNS) RECIST 1.1 for intracranial, and RECIST 1.1 for extracranial response. The primary endpoint was determination of intracranial objective response rate (iORR). RESULTS Median age was 47.5 years. Safety profile was similar to that of paclitaxel with myelosuppression as the predominating toxicity. Average number of prior CNS-directed therapies was 2.8 and 94% of the patients had prior taxane treatment. Patient benefit (stable disease or better) was seen in 77% (intracranial) and 86% (extracranial) of the evaluable patients, with iORR of 15% (investigator) or 8% (independent radiology facility [IRF] review). In the leptomeningeal carcinomatosis subset, 79% of the patients had intracranial disease control and estimated median overall survival of 8.0 months (95% CI, 5.4-9.4). CONCLUSIONS Even though the study preset rule for iORR per IRF was not met in this heavily pretreated population, a notable CNS and systemic treatment effect was seen in all patients including symptom improvement and prolonged overall survival compared to historical control for the subset of patients with leptomeningeal carcinomatosis (n = 28).
Collapse
Affiliation(s)
- Priya Kumthekar
- Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - Shou-Ching Tang
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, Mississippi
| | | | - Santosh Kesari
- John Wayne Cancer Institute and Pacific Neuroscience Institute, Santa Monica, California
| | - David E Piccioni
- Department of Neurosciences, UC San Diego Moores Cancer Center, La Jolla, California
| | | | - Jose Carrillo
- John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, California
| | | | - Peter Kabos
- University of Colorado, Anschutz Medical Campus, Greenwood Village, Colorado
| | - Shannon Puhalla
- University of Pittsburgh Magee Women's Cancer Program, Pittsburgh, Pennsylvania
| | - Katherine Tkaczuk
- University Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | | | - Manmeet S Ahluwalia
- Miller Family Endowed Chair in NeuroOncology; Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio
| | - Jeffrey S Wefel
- Departments of Neuro-Oncology and Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nehal Lakhani
- Cancer and Hematology Centers of Western Michigan, Grand Rapids, Michigan
| | - Nuhad Ibrahim
- Department of Breast Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center; Houston, Texas
| |
Collapse
|
38
|
Banks WA. The Blood-Brain Barrier Interface in Diabetes Mellitus: Dysfunctions, Mechanisms and Approaches to Treatment. Curr Pharm Des 2020; 26:1438-1447. [DOI: 10.2174/1381612826666200325110014] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/27/2020] [Indexed: 12/24/2022]
Abstract
Diabetes mellitus (DM) is one of the most common diseases in the world. Among its effects are an increase in the risk of cognitive impairment, including Alzheimer’s disease, and blood-brain barrier (BBB) dysfunction. DM is characterized by high blood glucose levels that are caused by either lack of insulin (Type I) or resistance to the actions of insulin (Type II). The phenotypes of these two types are dramatically different, with Type I animals being thin, with low levels of leptin as well as insulin, whereas Type II animals are often obese with high levels of both leptin and insulin. The best characterized change in BBB dysfunction is that of disruption. The brain regions that are disrupted, however, vary between Type I vs Type II DM, suggesting that factors other than hyperglycemia, perhaps hormonal factors such as leptin and insulin, play a regionally diverse role in BBB vulnerability or protection. Some BBB transporters are also altered in DM, including P-glycoprotein, lowdensity lipoprotein receptor-related protein 1, and the insulin transporter as other functions of the BBB, such as brain endothelial cell (BEC) expression of matrix metalloproteinases (MMPs) and immune cell trafficking. Pericyte loss secondary to the increased oxidative stress of processing excess glucose through the Krebs cycle is one mechanism that has shown to result in BBB disruption. Vascular endothelial growth factor (VEGF) induced by advanced glycation endproducts can increase the production of matrix metalloproteinases, which in turn affects tight junction proteins, providing another mechanism for BBB disruption as well as effects on P-glycoprotein. Through the enhanced expression of the redox-related mitochondrial transporter ABCB10, redox-sensitive transcription factor NF-E2 related factor-2 (Nrf2) inhibits BEC-monocyte adhesion. Several potential therapies, in addition to those of restoring euglycemia, can prevent some aspects of BBB dysfunction. Carbonic anhydrase inhibition decreases glucose metabolism and so reduces oxidative stress, preserving pericytes and blocking or reversing BBB disruption. Statins or N-acetylcysteine can reverse the BBB opening in some models of DM, fibroblast growth factor-21 improves BBB permeability through an Nrf2-dependent pathway, and nifedipine or VEGF improves memory in DM models. In summary, DM alters various aspects of BBB function through a number of mechanisms. A variety of treatments based on those mechanisms, as well as restoration of euglycemia, may be able to restore BBB functions., including reversal of BBB disruption.
