1
|
Arshavsky YI. Autoimmune hypothesis of Alzheimer's disease: unanswered question. J Neurophysiol 2024; 132:929-942. [PMID: 39163023 DOI: 10.1152/jn.00259.2024] [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: 06/18/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Alzheimer's disease (AD) was described more than a century ago. However, there are still no effective approaches to its treatment, which may suggest that the search for the cure is not being conducted in the most productive direction. AD begins as selective impairments of declarative memory with no deficits in other cognitive functions. Therefore, understanding of the AD pathogenesis has to include the understanding of this selectivity. Currently, the main efforts aimed at prevention and treatment of AD are based on the dominating hypothesis for the AD pathogenesis: the amyloid hypothesis. But this hypothesis does not explain selective memory impairments since β-amyloid accumulates extracellularly and should be toxic to all types of cerebral neurons, not only to "memory engram neurons." To explain selective memory impairment, I propose the autoimmune hypothesis of AD, based on the analysis of risk factors for AD and molecular mechanisms of memory formation. Memory formation is associated with epigenetic modifications of chromatin in memory engram neurons and, therefore, might be accompanied by the expression of memory-specific proteins recognized by the adaptive immune system as "non-self" antigens. Normally, the brain is protected by the blood-brain barrier (BBB). All risk factors for AD provoke BBB disruptions, possibly leading to an autoimmune reaction against memory engram neurons. This reaction would make them selectively sensitive to tauopathy. If this hypothesis is confirmed, the strategies for AD prevention and treatment would be radically changed.
Collapse
Affiliation(s)
- Yuri I Arshavsky
- BioCircuits Institute, University of California, San Diego, La Jolla, California, United States
| |
Collapse
|
2
|
Behl T, Kaur I, Sehgal A, Khandige PS, Imran M, Gulati M, Khalid Anwer M, Elossaily GM, Ali N, Wal P, Gasmi A. The link between Alzheimer's disease and stroke: A detrimental synergism. Ageing Res Rev 2024; 99:102388. [PMID: 38914265 DOI: 10.1016/j.arr.2024.102388] [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: 04/02/2024] [Revised: 05/05/2024] [Accepted: 05/06/2024] [Indexed: 06/26/2024]
Abstract
Being age-related disorders, both Alzheimer's disease (AD) and stroke share multiple risk factors, such as hypertension, smoking, diabetes, and apolipoprotein E (APOE) Ɛ4 genotype, and coexist in patients. Accumulation of amyloid-β plaques and neurofibrillary tangled impair cognitive potential, leading to AD. Blocked blood flow in the neuronal tissues, causes neurodegeneration and cell death in stroke. AD is commonly characterized by cerebral amyloid angiopathy, which significantly elevates the risk of hemorrhagic stroke. Patients with AD and stroke have been both reported to exhibit greater cognitive impairment, followed by multiple pathophysiological mechanisms shared between the two. The manuscript aims to elucidate the relationship between AD and stroke, as well as the common pathways and risk factors while understanding the preventive therapies that might limit the negative impacts of this correlation, with diagnostic modalities and current AD treatments. The authors provide a comprehensive review of the link and aid the healthcare professionals to identify suitable targets and risk factors, that may retard cognitive decline and neurodegeneration in patients. However, more intricate research is required in this regard and an interdisciplinary approach that would target both the vascular and neurodegenerative factors would improve the quality of life in AD patients.
Collapse
Affiliation(s)
- Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India.
| | - Ishnoor Kaur
- University of Glasgow, College of Medical, Veterinary and Life Sciences, Glasgow, United Kingdom
| | - Aayush Sehgal
- GHG Khalsa College of Pharmacy, Gurusar Sadhar, Ludhiana, Punjab, India
| | - Prasanna Shama Khandige
- NITTE (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmacology, Mangaluru, Karnataka, India
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Md Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Gehan M Elossaily
- Department of Baisc Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Pranay Wal
- PSIT Kanpur, Department of Pharmacy, Uttar Pradesh, India
| | - Amin Gasmi
- Societe Francophone de Nutritherapie et de Nutrigenetique Appliquee, Villeurbanne, France; International Institute of Nutrition and Micronutrition Sciences, Saint Etienne, France
| |
Collapse
|
3
|
Vasantharekha R, Priyanka HP, Nair RS, Hima L, Pratap UP, Srinivasan AV, ThyagaRajan S. Alterations in Immune Responses Are Associated with Dysfunctional Intracellular Signaling in Peripheral Blood Mononuclear Cells of Men and Women with Mild Cognitive Impairment and Alzheimer's disease. Mol Neurobiol 2024; 61:2964-2977. [PMID: 37957423 DOI: 10.1007/s12035-023-03764-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023]
Abstract
Deficits in the neuroendocrine-immune network in the periphery associated with the onset and progression of mild cognitive impairment (MCI) and Alzheimer's disease (AD) have not been extensively studied. The present study correlatively examines the association between cell-mediated immune responses, stress hormones, amyloid precursor protein (APP) expression, peripheral blood mononuclear cells (PBMC), and intracellular signaling molecules in the pathophysiology of MCI and AD compared to adults. Serum APP, lymphocyte proliferation, total cholinesterase (TChE), butyrylcholinesterase (BChE) activities, cytokines (IL-2, IFN-γ, IL-6, and TNF-α), and intracellular signaling molecules (p-ERK, p-CREB, and p-Akt) were measured in the PBMCs of adult, old, MCI, and AD men and women initially and after 3 years in the same population. An age- and disease-associated decline in mini-mental state examination (MMSE) scores and lymphocyte proliferation of MCI and AD men and women were observed. An age- and disease-related increase in serum APP, cortisol levels, and TChE activity were observed in men and women. Enhanced production of Th1 cytokine, IL-2, pro-inflammatory cytokines, and suppressed intracellular transcription factors may promote the inflammatory environment in MCI and AD patients. The expression of CREB and Akt was lower in MCI and AD men, while the expression of p-ERK was higher, and p-CREB was lower in MCI and AD women after 3 years. These results suggest that changes in specific intracellular signaling pathways may influence alterations in cell-mediated immunity to promote disease progression in MCI and AD patients.
Collapse
Affiliation(s)
- Ramasamy Vasantharekha
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science & Technology, Kattankulathur, 603203, Tamil Nadu, India.
| | - Hannah P Priyanka
- Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Omandurar Government Estate, Chennai, Tamil Nadu, India
| | - Rahul S Nair
- Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Omandurar Government Estate, Chennai, Tamil Nadu, India
| | - Lalgi Hima
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science & Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Uday P Pratap
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science & Technology, Kattankulathur, 603203, Tamil Nadu, India
| | | | - Srinivasan ThyagaRajan
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science & Technology, Kattankulathur, 603203, Tamil Nadu, India
| |
Collapse
|
4
|
Acharya NK, Grossman HC, Clifford PM, Levin EC, Light KR, Choi H, Swanson II RL, Kosciuk MC, Venkataraman V, Libon DJ, Matzel LD, Nagele RG. A Chronic Increase in Blood-Brain Barrier Permeability Facilitates Intraneuronal Deposition of Exogenous Bloodborne Amyloid-Beta1-42 Peptide in the Brain and Leads to Alzheimer's Disease-Relevant Cognitive Changes in a Mouse Model. J Alzheimers Dis 2024; 98:163-186. [PMID: 38393907 PMCID: PMC10977376 DOI: 10.3233/jad-231028] [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] [Accepted: 12/26/2023] [Indexed: 02/25/2024]
Abstract
Background Increased blood-brain barrier (BBB) permeability and amyloid-β (Aβ) peptides (especially Aβ1-42) (Aβ42) have been linked to Alzheimer's disease (AD) pathogenesis, but the nature of their involvement in AD-related neuropathological changes leading to cognitive changes remains poorly understood. Objective To test the hypothesis that chronic extravasation of bloodborne Aβ42 peptide and brain-reactive autoantibodies and their entry into the brain parenchyma via a permeable BBB contribute to AD-related pathological changes and cognitive changes in a mouse model. Methods The BBB was rendered chronically permeable through repeated injections of Pertussis toxin (PT), and soluble monomeric, fluorescein isothiocyanate (FITC)-labeled or unlabeled Aβ42 was injected into the tail-vein of 10-month-old male CD1 mice at designated intervals spanning ∼3 months. Acquisition of learned behaviors and long-term retention were assessed via a battery of cognitive and behavioral tests and linked to neuropathological changes. Results Mice injected with both PT and Aβ42 demonstrated a preferential deficit in the capacity for long-term retention and an increased susceptibility to interference in selective attention compared to mice exposed to PT or saline only. Immunohistochemical analyses revealed increased BBB permeability and entry of bloodborne Aβ42 and immunoglobulin G (IgG) into the brain parenchyma, selective neuronal binding of IgG and neuronal accumulation of Aβ42 in animals injected with both PT and Aβ42 compared to controls. Conclusion Results highlight the potential synergistic role of BBB compromise and the influx of bloodborne Aβ42 into the brain in both the initiation and progression of neuropathologic and cognitive changes associated with AD.
Collapse
Affiliation(s)
- Nimish K. Acharya
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Biomarker Discovery Center, New Jersey Institute for Successful Aging (NJISA), Rowan-Virtua School of Osteopathic Medicine, Stratford, NJ, USA
- Rowan-Virtua Graduate School of Biomedical Sciences, Stratford, NJ, USA
- Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Rowan University, Glassboro, NJ, USA
| | | | - Peter M. Clifford
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- HNL Lab Medicine, Allentown, PA, USA
| | - Eli C. Levin
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Department of Graduate Medical Education, Bayhealth Medical Center, Dover, DE, USA
| | - Kenneth R. Light
- Department of Psychology, Barnard College of Columbia University, New York, NY, USA
| | - Hana Choi
- Rowan-Virtua Graduate School of Biomedical Sciences, Stratford, NJ, USA
| | - Randel L. Swanson II
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Rehab Medicine Service, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Physical Medicine and Rehabilitation, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mary C. Kosciuk
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
| | - Venkat Venkataraman
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Department of Academic and Student Affairs, Rowan-Virtua School of Osteopathic Medicine, Stratford, NJ, USA
| | - David J. Libon
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Department of Psychology, Rowan University, Glassboro, NJ, USA
| | - Louis D. Matzel
- Department of Psychology, Rutgers University, Piscataway, NJ, USA
| | - Robert G. Nagele
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Biomarker Discovery Center, New Jersey Institute for Successful Aging (NJISA), Rowan-Virtua School of Osteopathic Medicine, Stratford, NJ, USA
- Rowan-Virtua Graduate School of Biomedical Sciences, Stratford, NJ, USA
- Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Rowan University, Glassboro, NJ, USA
| |
Collapse
|
5
|
Shekarian M, Salehi I, Raoufi S, Asadbegi M, Kourosh-Arami M, Komaki A. Neuroprotective effects of vinpocetine, as a phosphodiesterase 1 inhibitor, on long-term potentiation in a rat model of Alzheimer's disease. BMC Neurosci 2023; 24:20. [PMID: 36927298 PMCID: PMC10018848 DOI: 10.1186/s12868-023-00790-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Vinpocetine (Vin) is known as a phosphodiesterase 1 inhibitor (PDE1-I) drug with multilateral effects, including antioxidant and anti-inflammatory activity. In this research, we investigated the neuroprotective and therapeutic effects of Vin through hippocampal synaptic plasticity on a rat's model of Alzheimer's disease (AD) induced by an intracerebroventricular (ICV) injection of beta-amyloid (Aβ). METHODS Sixty adult male Wistar rats were randomly divided into six groups: 1. control, 2. sham, 3. Aβ, 4. pretreatment (Vin + Aβ): Vin (4 mg/kg, gavage) for 30 days and then, inducing an AD model by an ICV injection of Aβ(1-42), 5. treatment (Aβ + Vin): inducing an AD model and then receiving Vin for 30 days by gavage, and 7. pretreatment + treatment (Vin + Aβ + Vin): receiving Vin by gavage for 30 days before and 30 days after the induction of an AD model. After these procedures, via stereotaxic surgery, the stimulating electrodes were placed at the perforant pathway (PP) and the recording electrodes were implanted in the dentate gyrus. RESULTS Excitatory postsynaptic potential (EPSP) slope and population spike (PS) amplitude in the Aβ group meaningfully diminished compared to the control group after the induction of long-term potentiation (LTP). CONCLUSIONS Vin could significantly prevent the Aβ effects on LTP. It can be concluded that pretreatment and treatment with Vin can be neuroprotective against harmful consequences of Aβ on hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Meysam Shekarian
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
| | - Iraj Salehi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
| | - Safoura Raoufi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
| | - Masoumeh Asadbegi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Komaki
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran.
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
6
|
Andrews EJ, Martini AC, Head E. Exploring the role of sex differences in Alzheimer's disease pathogenesis in Down syndrome. Front Neurosci 2022; 16:954999. [PMID: 36033603 PMCID: PMC9411995 DOI: 10.3389/fnins.2022.954999] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/25/2022] [Indexed: 11/14/2022] Open
Abstract
Women are disproportionately affected by Alzheimer's disease (AD), yet little is known about sex-specific effects on the development of AD in the Down syndrome (DS) population. DS is caused by a full or partial triplication of chromosome 21, which harbors the amyloid precursor protein (APP) gene, among others. The majority of people with DS in their early- to mid-40s will accumulate sufficient amyloid-beta (Aβ) in their brains along with neurofibrillary tangles (NFT) for a neuropathological diagnosis of AD, and the triplication of the APP gene is regarded as the main cause. Studies addressing sex differences with age and impact on dementia in people with DS are inconsistent. However, women with DS experience earlier age of onset of menopause, marked by a drop in estrogen, than women without DS. This review focuses on key sex differences observed with age and AD in people with DS and a discussion of possible underlying mechanisms that could be driving or protecting from AD development in DS. Understanding how biological sex influences the brain will lead to development of dedicated therapeutics and interventions to improve the quality of life for people with DS and AD.
Collapse
Affiliation(s)
- Elizabeth J. Andrews
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| | - Alessandra C. Martini
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
7
|
Gupta A, Uthayaseelan K, Uthayaseelan K, Kadari M, Subhan M, Saji Parel N, Krishna PV, Sange I. Alzheimer's Disease and Stroke: A Tangled Neurological Conundrum. Cureus 2022; 14:e25005. [PMID: 35712342 PMCID: PMC9194877 DOI: 10.7759/cureus.25005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2022] [Indexed: 11/05/2022] Open
|
8
|
Zhang Y, Wang X, Yang X, Yang X, Xue J, Yang Y. Ganoderic Acid A To Alleviate Neuroinflammation of Alzheimer's Disease in Mice by Regulating the Imbalance of the Th17/Tregs Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:14204-14214. [PMID: 34798773 DOI: 10.1021/acs.jafc.1c06304] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Ganoderic acid A (GAA) is a kind of lanostane-type triterpenoid isolated from Ganoderma lucidum. Imbalance of the Th17/Tregs axis exists in the progress of neuroinflammation of Alzheimer's disease (AD). In this study, the alleviating neuroinflammatory effect of GAA on d-galactose mice was studied from the aspect of regulating the imbalance of the Th17/Tregs axis. The Morris water maze test was used to evaluate the cognitive ability of AD mice. Flow cytometry was used to detect the percentages of IL-17A, IL-17F, IL-21, IL-22, and CD4+CD25+Foxp3+ in peripheral blood. Transmission electron microscopy was used to assess the cerebral mitochondrial ultrastructure. Metabolomic analysis based on gas chromatography-mass spectrometry was used to evaluate the mitochondrial dysfunction metabolism. Western blot analysis was used to detect the protein expressions of cytokines secreted by Th17 cells and Treg cells in the brain. As the results show, GAA has an alleviating neuroinflammatory effect on AD mice via regulating the imbalance of the Th17/Tregs axis. The potential mechanism was related to inhibition of the JAK/STAT signaling pathway induced by Th17 cells and enhancement of the mitochondrial oxidative phosphorylation by regulating Treg cells, thereby improving mitochondrial dysfunction of AD mice.