Collapse
Affiliation(s)
- William A. Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, United States
| |
Collapse
|
39
|
Bhalerao A, Sivandzade F, Archie SR, Chowdhury EA, Noorani B, Cucullo L. In vitro modeling of the neurovascular unit: advances in the field. Fluids Barriers CNS 2020; 17:22. [PMID: 32178700 PMCID: PMC7077137 DOI: 10.1186/s12987-020-00183-7] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
The blood–brain barrier (BBB) is a fundamental component of the central nervous system. Its functional and structural integrity is vital in maintaining the homeostasis of the brain microenvironment. On the other hand, the BBB is also a major hindering obstacle for the delivery of effective therapies to treat disorders of the Central Nervous System (CNS). Over time, various model systems have been established to simulate the complexities of the BBB. The development of realistic in vitro BBB models that accurately mimic the physiological characteristics of the brain microcapillaries in situ is of fundamental importance not only in CNS drug discovery but also in translational research. Successful modeling of the Neurovascular Unit (NVU) would provide an invaluable tool that would aid in dissecting out the pathological factors, mechanisms of action, and corresponding targets prodromal to the onset of CNS disorders. The field of BBB in vitro modeling has seen many fundamental changes in the last few years with the introduction of novel tools and methods to improve existing models and enable new ones. The development of CNS organoids, organ-on-chip, spheroids, 3D printed microfluidics, and other innovative technologies have the potential to advance the field of BBB and NVU modeling. Therefore, in this review, summarize the advances and progress in the design and application of functional in vitro BBB platforms with a focus on rapidly advancing technologies.
Collapse
Affiliation(s)
- Aditya Bhalerao
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Farzane Sivandzade
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Sabrina Rahman Archie
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Ekram Ahmed Chowdhury
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Behnam Noorani
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Luca Cucullo
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA. .,Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA.
| |
Collapse
|
40
|
Therapeutic Potential of Direct Clearance of the Amyloid-β in Alzheimer's Disease. Brain Sci 2020; 10:brainsci10020093. [PMID: 32050618 PMCID: PMC7071829 DOI: 10.3390/brainsci10020093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/05/2020] [Accepted: 02/05/2020] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by deposition and accumulation of amyloid-β (Aβ) and its corresponding plaques within the brain. Although much debate exists whether these plaques are the cause or the effect of AD, the accumulation of Aβ is linked with the imbalance between the production and clearance of Aβ. The receptor for advanced glycation endproducts (RAGE) facilitates entry of free Aβ from the peripheral stream. Conversely, lipoprotein receptor-related protein 1 (LRP1), located in the abluminal side at the blood–brain barrier mediates the efflux of Aβ. Research on altering the rates of clearance of Aβ by targeting these two pathways has been extensively study. Additionally, a cerebrospinal fluid (CSF) circulation assistant device has also been evaluated as an approach to increase solute concentration in the CSF via mechanical drainage, to allow for removal of Aβ from the brain. Herein, we provide a brief review of these approaches that are designed to re-establish a homeostatic Aβ balance in the brain.