Collapse
Affiliation(s)
- Yan Zhang
- School of Chemical and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, P. R. China
| | - Xinyan Wang
- Graduate School, Jilin Institute of Chemical Technology, Jilin 132022, P. R. China
| | - Xiaomei Yang
- Nutritional Department, Jilin Medical University Affiliated Hospital, Jilin 132013, P. R. China
| | - Xiudong Yang
- School of Chemical and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, P. R. China
| | - Jianfei Xue
- School of Chemical and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, P. R. China
| | - Yanjun Yang
- School of Chemical and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, P. R. China
| |
Collapse
|
9
|
Archie SR, Al Shoyaib A, Cucullo L. Blood-Brain Barrier Dysfunction in CNS Disorders and Putative Therapeutic Targets: An Overview. Pharmaceutics 2021; 13:pharmaceutics13111779. [PMID: 34834200 PMCID: PMC8622070 DOI: 10.3390/pharmaceutics13111779] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 01/22/2023] Open
Abstract
The blood-brain barrier (BBB) is a fundamental component of the central nervous system (CNS). Its functional and structural integrity is vital to maintain the homeostasis of the brain microenvironment by controlling the passage of substances and regulating the trafficking of immune cells between the blood and the brain. The BBB is primarily composed of highly specialized microvascular endothelial cells. These cells’ special features and physiological properties are acquired and maintained through the concerted effort of hemodynamic and cellular cues from the surrounding environment. This complex multicellular system, comprising endothelial cells, astrocytes, pericytes, and neurons, is known as the neurovascular unit (NVU). The BBB strictly controls the transport of nutrients and metabolites into brain parenchyma through a tightly regulated transport system while limiting the access of potentially harmful substances via efflux transcytosis and metabolic mechanisms. Not surprisingly, a disruption of the BBB has been associated with the onset and/or progression of major neurological disorders. Although the association between disease and BBB disruption is clear, its nature is not always evident, specifically with regard to whether an impaired BBB function results from the pathological condition or whether the BBB damage is the primary pathogenic factor prodromal to the onset of the disease. In either case, repairing the barrier could be a viable option for treating and/or reducing the effects of CNS disorders. In this review, we describe the fundamental structure and function of the BBB in both healthy and altered/diseased conditions. Additionally, we provide an overview of the potential therapeutic targets that could be leveraged to restore the integrity of the BBB concomitant to the treatment of these brain disorders.
Collapse
Affiliation(s)
- Sabrina Rahman Archie
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (S.R.A.); (A.A.S.)
| | - Abdullah Al Shoyaib
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (S.R.A.); (A.A.S.)
| | - Luca Cucullo
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
- Correspondence: ; Tel.: +1-248-370-3884; Fax: +1-248-370-4060
| |
Collapse
|
10
|
Mesnil M, Defamie N, Naus C, Sarrouilhe D. Brain Disorders and Chemical Pollutants: A Gap Junction Link? Biomolecules 2020; 11:51. [PMID: 33396565 PMCID: PMC7824109 DOI: 10.3390/biom11010051] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
The incidence of brain pathologies has increased during last decades. Better diagnosis (autism spectrum disorders) and longer life expectancy (Parkinson's disease, Alzheimer's disease) partly explain this increase, while emerging data suggest pollutant exposures as a possible but still underestimated cause of major brain disorders. Taking into account that the brain parenchyma is rich in gap junctions and that most pollutants inhibit their function; brain disorders might be the consequence of gap-junctional alterations due to long-term exposures to pollutants. In this article, this hypothesis is addressed through three complementary aspects: (1) the gap-junctional organization and connexin expression in brain parenchyma and their function; (2) the effect of major pollutants (pesticides, bisphenol A, phthalates, heavy metals, airborne particles, etc.) on gap-junctional and connexin functions; (3) a description of the major brain disorders categorized as neurodevelopmental (autism spectrum disorders, attention deficit hyperactivity disorders, epilepsy), neurobehavioral (migraines, major depressive disorders), neurodegenerative (Parkinson's and Alzheimer's diseases) and cancers (glioma), in which both connexin dysfunction and pollutant involvement have been described. Based on these different aspects, the possible involvement of pollutant-inhibited gap junctions in brain disorders is discussed for prenatal and postnatal exposures.
Collapse
Affiliation(s)
- Marc Mesnil
- Laboratoire STIM, ERL7003 CNRS-Université de Poitiers, 1 rue G. Bonnet–TSA 51 106, 86073 Poitiers, France; (M.M.); (N.D.)
| | - Norah Defamie
- Laboratoire STIM, ERL7003 CNRS-Université de Poitiers, 1 rue G. Bonnet–TSA 51 106, 86073 Poitiers, France; (M.M.); (N.D.)
| | - Christian Naus
- Faculty of Medicine, Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T1Z3, Canada;
| | - Denis Sarrouilhe
- Laboratoire de Physiologie Humaine, Faculté de Médecine et Pharmacie, 6 rue de La Milétrie, bât D1, TSA 51115, 86073 Poitiers, France
| |
Collapse
|
11
|
Abstract
OBJECTIVES Many survivors of sepsis suffer long-term cognitive impairment, but the mechanisms of this association remain unknown. The objective of this study was to determine whether sepsis is associated with cerebral microinfarcts on brain autopsy. DESIGN Retrospective cohort study. SETTING AND SUBJECTS Five-hundred twenty-nine participants of the Adult Changes in Thought, a population-based prospective cohort study of older adults carried out in Kaiser Permanente Washington greater than or equal to 65 years old without dementia at study entry and who underwent brain autopsy. MEASUREMENTS AND MAIN RESULTS Late-life sepsis hospitalization was identified using administrative data. We identified 89 individuals with greater than or equal to 1 sepsis hospitalization during study participation, 80 of whom survived hospitalization and died a median of 169 days after discharge. Thirty percent of participants with one or more sepsis hospitalization had greater than two microinfarcts, compared with 19% participants without (χ p = 0.02); 20% of those with sepsis hospitalization had greater than two microinfarcts in the cerebral cortex, compared with 10% of those without (χ p = 0.01). The adjusted relative risk of greater than two microinfarcts was 1.61 (95% CI, 1.01-2.57; p = 0.04); the relative risk for having greater than two microinfarcts in the cerebral cortex was 2.12 (95% CI, 1.12-4.02; p = 0.02). There was no difference in Braak stage for neurofibrillary tangles or consortium to establish a registry for Alzheimer's disease score for neuritic plaques between, but Lewy bodies were less significantly common in those with sepsis. CONCLUSIONS Sepsis was specifically associated with moderate to severe vascular brain injury as assessed by microvascular infarcts. This association was stronger for microinfarcts within the cerebral cortex, with those who experienced severe sepsis hospitalization being more than twice as likely to have evidence of moderate to severe cerebral cortical injury in adjusted analyses. Further study to identify mechanisms for the association of sepsis and microinfarcts is needed.
Collapse
|
12
|
Jeon SG, Yoo A, Chun DW, Hong SB, Chung H, Kim JI, Moon M. The Critical Role of Nurr1 as a Mediator and Therapeutic Target in Alzheimer's Disease-related Pathogenesis. Aging Dis 2020; 11:705-724. [PMID: 32489714 PMCID: PMC7220289 DOI: 10.14336/ad.2019.0718] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/18/2019] [Indexed: 01/16/2023] Open
Abstract
Several studies have revealed that the transcription factor nuclear receptor related 1 (Nurr1) plays several roles not only in the regulation of gene expression related to dopamine synthesis, but also in alternative splicing, and miRNA targeting. Moreover, it regulates cognitive functions and protects against inflammation-induced neuronal death. In particular, the role of Nurr1 in the pathogenesis of Parkinson's disease (PD) has been well investigated; for example, it has been shown that it restores behavioral and histological impairments in PD models. Although many studies have evaluated the connection between Nurr1 and PD pathogenesis, the role of Nurr1 in Alzheimer's disease (AD) remain to be studied. There have been several studies describing Nurr1 protein expression in the AD brain. However, only a few studies have examined the role of Nurr1 in the context of AD. Therefore, in this review, we highlight the overall effects of Nurr1 under the neuropathologic conditions related to AD. Furthermore, we suggest the possibility of using Nurr1 as a therapeutic target for AD or other neurodegenerative disorders.
Collapse
Affiliation(s)
- Seong Gak Jeon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Anji Yoo
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Dong Wook Chun
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Sang Bum Hong
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Hyunju Chung
- Department of Core Research Laboratory, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
| | - Jin-il Kim
- Department of Nursing, College of Nursing, Jeju National University, Jeju-si 63243, Republic of Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| |
Collapse
|
13
|
Hui L, Soliman ML, Geiger NH, Miller NM, Afghah Z, Lakpa KL, Chen X, Geiger JD. Acidifying Endolysosomes Prevented Low-Density Lipoprotein-Induced Amyloidogenesis. J Alzheimers Dis 2020; 67:393-410. [PMID: 30594929 DOI: 10.3233/jad-180941] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cholesterol dyshomeostasis has been linked to the pathogenesis of sporadic Alzheimer's disease (AD). In furthering the understanding of mechanisms by which increased levels of circulating cholesterol augments the risk of developing sporadic AD, others and we have reported that low-density lipoprotein (LDL) enters brain parenchyma by disrupting the blood-brain barrier and that endolysosome de-acidification plays a role in LDL-induced amyloidogenesis in neurons. Here, we tested the hypothesis that endolysosome de-acidification was central to amyloid-β (Aβ) generation and that acidifying endolysosomes protects against LDL-induced increases in Aβ levels in neurons. We demonstrated that LDL, but not HDL, de-acidified endolysosomes and increased intraneuronal and secreted levels of Aβ. ML-SA1, an agonist of endolysosome-resident TRPML1 channels, acidified endolysosomes, and TRPML1 knockdown attenuated ML-SA1-induced endolysosome acidification. ML-SA1 blocked LDL-induced increases in intraneuronal and secreted levels of Aβ as well as Aβ accumulation in endolysosomes, prevented BACE1 accumulation in endolysosomes, and decreased BACE1 activity levels. LDL downregulated TRPML1 protein levels, and TRPML1 knockdown worsens LDL-induced increases in Aβ. Our findings suggest that endolysosome acidification by activating TRPML1 may represent a protective strategy against sporadic AD.
Collapse
Affiliation(s)
- Liang Hui
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Mahmoud L Soliman
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Nicholas H Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Nicole M Miller
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Zahra Afghah
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Koffi L Lakpa
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| |
Collapse
|
14
|
Abstract
Although Alzheimer's disease (AD) was described over a century ago, there are no effective approaches to its prevention and treatment. Such a slow progress is explained, at least in part, by our incomplete understanding of the mechanisms underlying the pathogenesis of AD. Here, I champion a hypothesis whereby AD is initiated on a disruption of the blood-brain barrier (BBB) caused by either genetic or non-genetic risk factors. The BBB disruption leads to an autoimmune response against pyramidal neurons located in the allo- and neocortical structures involved in memory formation and storage. The response caused by the adaptive immune system is not strong enough to directly kill neurons but may be sufficient to make them selectively vulnerable to neurofibrillary pathology. This hypothesis is based on the recent data showing that memory formation is associated with epigenetic chromatin modifications and, therefore, may be accompanied by expression of memory-specific proteins recognized by the immune system as "non-self" antigens. The autoimmune hypothesis is testable, and I discuss potential ways for its experimental and clinical verification. If confirmed, this hypothesis can radically change therapeutic approaches to AD prevention and treatment.
Collapse
Affiliation(s)
- Yuri I Arshavsky
- BioCircuits Institute, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
15
|
Katt ME, Mayo LN, Ellis SE, Mahairaki V, Rothstein JD, Cheng L, Searson PC. The role of mutations associated with familial neurodegenerative disorders on blood-brain barrier function in an iPSC model. Fluids Barriers CNS 2019; 16:20. [PMID: 31303172 PMCID: PMC6628493 DOI: 10.1186/s12987-019-0139-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/27/2019] [Indexed: 01/23/2023] Open
Abstract
Background Blood–brain barrier dysfunction is associated with many late-stage neurodegenerative diseases. An emerging question is whether the mutations associated with neurodegenerative diseases can independently lead to blood–brain barrier (BBB) dysfunction. Studies from patient-derived induced pluripotent stem cells suggest that mutations associated with neurodegenerative disease are non-cell autonomous, resulting in gain of toxic function in derived neurons and astrocytes. Here we assess whether selected mutations associated with neurodegenerative diseases can contribute to impairment of the blood–brain barrier. Methods We assessed barrier function of confluent monolayers of human brain microvascular endothelial cells (hBMECs) derived from induced pluripotent stem cells (iPSC) from three healthy individuals and eight individuals with neurodegenerative disease. We systematically assessed protein and gene expression of BBB biomarkers, transendothelial resistance (TEER), permeability of Lucifer yellow, permeability of d-glucose, permeability of rhodamine 123, the efflux ratio of rhodamine 123, and P-gp inhibition using Tariquidar for confluent monolayers of human brain microvascular endothelial cell (hBMECs). Results We provide evidence supporting the hypothesis that mutations associated with neurodegenerative disease can independently cause BBB dysfunction. These functional changes are not catastrophic since barrier breakdown would result in BBB impairment during development. Synergistic interactions between non-cell autonomous cerebrovascular dysfunction and the effects of gain-of-toxic function in neurons (e.g. toxic oligomers) are likely to increase disease burden through a positive feedback mechanism. Conclusions These results suggest that the accumulation of defects in brain microvascular endothelial cells may ultimately lead to impairment of the BBB. Small changes in barrier function over time could lead to accumulated defects that result in positive feedback to unrelated central nervous system diseases. Electronic supplementary material The online version of this article (10.1186/s12987-019-0139-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Moriah E Katt
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, 21218, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Lakyn N Mayo
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, 21218, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Shannon E Ellis
- Department of Biostatistics, Johns Hopkins University School of Public Health, Baltimore, MD, USA
| | - Vasiliki Mahairaki
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeffrey D Rothstein
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Linzhao Cheng
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, 21218, USA. .,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA. .,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
16
|
Paley EL. Diet-Related Metabolic Perturbations of Gut Microbial Shikimate Pathway-Tryptamine-tRNA Aminoacylation-Protein Synthesis in Human Health and Disease. Int J Tryptophan Res 2019; 12:1178646919834550. [PMID: 30944520 PMCID: PMC6440052 DOI: 10.1177/1178646919834550] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 02/04/2019] [Indexed: 12/26/2022] Open
Abstract
Human gut bacterial Na(+)-transporting NADH:ubiquinone reductase (NQR) sequence is associated with Alzheimer disease (AD). Here, Alzheimer disease-associated sequence (ADAS) is further characterized in cultured spore-forming Clostridium sp. Tryptophan and NQR substrate ubiquinone have common precursor chorismate in microbial shikimate pathway. Tryptophan-derived tryptamine presents in human diet and gut microbiome. Tryptamine inhibits tryptophanyl-tRNA synthetase (TrpRS) with consequent neurodegeneration in cell and animal models. Tryptophanyl-tRNA synthetase inhibition causes protein biosynthesis impairment similar to that revealed in AD. Tryptamine-induced TrpRS gene-dose reduction is associated with TrpRS protein deficiency and cell death. In animals, tryptamine treatment results in toxicity, weight gain, and prediabetes-related hypoglycemia. Sequence analysis of gut microbiome database reveals 89% to 100% ADAS nucleotide identity in American Indian (Cheyenne and Arapaho [C&A]) Oklahomans, of which ~93% being overweight or obese and 50% self-reporting type 2 diabetes (T2D). Alzheimer disease-associated sequence occurs in 10.8% of C&A vs 1.3% of healthy American population. This observation is of considerable interest because T2D links to AD and obesity. Alzheimer disease-associated sequence prevails in gut microbiome of colorectal cancer, which linked to AD. Metabolomics revealed that tryptamine, chorismate precursor quinate, and chorismate product 4-hydroxybenzoate (ubiquinone precursor) are significantly higher, while tryptophan-containing dipeptides are lower due to tRNA aminoacylation deficiency in C&A compared with non-native Oklahoman who showed no ADAS. Thus, gut microbial tryptamine overproduction correlates with ADAS occurrence. Antibiotic and diet additives induce ADAS and tryptamine. Mitogenic/cytotoxic tryptamine cause microbial and human cell death, gut dysbiosis, and consequent disruption of host-microbe homeostasis. Present analysis of 1246 participants from 17 human gut metagenomics studies revealed ADAS in cell death diseases.