Collapse
|
41
|
Versele R, Corsi M, Fuso A, Sevin E, Businaro R, Gosselet F, Fenart L, Candela P. Ketone Bodies Promote Amyloid-β 1-40 Clearance in a Human in Vitro Blood-Brain Barrier Model. Int J Mol Sci 2020; 21:E934. [PMID: 32023814 PMCID: PMC7037612 DOI: 10.3390/ijms21030934] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the abnormal accumulation of amyloid-β (Aβ) peptides in the brain. The pathological process has not yet been clarified, although dysfunctional transport of Aβ across the blood-brain barrier (BBB) appears to be integral to disease development. At present, no effective therapeutic treatment against AD exists, and the adoption of a ketogenic diet (KD) or ketone body (KB) supplements have been investigated as potential new therapeutic approaches. Despite experimental evidence supporting the hypothesis that KBs reduce the Aβ load in the AD brain, little information is available about the effect of KBs on BBB and their effect on Aβ transport. Therefore, we used a human in vitro BBB model, brain-like endothelial cells (BLECs), to investigate the effect of KBs on the BBB and on Aβ transport. Our results show that KBs do not modify BBB integrity and do not cause toxicity to BLECs. Furthermore, the presence of KBs in the culture media was combined with higher MCT1 and GLUT1 protein levels in BLECs. In addition, KBs significantly enhanced the protein levels of LRP1, P-gp, and PICALM, described to be involved in Aβ clearance. Finally, the combined use of KBs promotes Aβ efflux across the BBB. Inhibition experiments demonstrated the involvement of LRP1 and P-gp in the efflux. This work provides evidence that KBs promote Aβ clearance from the brain to blood in addition to exciting perspectives for studying the use of KBs in therapeutic approaches.
Collapse
Affiliation(s)
- Romain Versele
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| | - Mariangela Corsi
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy;
| | - Andrea Fuso
- Department of Experimental Medicine, Sapienza University of Rome, Dip. di Chirurgia “P. Valdoni”, Via A. Scarpa 16, 00161 Rome, Italy;
| | - Emmanuel Sevin
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| | - Rita Businaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy;
| | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| | - Laurence Fenart
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| | - Pietra Candela
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d'Artois, F-62300 Lens, France; (R.V.); (M.C.); (E.S.); (F.G.); (L.F.)
| |
Collapse
|
42
|
Abstract
The blood-brain barrier (BBB) was first noted for its ability to prevent the unregulated exchange of substances between the blood and the central nervous system (CNS). Over time, its characterization as an interface that enables regulated exchanges between the CNS and substances that are carried in the blood in a hormone-like fashion have emerged. Therefore, communication between the CNS, BBB and peripheral tissues has many endocrine-like properties. In this Review, I examine the various ways in which the BBB exhibits endocrine-related properties. The BBB is a target for hormones, such as leptin and insulin, that affect many of its functions. The BBB is also a secretory body, releasing substances either into the blood or the interstitial fluid of the brain. The BBB selectively allows classical and non-classical hormones entry to and exit from the CNS, thus allowing the CNS to be both an endocrine target and a secretory tissue. The BBB is affected by endocrine diseases such as diabetes mellitus and can cause or participate in endocrine diseases, including those related to thyroid hormones and obesity. The endocrine-like mechanisms of the BBB can extend the definition of endocrine disease to include neurodegenerative conditions, including Alzheimer disease, and of hormones to include cytokines, triglycerides and fatty acids.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
43
|
Wang J, Qiao F, Shang S, Li P, Chen C, Dang L, Jiang Y, Huo K, Deng M, Wang J, Qu Q. Elevation of Plasma Amyloid-β Level is More Significant in Early Stage of Cognitive Impairment: A Population-Based Cross-Sectional Study. J Alzheimers Dis 2019; 64:61-69. [PMID: 29865072 DOI: 10.3233/jad-180140] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Aggregation and deposition of amyloid-β (Aβ) in the brain is the main pathological change of Alzheimer's disease (AD). Decreased Aβ42 in the cerebrospinal fluid has been confirmed as a biomarker of AD; however, the relationship between plasma Aβ and cognitive impairment is currently unclear. OBJECTIVE The aim was to explore the relationship between plasma Aβ and cognitive impairment in a cross-sectional study. METHODS A total of 1,314 subjects (age above 40) from a village in the suburbs of Xi'an, China were enrolled between October 8, 2014 and March 30, 2015. A validated Chinese version of the Mini-Mental State Examination and neuropsychological battery were used to assess cognition. Levels of plasma Aβ42 and Aβ40 were tested using commercial enzyme-linked immunosorbent assay. Relationship of plasma Aβ and cognitive impairment was analyzed using logistic regression analysis. RESULTS Of the enrolled subjects, 1,180 (89.80%) had normal cognition, 85 (6.47%) had possible cognitive impairment and 49 (3.73%) had probable cognitive impairment. Logistic regression analysis showed that the Aβ42/Aβ40 ratio (OR = 4.042, 95% CI: 1.248-11.098, p = 0.012) and plasma Aβ42 (OR = 1.036, 95% CI: 1.003-1.071, p = 0.031) was higher in the possible cognitive impairment than that in the normal cognition group. Furthermore, the plasma Aβ42/Aβ40 ratio was higher in the possible cognitive impairment group than that in the probable cognitive impairment group (OR = 0.029, 95% CI: 0.002-0.450, p = 0.011). CONCLUSIONS Levels of plasma Aβ42 and Aβ42/Aβ40 ratio were elevated in patients with possible cognitive impairment, indicating that plasma Aβ42 and Aβ42/Aβ40 ratio increases may be more pronounced in early stage of cognitive impairment.