Collapse
Affiliation(s)
- Elena L Paley
- Expert BioMed, Inc., Miami Dade, FL, USA.,Stop Alzheimers Corp, Miami Dade, FL, USA.,Nova Southeastern University, Fort Lauderdale, FL, USA
| |
Collapse
|
17
|
Moura RP, Martins C, Pinto S, Sousa F, Sarmento B. Blood-brain barrier receptors and transporters: an insight on their function and how to exploit them through nanotechnology. Expert Opin Drug Deliv 2019; 16:271-285. [PMID: 30767695 DOI: 10.1080/17425247.2019.1583205] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The blood-brain barrier (BBB) is a highly limiting barrier that prevents the brain from contacting with several circulating molecules, including harmful agents. However, certain systemic nutrients and macromolecules are able to cross the BBB and reach the brain parenchyma, involving the interaction with multiple receptors and/or transporters at the BBB surface. Nanotechnology allows the creation of drug vehicles, functionalized with targeting ligands for binding specific BBB receptors and/or transporters, hence triggering the transport through this biobarrier. AREAS COVERED This review focuses the BBB receptors/transporters to be exploited in regard to their overall structure and biologic function, as well as their role in the development of strategies envisaging drug delivery to the brain. Then, the interplay between the targeting of these BBB receptors/transporters and nanotechnology is explored, as they can increase by several-fold the effectiveness of brain-targeted therapies. EXPERT OPINION Nanomedicine may be particularly useful in brain drug delivery, mainly due to the possibility of functionalizing nanoparticles to target specific receptors/transporters. Since the BBB is endowed with numerous receptors and transporters responsible for regulating the proper metabolic activity of the brain, their targeting can be a promising bypass strategy to circumvent the hurdle that the BBB represents for brain drug delivery.
Collapse
Affiliation(s)
- Rui Pedro Moura
- a CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde , Gandra , Portugal
| | - Cláudia Martins
- b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal.,d ICBAS - Instituto de Ciências Biomédicas Abel Salazar , Universidade do Porto , Porto , Portugal
| | - Soraia Pinto
- b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal
| | - Flávia Sousa
- a CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde , Gandra , Portugal.,b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal.,d ICBAS - Instituto de Ciências Biomédicas Abel Salazar , Universidade do Porto , Porto , Portugal
| | - Bruno Sarmento
- a CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde , Gandra , Portugal.,b I3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal
| |
Collapse
|
18
|
Ott BR, Jones RN, Daiello LA, de la Monte SM, Stopa EG, Johanson CE, Denby C, Grammas P. Blood-Cerebrospinal Fluid Barrier Gradients in Mild Cognitive Impairment and Alzheimer's Disease: Relationship to Inflammatory Cytokines and Chemokines. Front Aging Neurosci 2018; 10:245. [PMID: 30186149 PMCID: PMC6110816 DOI: 10.3389/fnagi.2018.00245] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 07/25/2018] [Indexed: 01/30/2023] Open
Abstract
Background: The pathophysiology underlying altered blood-cerebrospinal fluid barrier (BCSFB) function in Alzheimer's disease (AD) is unknown but may relate to endothelial cell activation and cytokine mediated inflammation. Methods: Cerebrospinal fluid (CSF) and peripheral blood were concurrently collected from cognitively healthy controls (N = 21) and patients with mild cognitive impairment (MCI) (N = 8) or AD (N = 11). The paired serum and CSF samples were assayed for a panel of cytokines, chemokines, and related trophic factors using multiplex ELISAs. Dominance analysis models were conducted to determine the relative importance of the inflammatory factors in relationship to BCSFB permeability, as measured by CSF/serum ratios for urea, creatinine, and albumin. Results: BCSFB disruption to urea, a small molecule distributed by passive diffusion, had a full model coefficient of determination (r2) = 0.35, and large standardized dominance weights (>0.1) for monocyte chemoattractant protein-1, interleukin (IL)-15, IL-1rα, and IL-2 in serum. BCSFB disruption to creatinine, a larger molecule governed by active transport, had a full model r2 = 0.78, and large standardized dominance weights for monocyte inhibitor protein-1b in CSF and tumor necrosis factor-α in serum. BCSFB disruption to albumin, a much larger molecule, had a full model r2 = 0.62, and large standardized dominance weights for IL-17a, interferon-gamma, IL-2, and VEGF in CSF, as well IL-4 in serum. Conclusions: Inflammatory proteins have been widely documented in the AD brain. The results of the current study suggest that changes in BCSFB function resulting in altered permeability and transport are related to expression of specific inflammatory proteins, and that the shifting distribution of these proteins from serum to CSF in AD and MCI is correlated with more severe perturbations in BCSFB function.
Collapse
Affiliation(s)
- Brian R. Ott
- Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States,George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States,*Correspondence: Brian R. Ott
| | - Richard N. Jones
- Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Lori A. Daiello
- Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Suzanne M. de la Monte
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States,Division of Neuropathology, Department of Pathology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Edward G. Stopa
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States,Division of Neuropathology, Department of Pathology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Conrad E. Johanson
- Department of Neurosurgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Charles Denby
- Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Paula Grammas
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States
| |
Collapse
|
19
|
Vojdani A, Vojdani E. Amyloid-Beta 1-42 Cross-Reactive Antibody Prevalent in Human Sera May Contribute to Intraneuronal Deposition of A-Beta-P-42. Int J Alzheimers Dis 2018; 2018:1672568. [PMID: 30034864 PMCID: PMC6032666 DOI: 10.1155/2018/1672568] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/13/2018] [Accepted: 04/29/2018] [Indexed: 12/11/2022] Open
Abstract
Antibodies against many neural antigens are detected in the sera of both patients with Alzheimer's disease (AD) and some healthy individuals. Blood-brain barrier dysfunction could make it possible for brain-reactive autoantibodies to reach the brain, where they can react with amyloid ß peptide (AßP). The origin of these autoreactive antibodies in the blood is unclear. The goals of this study were as follows: (1) to examine the immune reactivity of anti-AßP-42 with 22 neuronal and other associated antigens, some of which are involved in the pathophysiology of AD; (2) to classify antibodies to these 22 different antigens into those that cross-react with AßP-42 and those that do not; (3) to determine whether these antibodies react with BBB proteins, nerve growth factors, and enteric neuronal antigens. Using monoclonal AßP-42 antibody and ELISA methodology, we found that the antibody was highly reactive with Aß protein, tau protein, presenilin, rabaptin-5, β-NGF, BDNF, mTG, and enteric nerve. The same antibody produced equivocal to moderate reactions with glutamate-R, S100B, AQP4, GFAP, MBP, α-synuclein, tTG-2, and tTG-3, and not with the rest. These antibodies were also measured in blood samples from 47 AD patients and 47 controls. IgG antibodies were found to be elevated against AßP-42 and many other antigens in a significant percentage of controls. Overall, the mean OD values were significantly higher against 9/23 tested antigens (p <0.001) in the samples with AD. We were indeed able to classify the detected neuronal antibodies into those that cross-react with AßP-42 and those that do not. Our main finding is that although these antibodies may be harmless in a subgroup of controls, in individuals with compromised BBBs these antibodies that cross-react with AßP-42 can reach the brain, where their cross-reactivity with AßP-42 may contribute to the onset and progression of AD, and perhaps other neurodegenerative disorders.
Collapse
Affiliation(s)
- Aristo Vojdani
- Immunosciences Lab., Inc., 822 S. Robertson Blvd., Ste. 312, Los Angeles, CA 90035, USA
- Department of Preventive Medicine, Loma Linda University School of Medicine, 24785 Stewart St., Evans Hall, Ste. 111, Loma Linda, CA 92354, USA
| | - Elroy Vojdani
- Regenera Medical, 11860 Wilshire Blvd., Ste. 301, Los Angeles, CA 90025, USA
| |
Collapse
|
20
|
The effects of cerebral amyloid angiopathy on integrity of the blood-brain barrier. Neurobiol Aging 2018; 70:70-77. [PMID: 30007166 DOI: 10.1016/j.neurobiolaging.2018.06.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 05/21/2018] [Accepted: 06/04/2018] [Indexed: 12/12/2022]
Abstract
Cerebral amyloid angiopathy (CAA), in which amyloid accumulates predominantly in the walls of arterioles and capillaries, is seen in most patients with Alzheimer disease (AD) and may contribute to compromise of blood-brain barrier (BBB) function seen in AD. We investigated the effects of CAA on BBB integrity by examining the expression of the endothelial marker CD31, basement membrane protein collagen IV (COL4), tight junction protein claudin-5, and fibrinogen, a marker of BBB leakage, by immunohistochemistry in the occipital cortex of autopsy brains with AD and capillary CAA (CAA type 1; n = 8), AD with noncapillary CAA (CAA type 2; n = 10), and AD without CAA (n = 7) compared with elderly controls (n = 10). Given the difference in pathogenesis of capillary and noncapillary CAA, we hypothesize that features of BBB breakdown are observed only in capillary CAA. We found decreased expression of CD31 in AD subjects with CAA types 1 and 2 compared with AD without CAA and an increase in COL4 in AD without CAA compared with controls. Furthermore, there was increased immunoreactivity for fibrinogen in AD with CAA type 1 compared with controls. These findings suggest that capillary CAA is associated with morphologic and possibly physiologic alterations of the neurovascular unit and increased BBB permeability in AD.
Collapse
|
21
|
Oberstein TJ, Taha L, Spitzer P, Hellstern J, Herrmann M, Kornhuber J, Maler JM. Imbalance of Circulating T h17 and Regulatory T Cells in Alzheimer's Disease: A Case Control Study. Front Immunol 2018; 9:1213. [PMID: 29915582 PMCID: PMC5994416 DOI: 10.3389/fimmu.2018.01213] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/15/2018] [Indexed: 01/01/2023] Open
Abstract
The neuropathological hallmarks of Alzheimer's disease (AD), i.e., neuritic plaques and neurofibrillary tangles, consist of beta amyloid peptides (Aβ) and hyperphosphorylated Tau. These are accompanied by reactive microglia and astrocytes in the vicinity of the neuritic plaques and by changes to the peripheral immune system, e.g., an increase of the pro-inflammatory cytokines IL-1β, IL-6, and TNF-α in the peripheral blood. To address a potential involvement of peripheral T helper cell (Th) subsets in AD, we conducted a case control study with 54 individuals with AD dementia (n = 14), with mild cognitive impairment (MCI) due to AD (MCIAD, n = 14), with MCI unlikely due to AD (MCIother, n = 13), and controls without cognitive impairment (controls, n = 13). The proportions of CD3+CD8-IL-17A+IFNγ- Th17 cells, CD3+CD8-IL-17A-IFNγ+ Th1 cells, and CD4+CD127lowCD25+ regulatory T cells (Tregs) were assessed by flow cytometry. In addition, the correlations of the proportions of Th subsets to cerebrospinal fluid biomarkers were studied. CD3+CD8-IL-17A+IFNγ- Th17 cells were significantly increased in subjects with MCIAD compared to age- and sex-matched subjects with MCIother and controls (MCIAD mean = 1.13, SD = 0.77; MCIother mean = 0.58, SD = 0.28; and controls mean = 0.52, SD = 0.22; p = 0.008). The proportion of CD4+CD127lowCD25+ Tregs was not altered between the different groups, but it significantly positively related with the levels of total Tau and pTau181 (rTreg|totalTau = 0.43, p = 0.021, n = 28; rTreg|pTau181 = 0.46; p = 0.024, n = 28) in subjects with AD but not in nonAD controls (rTreg|totalTau = -0.51, p = 0.007, n = 26). The increase of circulating CD3+CD8-IL-17A+IFNγ- Th17 cells in the early stages of AD and the association of CD4+CD127lowCD25+ Tregs with neurodegeneration marker Tau may indicate that the adaptive immune system relates to neuropathological changes in AD.
Collapse
Affiliation(s)
- Timo Jan Oberstein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Lava Taha
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Philipp Spitzer
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Janina Hellstern
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University of Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Juan Manuel Maler
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
22
|
Monteiro J, Alves MG, Oliveira PF, Silva BM. Pharmacological potential of methylxanthines: Retrospective analysis and future expectations. Crit Rev Food Sci Nutr 2018; 59:2597-2625. [PMID: 29624433 DOI: 10.1080/10408398.2018.1461607] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Methylated xanthines (methylxanthines) are available from a significant number of different botanical species. They are ordinarily included in daily diet, in many extremely common beverages and foods. Caffeine, theophylline and theobromine are the main methylxanthines available from natural sources. The supposedly relatively low toxicity of methylxanthines, combined with the many beneficial effects that have been attributed to these compounds through time, generated a justified attention and a very prolific ground for dedicated scientific reports. Methylxanthines have been widely used as therapeutical tools, in an intriguing range of medicinal scopes. In fact, methylxanthines have been/were medically used as Central Nervous System stimulants, bronchodilators, coronary dilators, diuretics and anti-cancer adjuvant treatments. Other than these applications, methylxanthines have also been hinted to hold other beneficial health effects, namely regarding neurodegenerative diseases, cardioprotection, diabetes and fertility. However, it seems now consensual that toxicity concerns related to methylxanthine consumption and/or therapeutic use should not be dismissed. Taking all the knowledge and expectations on the potential of methylxanthines into account, we propose a systematic look at the past and future of methylxanthine pharmacologic applications, discussing all the promise and anticipating possible constraints. Anyways, methylxanthines will still substantiate considerable meaningful research and discussion for years to come.
Collapse
Affiliation(s)
- João Monteiro
- Mass Spectrometry Centre, Department of Chemistry & CESAM, University of Aveiro, Campus Universitário de Santiago , Aveiro , Portugal
| | - Marco G Alves
- Department of Microscopy, Laboratory of Cell Biology, Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto , Portugal
| | - Pedro F Oliveira
- Department of Microscopy, Laboratory of Cell Biology, Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto , Portugal.,Institute of Health Research an Innovation (i3S), University of Porto , Porto , Portugal
| | | |
Collapse
|
23
|
Gioscia-Ryan RA, Battson ML, Cuevas LM, Eng JS, Murphy MP, Seals DR. Mitochondria-targeted antioxidant therapy with MitoQ ameliorates aortic stiffening in old mice. J Appl Physiol (1985) 2018; 124:1194-1202. [PMID: 29074712 PMCID: PMC6008077 DOI: 10.1152/japplphysiol.00670.2017] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/10/2017] [Accepted: 10/20/2017] [Indexed: 12/21/2022] Open
Abstract
Aortic stiffening is a major independent risk factor for cardiovascular diseases, cognitive dysfunction, and other chronic disorders of aging. Mitochondria-derived reactive oxygen species are a key source of arterial oxidative stress, which may contribute to arterial stiffening by promoting adverse structural changes-including collagen overabundance and elastin degradation-and enhancing inflammation, but the potential for mitochondria-targeted therapeutic strategies to ameliorate aortic stiffening with primary aging is unknown. We assessed aortic stiffness [pulse-wave velocity (aPWV)], ex vivo aortic intrinsic mechanical properties [elastic modulus (EM) of collagen and elastin regions], and aortic protein expression in young (~6 mo) and old (~27 mo) male C57BL/6 mice consuming normal drinking water (YC and OC) or water containing mitochondria-targeted antioxidant MitoQ (250 µM; YMQ and OMQ) for 4 wk. Both baseline and postintervention aPWV values were higher in OC vs. YC (post: 482 ± 21 vs. 420 ± 5 cm/s, P < 0.05). MitoQ had no effect in young mice but decreased aPWV in old mice (OMQ, 426 ± 20, P < 0.05 vs. OC). MitoQ did not affect age-associated increases in aortic collagen-region EM, collagen expression, or proinflammatory cytokine expression, but partially attenuated age-associated decreases in elastin region EM and elastin expression. Our results demonstrate that MitoQ reverses in vivo aortic stiffness in old mice and suggest that mitochondria-targeted antioxidants may represent a novel, promising therapeutic strategy for decreasing aortic stiffness with primary aging and, possibly, age-related clinical disorders in humans. The destiffening effects of MitoQ treatment may be at least partially mediated by attenuation/reversal of age-related aortic elastin degradation. NEW & NOTEWORTHY We show that 4 wk of treatment with the mitochondria-specific antioxidant MitoQ in mice completely reverses the age-associated elevation in aortic stiffness, assessed as aortic pulse-wave velocity. The destiffening effects of MitoQ treatment may be at least partially mediated by attenuation of age-related aortic elastin degradation. Our results suggest that mitochondria-targeted therapeutic strategies may hold promise for decreasing arterial stiffening with aging in humans, possibly decreasing the risk of many chronic age-related clinical disorders.