Collapse
Affiliation(s)
- Jin Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fan Qiao
- Weinan Central hospital, Shaanxi, China
| | - Suhang Shang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Pei Li
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chen Chen
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liangjun Dang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yu Jiang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kang Huo
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meiying Deng
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingyi Wang
- Huxian Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
44
|
Kehoe PG. The Coming of Age of the Angiotensin Hypothesis in Alzheimer's Disease: Progress Toward Disease Prevention and Treatment? J Alzheimers Dis 2019; 62:1443-1466. [PMID: 29562545 PMCID: PMC5870007 DOI: 10.3233/jad-171119] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There is wide recognition of a complex association between midlife hypertension and cardiovascular disease and later development of Alzheimer’s disease (AD) and cognitive impairment. While significant progress has been made in reducing rates of mortality and morbidity due to cardiovascular disease over the last thirty years, progress towards effective treatments for AD has been slower. Despite the known association between hypertension and dementia, research into each disease has largely been undertaken in parallel and independently. Yet over the last decade and a half, the emergence of converging findings from pre-clinical and clinical research has shown how the renin angiotensin system (RAS), which is very important in blood pressure regulation and cardiovascular disease, warrants careful consideration in the pathogenesis of AD. Numerous components of the RAS have now been found to be altered in AD such that the multifunctional and potent vasoconstrictor angiotensin II, and similarly acting angiotensin III, are greatly altered at the expense of other RAS signaling peptides considered to contribute to neuronal and cognitive function. Collectively these changes may contribute to many of the neuropathological hallmarks of AD, as well as observed progressive deficiencies in cognitive function, while also linking elements of a number of the proposed hypotheses for the cause of AD. This review discusses the emergence of the RAS and its likely importance in AD, not only because of the multiple facets of its involvement, but also perhaps fortuitously because of the ready availability of numerous RAS-acting drugs, that could be repurposed as interventions in AD.