Collapse
Affiliation(s)
- Rachel A Gioscia-Ryan
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, Colorado
| | - Micah L Battson
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, Colorado
| | - Lauren M Cuevas
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, Colorado
| | - Jason S Eng
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, Colorado
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge , Cambridge , United Kingdom
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, Colorado
| |
Collapse
|
24
|
Izawa Y, Gu YH, Osada T, Kanazawa M, Hawkins BT, Koziol JA, Papayannopoulou T, Spatz M, Del Zoppo GJ. β1-integrin-matrix interactions modulate cerebral microvessel endothelial cell tight junction expression and permeability. J Cereb Blood Flow Metab 2018; 38:641-658. [PMID: 28787238 PMCID: PMC5888854 DOI: 10.1177/0271678x17722108] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Acutely following focal cerebral ischemia disruption of the microvessel blood-brain barrier allows transit of plasma proteins into the neuropil as edema formation that coincides with loss of microvessel endothelial β1-integrins. We extend previous findings to show that interference with endothelial β1-integrin-matrix adhesion by the monoclonal IgM Ha2/5 increases the permeability of primary cerebral microvascular endothelial cell monolayers through reorganization of claudin-5, occludin, and zonula occludens-1 (ZO-1) from inter-endothelial borders. Interference with β1-integrin-matrix adhesion initiates F-actin conformational changes that coincide with claudin-5 redistribution. β1-integrin-matrix interference simultaneously increases phosphorylation of myosin light chain (MLC), while inhibition of MLC kinase (MLCK) and Rho kinase (ROCK) abolishes the Ha2/5-dependent increased endothelial permeability by 6 h after β1-integrin-matrix interference. These observations are supported by concordant observations in the cortex of a high-quality murine conditional β1-integrin deletion construct. Together they support the hypothesis that detachment of β1-integrins from abluminal matrix ligands increases vascular endothelial permeability through reorganization of tight junction (TJ) proteins via altered F-actin conformation, and indicate that the β1-integrin-MLC signaling pathway is engaged when β1-integrin detachment occurs. These findings provide a novel approach to the research and treatment of cerebral disorders where the breakdown of the blood-brain barrier accounts for their progression and complication.
Collapse
Affiliation(s)
- Yoshikane Izawa
- 1 Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.,2 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Yu-Huan Gu
- 1 Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Takashi Osada
- 1 Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.,2 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Masato Kanazawa
- 1 Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.,3 Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Brian T Hawkins
- 1 Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.,4 Discovery, Science, & Technology, RTI International, Research Triangle Park, NC, USA
| | - James A Koziol
- 5 Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Thalia Papayannopoulou
- 1 Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Maria Spatz
- 6 Stroke Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Gregory J Del Zoppo
- 1 Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.,7 Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
25
|
Vergauwen E, Vanbinst AM, Brussaard C, Janssens P, De Clerck D, Van Lint M, Houtman AC, Michel O, Keymolen K, Lefevere B, Bohler S, Michielsen D, Jansen AC, Van Velthoven V, Gläsker S. Central nervous system gadolinium accumulation in patients undergoing periodical contrast MRI screening for hereditary tumor syndromes. Hered Cancer Clin Pract 2018; 16:2. [PMID: 29312473 PMCID: PMC5756358 DOI: 10.1186/s13053-017-0084-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/28/2017] [Indexed: 12/17/2022] Open
Abstract
Background Patients with hereditary tumor syndromes undergo periodical magnetic resonance imaging (MRI) screening with Gadolinium contrast. Gadolinium accumulation has recently been described in the central nervous system after repeated administrations. The prevalence and rate of accumulation in different subgroups of patients are unknown. Neither are the mechanism nor clinical impact. This may cause uncertainty about the screening. To explore the prevalence and rate of Gadolinium accumulation in different subgroups, we retrospectively analyzed MRIs of patients with von Hippel-Lindau disease (VHL) and Tuberous Sclerosis Complex (TSC). Methods We determined the prevalence and rate of accumulation in the dentate nucleus and globus pallidus on unenhanced T1-weighted MRI from VHL and TSC patients. We compared the signal intensities of these regions to the signal intensity of the pons. We evaluated the impact of number of MRIs, kidney function and liver function on Gadolinium accumulation. Results Twenty eight VHL patients and 24 TSC patients were included. The prevalence of accumulation in the dentate nucleus and globus pallidus increased linearly according to number of Gadolinium enhanced MRIs and was higher in the VHL group (100%). A significant linear correlation between number of MRIs and increased signal intensity was observed in the VHL group. Conclusions Gadolinium accumulation occurs in almost all patients undergoing contrast MRI screening after >5 MRIs. We advocate a screening protocol for patients with hereditary tumor syndromes that minimizes the Gadolinium dose. This can be accomplished by using a single administration to simultaneously screen for brain, spine and/or abdominal lesions, using an MRI protocol focused on either VHL- or TSC-specific lesions. Higher prevalence and rate of accumulation in VHL patients may be explained by the typical vascular leakage accompanying central nervous system hemangioblastomas.
Collapse
Affiliation(s)
- Evelynn Vergauwen
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| | | | - Carola Brussaard
- Department of Radiology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Peter Janssens
- Department of Nephrology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Dieter De Clerck
- Department of Nephrology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Michel Van Lint
- Department of Ophthalmology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Anne C Houtman
- Department of Ophthalmology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Olaf Michel
- Department of Otorhinolaryngology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Kathelijn Keymolen
- Department of Genetics, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Bieke Lefevere
- Department of Psychology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Susanne Bohler
- Department of Psychology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Dirk Michielsen
- Department of Urology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Anna C Jansen
- Department of Pediatrics, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Vera Van Velthoven
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Sven Gläsker
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| |
Collapse
|
26
|
Lai AY, McLaurin J. Rho-associated protein kinases as therapeutic targets for both vascular and parenchymal pathologies in Alzheimer's disease. J Neurochem 2017; 144:659-668. [PMID: 28722749 DOI: 10.1111/jnc.14130] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/21/2017] [Accepted: 07/14/2017] [Indexed: 12/30/2022]
Abstract
The causes of late-onset Alzheimer's disease are unclear and likely multifactorial. Rho-associated protein kinases (ROCKs) are ubiquitously expressed signaling messengers that mediate a wide array of cellular processes. Interestingly, they play an important role in several vascular and brain pathologies implicated in Alzheimer's etiology, including hypertension, hypercholesterolemia, blood-brain barrier disruption, oxidative stress, deposition of vascular and parenchymal amyloid-beta peptides, tau hyperphosphorylation, and cognitive decline. The current review summarizes the functions of ROCKs with respect to the various risk factors and pathologies on both sides of the blood-brain barrier and present support for targeting ROCK signaling as a multifactorial and multi-effect approach for the prevention and amelioration of late-onset Alzheimer's disease. This article is part of the Special Issue "Vascular Dementia".