Collapse
Affiliation(s)
- Patrick Gavin Kehoe
- Dementia Research Group, Translational Health Sciences, Bristol Medical School, University of Bristol, Southmead Hospital, Bristol, UK
| |
Collapse
|
45
|
Liebsch F, Kulic L, Teunissen C, Shobo A, Ulku I, Engelschalt V, Hancock MA, van der Flier WM, Kunach P, Rosa-Neto P, Scheltens P, Poirier J, Saftig P, Bateman RJ, Breitner J, Hock C, Multhaup G. Aβ34 is a BACE1-derived degradation intermediate associated with amyloid clearance and Alzheimer's disease progression. Nat Commun 2019; 10:2240. [PMID: 31110178 PMCID: PMC6527709 DOI: 10.1038/s41467-019-10152-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 04/17/2019] [Indexed: 12/13/2022] Open
Abstract
The beta-site APP cleaving enzyme 1 (BACE1) is known primarily for its initial cleavage of the amyloid precursor protein (APP), which ultimately leads to the generation of Aβ peptides. Here, we provide evidence that altered BACE1 levels and activity impact the degradation of Aβ40 and Aβ42 into a common Aβ34 intermediate. Using human cerebrospinal fluid (CSF) samples from the Amsterdam Dementia Cohort, we show that Aβ34 is elevated in individuals with mild cognitive impairment who later progressed to dementia. Furthermore, Aβ34 levels correlate with the overall Aβ clearance rates in amyloid positive individuals. Using CSF samples from the PREVENT-AD cohort (cognitively normal individuals at risk for Alzheimer’s disease), we further demonstrate that the Aβ34/Aβ42 ratio, representing Aβ degradation and cortical deposition, associates with pre-clinical markers of neurodegeneration. We propose that Aβ34 represents a marker of amyloid clearance and may be helpful for the characterization of Aβ turnover in clinical samples. Aβ34 is generated from degradation of Aβ40 and Aβ42 by β-secretase. Here, the authors show that Aβ34 is a marker for amyloid clearance and is elevated in the CSF of patients that go on to convert from mild cognitive impairment to Alzheimer’s disease, suggesting it may be a useful biomarker.
Collapse
Affiliation(s)
- Filip Liebsch
- Department of Pharmacology and Therapeutics and Integrated Program in Neuroscience, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Luka Kulic
- Institute for Regenerative Medicine, University of Zurich, CH-8952, Schlieren, Switzerland
| | - Charlotte Teunissen
- Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081HZ, Amsterdam, The Netherlands
| | - Adeola Shobo
- Department of Pharmacology and Therapeutics and Integrated Program in Neuroscience, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Irem Ulku
- Department of Pharmacology and Therapeutics and Integrated Program in Neuroscience, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Vivienne Engelschalt
- Institut für Chemie und Biochemie, Freie Universität Berlin, 14195, Berlin, Germany
| | - Mark A Hancock
- SPR-MS Facility, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, 1081HZ, The Netherlands
| | - Peter Kunach
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, McGill University, Montreal, H4H 1R3, QC, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, McGill University, Montreal, H4H 1R3, QC, Canada
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, 1081HZ, The Netherlands
| | - Judes Poirier
- Department of Psychiatry, McGill University, Montreal, QC, H4H 1R3, Canada
| | - Paul Saftig
- Biochemisches Institut, Christian-Albrechts-Universität-Kiel, 24118, Kiel, Germany
| | - Randall J Bateman
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - John Breitner
- Department of Psychiatry, McGill University, Montreal, QC, H4H 1R3, Canada
| | - Christoph Hock
- Institute for Regenerative Medicine, University of Zurich, CH-8952, Schlieren, Switzerland.,Neurimmune, CH-8952, Schlieren, Switzerland
| | - Gerhard Multhaup
- Department of Pharmacology and Therapeutics and Integrated Program in Neuroscience, McGill University, Montreal, QC, H3G 1Y6, Canada.
| |
Collapse
|
46
|
Aldea R, Weller RO, Wilcock DM, Carare RO, Richardson G. Cerebrovascular Smooth Muscle Cells as the Drivers of Intramural Periarterial Drainage of the Brain. Front Aging Neurosci 2019; 11:1. [PMID: 30740048 PMCID: PMC6357927 DOI: 10.3389/fnagi.2019.00001] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/07/2019] [Indexed: 12/25/2022] Open
Abstract
The human brain is the organ with the highest metabolic activity but it lacks a traditional lymphatic system responsible for clearing waste products. We have demonstrated that the basement membranes of cerebral capillaries and arteries represent the lymphatic pathways of the brain along which intramural periarterial drainage (IPAD) of soluble metabolites occurs. Failure of IPAD could explain the vascular deposition of the amyloid-beta protein as cerebral amyloid angiopathy (CAA), which is a key pathological feature of Alzheimer's disease. The underlying mechanisms of IPAD, including its motive force, have not been clarified, delaying successful therapies for CAA. Although arterial pulsations from the heart were initially considered to be the motive force for IPAD, they are not strong enough for efficient IPAD. This study aims to unravel the driving force for IPAD, by shifting the perspective of a heart-driven clearance of soluble metabolites from the brain to an intrinsic mechanism of cerebral arteries (e.g., vasomotion-driven IPAD). We test the hypothesis that the cerebrovascular smooth muscle cells, whose cycles of contraction and relaxation generate vasomotion, are the drivers of IPAD. A novel multiscale model of arteries, in which we treat the basement membrane as a fluid-filled poroelastic medium deformed by the contractile cerebrovascular smooth muscle cells, is used to test the hypothesis. The vasomotion-induced intramural flow rates suggest that vasomotion-driven IPAD is the only mechanism postulated to date capable of explaining the available experimental observations. The cerebrovascular smooth muscle cells could represent valuable drug targets for prevention and early interventions in CAA.
Collapse
Affiliation(s)
- Roxana Aldea
- Mathematical Sciences, University of Southampton, Southampton, United Kingdom
| | - Roy O Weller
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Donna M Wilcock
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Roxana O Carare
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Giles Richardson
- Mathematical Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
47
|
Sharma R, Razdan K, Bansal Y, Kuhad A. Rollercoaster ride of kynurenines: steering the wheel towards neuroprotection in Alzheimer's disease. Expert Opin Ther Targets 2018; 22:849-867. [PMID: 30223691 DOI: 10.1080/14728222.2018.1524877] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is associated with cerebral cognitive deficits exhibiting two cardinal hallmarks: accruement of extracellular amyloid plaques and intracellular neurofibrillary tangles composed of hyperphosphorylated tau protein. The currently accessible therapeutic armamentarium merely provides symptomatic relief. Therefore, the cry for prospective neuroprotective strategies seems to be the need of the hour. Areas covered: This review comprehensively establishes correlation between kynurenine pathway (KP) metabolites and AD with major emphasis on its two functionally contrasting neuroactive metabolites i.e. kynurenic acid (KYNA) and quinolinic acid (QUIN) and enlists various clinical studies which hold a potential for future therapeutics in AD. Also, major hypotheses of AD and mechanisms underlying them have been scrutinized with the aim to brush up the readers with basic pathology of AD. Expert opinion: KP is unique in itself as it holds two completely different domains i.e. neurotoxic QUIN and neuroprotective KYNA and disrupted equilibrium between the two has a hand in neurodegeneration. KYNA has long been demonstrated to be neuroprotective but lately being disparaged for cognitive side effects. But we blaze a trail by amalgamating the pharmacological mechanistic studies of KYNA in kinship with α7nAChRs, NMDARs and GABA which lends aid in favour of KA.
Collapse
Affiliation(s)
- Radhika Sharma
- a Pharmacology Research Laboratory , University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University , Chandigarh , India
| | - Karan Razdan
- b Pharmaceutics division , University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University , Chandigarh , India
| | - Yashika Bansal
- a Pharmacology Research Laboratory , University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University , Chandigarh , India
| | - Anurag Kuhad
- a Pharmacology Research Laboratory , University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University , Chandigarh , India
| |
Collapse
|
48
|
Ryan P, Patel B, Makwana V, Jadhav HR, Kiefel M, Davey A, Reekie TA, Rudrawar S, Kassiou M. Peptides, Peptidomimetics, and Carbohydrate-Peptide Conjugates as Amyloidogenic Aggregation Inhibitors for Alzheimer's Disease. ACS Chem Neurosci 2018; 9:1530-1551. [PMID: 29782794 DOI: 10.1021/acschemneuro.8b00185] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder accounting for 60-80% of dementia cases. For many years, AD causality was attributed to amyloid-β (Aβ) aggregated species. Recently, multiple therapies that target Aβ aggregation have failed in clinical trials, since Aβ aggregation is found in AD and healthy patients. Attention has therefore shifted toward the aggregation of the tau protein as a major driver of AD. Numerous inhibitors of tau-based pathology have recently been developed. Diagnosis of AD has shifted from measuring late stage senile plaques to early stage biomarkers, amyloid-β and tau monomers and oligomeric assemblies. Synthetic peptides and some derivative structures are being explored for use as theranostic tools as they possess the capacity both to bind the biomarkers and to inhibit their pathological self-assembly. Several studies have demonstrated that O-linked glycoside addition can significantly alter amyloid aggregation kinetics. Furthermore, natural O-glycosylation of amyloid-forming proteins, including amyloid precursor protein (APP), tau, and α-synuclein, promotes alternative nonamyloidogenic processing pathways. As such, glycopeptides and related peptidomimetics are being investigated within the AD field. Here we review advancements made in the last 5 years, as well as the arrival of sugar-based derivatives.
Collapse
Affiliation(s)
- Philip Ryan
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast 4222, Australia
| | - Bhautikkumar Patel
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast 4222, Australia
| | - Vivek Makwana
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast 4222, Australia
| | - Hemant R. Jadhav
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani-333031, Rajasthan, India
| | - Milton Kiefel
- Institute for Glycomics, Griffith University, Gold Coast 4222, Australia
| | - Andrew Davey
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast 4222, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast 4222, Australia
- Quality Use of Medicines Network, Griffith University, Gold Coast 4222, Australia
| | | | - Santosh Rudrawar
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast 4222, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast 4222, Australia
- Quality Use of Medicines Network, Griffith University, Gold Coast 4222, Australia
- School of Chemistry, The University of Sydney, NSW 2006, Australia
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, NSW 2006, Australia
| |
Collapse
|
49
|
Fan LY, Tzen KY, Chen YF, Chen TF, Lai YM, Yen RF, Huang YY, Shiue CY, Yang SY, Chiu MJ. The Relation Between Brain Amyloid Deposition, Cortical Atrophy, and Plasma Biomarkers in Amnesic Mild Cognitive Impairment and Alzheimer's Disease. Front Aging Neurosci 2018; 10:175. [PMID: 29967578 PMCID: PMC6015901 DOI: 10.3389/fnagi.2018.00175] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/22/2018] [Indexed: 01/25/2023] Open
Abstract
Background: Neuritic plaques and neurofibrillary tangles are the pathological hallmarks of Alzheimer’s disease (AD), while the role of brain amyloid deposition in the clinical manifestation or brain atrophy remains unresolved. We aimed to explore the relation between brain amyloid deposition, cortical thickness, and plasma biomarkers. Methods: We used 11C-Pittsburgh compound B-positron emission tomography to assay brain amyloid deposition, magnetic resonance imaging to estimate cortical thickness, and an immunomagnetic reduction assay to measure plasma biomarkers. We recruited 39 controls, 25 subjects with amnesic mild cognitive impairment (aMCI), and 16 subjects with AD. PiB positivity (PiB+) was defined by the upper limit of the 95% confidence interval of the mean cortical SUVR from six predefined regions (1.0511 in this study). Results: All plasma biomarkers showed significant between-group differences. The plasma Aβ40 level was positively correlated with the mean cortical thickness of both the PiB+ and PiB- subjects. The plasma Aβ40 level of the subjects who were PiB+ was negatively correlated with brain amyloid deposition. In addition, the plasma tau level was negatively correlated with cortical thickness in both the PiB+ and PiB- subjects. Moreover, cortical thickness was negatively correlated with brain amyloid deposition in the PiB+ subjects. In addition, the cut-off point of plasma tau for differentiating between controls and AD was higher in the PiB- group than in the PiB+ group (37.5 versus 25.6 pg/ml, respectively). Lastly, ApoE4 increased the PiB+ rate in the aMCI and control groups. Conclusion: The contributions of brain amyloid deposition to cortical atrophy are spatially distinct. Plasma Aβ40 might be a protective indicator of less brain amyloid deposition and cortical atrophy. It takes more tau pathology to reach the same level of cognitive decline in subjects without brain amyloid deposition, and ApoE4 plays an early role in amyloid pathogenesis.
Collapse
Affiliation(s)
- Ling-Yun Fan
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.,Institute of Brain and Mind Sciences, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kai-Yuan Tzen
- PET Center, Department of Nuclear Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Nuclear Medicine, Changhua Christian Hospital, Changhua City, Taiwan.,Molecular Imaging Center, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Fang Chen
- Department of Medical Imaging, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Mei Lai
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ruoh-Fang Yen
- PET Center, Department of Nuclear Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.,Molecular Imaging Center, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Yao Huang
- Molecular Imaging Center, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chyng-Yann Shiue
- PET Center, Department of Nuclear Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.,Molecular Imaging Center, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.,PET Center, Tri-Service General Hospital, Taipei, Taiwan
| | | | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.,Institute of Brain and Mind Sciences, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Psychology, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
50
|
Zimering MB. Circulating Neurotoxic 5-HT2A Receptor Agonist Autoantibodies in Adult Type 2 Diabetes with Parkinson's Disease. JOURNAL OF ENDOCRINOLOGY AND DIABETES 2018; 5:10.15226/2374-6890/5/2/01102. [PMID: 29888323 PMCID: PMC5990037 DOI: 10.15226/2374-6890/5/2/01102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
AIMS To test whether circulating neurotoxic autoantibodies increase in adult type 2 diabetes mellitus with Parkinson's disease (PD) or dementia. To identify the G-protein coupled receptor on neuroblastoma cells mediating neural inhibitory effects in diabetic Parkinson's disease plasma autoantibodies. To determine the mechanism of accelerated neuroblastoma cell death and acute neurite retraction induced by diabetic Parkinson's disease and dementia autoantibodies. METHODS Protein-A eluates from plasma of twelve older adult male diabetic patients having Parkinson's disease (n=10) or dementia (n=2), and eight age-matched control diabetic patients were tested for ability to cause accelerated N2A neuroblastoma cell death and acute neurite retraction. Specific antagonists of G protein coupled receptors belonging to the G alpha q subfamily of heterotrimetric G-proteins, the phospholipase C/inositol triphosphate/Ca2+ pathway, or the RhoA/Rho kinase pathway were tested for ability to block diabetic Parkinson's disease/dementia autoantibody-induced neurite retraction or N2A accelerated cell loss. Sequential Liposorber LA-15 dextran sulfate cellulose/protein-A affinity chromatography was used to obtain highly-purified fractions of diabetic Parkinson's disease autoantibodies. RESULTS Mean accelerated neuroblastoma cell loss induced by diabetic Parkinson's disease or dementia autoantibodies significantly exceeded (P = 0.001) the level of N2A cell loss induced by an identical concentration of the diabetic autoantibodies in control patients without these two co-morbid neurodegenerative disorders. Co-incubation of diabetic Parkinson's disease and dementia autoantibodies with two-hundred nanomolar concentrations of M100907, a highly selective 5-HT2AR antagonist, completely prevented autoantibody-induced accelerated N2A cell loss and neurite retraction. A higher concentration (500 nM-10μM) of alpha-1 adrenergic, angiotensin II type 1, or endothelin A receptor antagonists did not substantially inhibit autoantibody-induced neuroblastoma cell death or prevent neurite retraction. Antagonists of the inositol triphosphate receptor (2-APB, 50μM), the intracellular calcium chelator (BAPTA-AM, 30 μM) and Y27632 (10 μM), a selective RhoA/Rho kinase inhibitor, each completely blocked acute neurite retraction induced by sixty nanomolar concentrations of diabetic Parkinson's disease autoantibodies. Co-incubation with 2-APB (1-2 μM) for 8 hours' prevented autoantibody-induced N2A cell loss. The highly-purified fraction obtained after Liposorber LA/protein-A affinity chromatography in hypertriglyceridemic diabetic dementia and Parkinson's disease plasmas had apparent MWs > 30 kD, and displayed enhanced N2A toxicity requiring substantially higher concentrations of 5-HT2AR antagonists (M100907, ketanserin, spiperone) to effectively neutralize. CONCLUSION These data suggest increased autoantibodies in older adult diabetes with Parkinson's disease or dementia cause accelerated neuron loss via the 5-hydroxytryptamine 2 receptor coupled to inositol triphosphate receptor-mediated cytosolic Ca2+ release.
Collapse
Affiliation(s)
- Mark B. Zimering
- Endocrinology, Veterans Affairs New Jersey Healthcare System, East Orange, NJ & Rutgers/Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|