Collapse
Affiliation(s)
- Aaron Y Lai
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - JoAnne McLaurin
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Theodorakis PE, Müller EA, Craster RV, Matar OK. Physical insights into the blood-brain barrier translocation mechanisms. Phys Biol 2017; 14:041001. [PMID: 28586313 DOI: 10.1088/1478-3975/aa708a] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The number of individuals suffering from diseases of the central nervous system (CNS) is growing with an aging population. While candidate drugs for many of these diseases are available, most of these pharmaceutical agents cannot reach the brain rendering most of the drug therapies that target the CNS inefficient. The reason is the blood-brain barrier (BBB), a complex and dynamic interface that controls the influx and efflux of substances through a number of different translocation mechanisms. Here, we present these mechanisms providing, also, the necessary background related to the morphology and various characteristics of the BBB. Moreover, we discuss various numerical and simulation approaches used to study the BBB, and possible future directions based on multi-scale methods. We anticipate that this review will motivate multi-disciplinary research on the BBB aiming at the design of effective drug therapies.
Collapse
|
28
|
Yu LE, Lai CL, Lee CT, Wang JY. Highly electronegative low-density lipoprotein L5 evokes microglial activation and creates a neuroinflammatory stress via Toll-like receptor 4 signaling. J Neurochem 2017; 142:231-245. [PMID: 28444734 DOI: 10.1111/jnc.14053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/30/2017] [Accepted: 04/19/2017] [Indexed: 12/23/2022]
Abstract
Atherogenic risk factors, such as hypercholesterolemia, are associated with increased risk of neurodegeneration, especially Alzheimer's dementia. Human plasma electronegative low-density lipoprotein [LDL(-)], especially L5, may serve as an important contributing factor. L5 promoting an inflammatory action in atherosclerosis has been extensively studied. However, the role of L5 in inducing neuroinflammation remains unknown. Here, we examined the impact of L5 on immune activation and cell viability in cultured BV-2 microglia. BV-2 cells treated with lipopolysaccharide or human LDLs (L1, L5, or oxLDL) were subjected to molecular/biochemical assays for measuring microglial activation, levels of inflammatory factors, and cell survival. A transwell BV-2/N2a co-culture was used to assess N2a cell viability following BV-2 cell exposure to L5. We found that L5 enables the activation of microglia and elicits an inflammatory response, as evidenced by increased oxygen/nitrogen free radicals (nitric oxide, reactive oxygen species, and peroxides), elevated tumor necrosis factor-α levels, decreased basal interleukin-10 levels, and augmented production of pro-inflammatory proteins (inducible nitric oxide synthase and cyclooxygenase-2). L5 also triggered BV-2 cell death primarily via apoptosis. These effects were markedly disrupted by the application of signaling pathway inhibitors, thus demonstrating that L5 interacts with Toll-like receptor 4 to modulate multiple pathways, including MAPKs, PI3K/Akt, and NF-κB. Decreased N2a cell viability in a transwell co-culture was mainly ascribed to L5-induced BV-2 cell activation. Together, our data suggest that L5 creates a neuroinflammatory stress via microglial Toll-like receptor 4, thereby leading to the death of BV-2 microglia and coexistent N2a cells. Atherogenic L5 possibly contributes to neuroinflammation-related neurodegeneration.
Collapse
Affiliation(s)
- Liang-En Yu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chiou-Lian Lai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ching-Tien Lee
- Department of Nursing, Hsin-Sheng College of Medical Care and Management, Taoyuan, Taiwan
| | - Jiz-Yuh Wang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
29
|
Depletion of coagulation factor XII ameliorates brain pathology and cognitive impairment in Alzheimer disease mice. Blood 2017; 129:2547-2556. [PMID: 28242605 DOI: 10.1182/blood-2016-11-753202] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/21/2017] [Indexed: 12/21/2022] Open
Abstract
Vascular abnormalities and inflammation are found in many Alzheimer disease (AD) patients, but whether these changes play a causative role in AD is not clear. The factor XII (FXII) -initiated contact system can trigger both vascular pathology and inflammation and is activated in AD patients and AD mice. We have investigated the role of the contact system in AD pathogenesis. Cleavage of high-molecular-weight kininogen (HK), a marker for activation of the inflammatory arm of the contact system, is increased in a mouse model of AD, and this cleavage is temporally correlated with the onset of brain inflammation. Depletion of FXII in AD mice inhibited HK cleavage in plasma and reduced neuroinflammation, fibrinogen deposition, and neurodegeneration in the brain. Moreover, FXII-depleted AD mice showed better cognitive function than untreated AD mice. These results indicate that FXII-mediated contact system activation contributes to AD pathogenesis, and therefore this system may offer novel targets for AD treatment.
Collapse
|
30
|
Wardlaw JM, Makin SJ, Valdés Hernández MC, Armitage PA, Heye AK, Chappell FM, Muñoz‐Maniega S, Sakka E, Shuler K, Dennis MS, Thrippleton MJ. Blood‐brain barrier failure as a core mechanism in cerebral small vessel disease and dementia: evidence from a cohort study. Alzheimers Dement 2016. [PMCID: PMC5472180 DOI: 10.1016/j.jalz.2016.09.006] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Introduction Methods Results Discussion In 201 patients with non-disabling stroke and white matter hyperintensities (WMH), we measured blood-brain barrier (BBB) leakage with contrast-enhanced MRI. BBB leakage was higher in WMH than in normal appearing white matter. BBB leakage increased in both WMH and normal appearing white matter with the burden of small vessel disease, hypertension, and age. BBB leakage predicted cognitive decline one year later. The leakage pattern supported the hypothesis that BBB leak was pathogenic of diffuse brain damage in cerebral small vessel disease.
Collapse
Affiliation(s)
- Joanna M. Wardlaw
- Centre for Clinical Brain Sciences University of Edinburgh Edinburgh UK
| | - Stephen J. Makin
- Centre for Clinical Brain Sciences University of Edinburgh Edinburgh UK
| | | | - Paul A. Armitage
- Academic Unit of Radiology, Department of Cardiovascular Science University of Sheffield, Royal Hallamshire Hospital Sheffield UK
| | - Anna K. Heye
- Centre for Clinical Brain Sciences University of Edinburgh Edinburgh UK
| | | | | | - Eleni Sakka
- Centre for Clinical Brain Sciences University of Edinburgh Edinburgh UK
| | - Kirsten Shuler
- Centre for Clinical Brain Sciences University of Edinburgh Edinburgh UK
| | - Martin S. Dennis
- Centre for Clinical Brain Sciences University of Edinburgh Edinburgh UK
| | | |
Collapse
|
31
|
Kajiwara Y, McKenzie A, Dorr N, Gama Sosa MA, Elder G, Schmeidler J, Dickstein DL, Bozdagi O, Zhang B, Buxbaum JD. The human-specific CASP4 gene product contributes to Alzheimer-related synaptic and behavioural deficits. Hum Mol Genet 2016; 25:4315-4327. [PMID: 27516385 DOI: 10.1093/hmg/ddw265] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 06/09/2016] [Accepted: 07/28/2016] [Indexed: 12/13/2022] Open
Abstract
Recent studies have indicated that innate immune signalling molecules are involved in late-onset Alzheimer's disease (LOAD) risk. Amyloid beta (Aβ) accumulates in AD brain, and has been proposed to act as a trigger of innate immune responses. Caspase-4 is an important part of the innate immune response. We recently characterized transgenic mice carrying human CASP4, and observed that the mice manifested profound innate immune responses to lipopolysaccharide (LPS). Since these inflammatory processes are important in the aetiology of AD, we have now analysed the correlation of expression of caspase-4 in human brain with AD risk genes, and studied caspase-4 effects on AD-related phenotypes in APPswe/PS1deltaE9 (APP/PS1) mice. We observed that the expression of caspase-4 was strongly correlated with AD risk genes including TYROBP, TREM2, CR1, PSEN1, MS4A4A and MS4A6A in LOAD brains. Caspase-4 expression was upregulated in CASP4/APP/PS1 mice in a region-specific manner, including hippocampus and prefrontal cortex. In APP/PS1 mice, caspase-4 expression led to impairments in the reversal phase of a Barnes maze task and in hippocampal synaptic plasticity, without affecting soluble or aggregated Aβ levels. Caspase-4 was expressed predominantly in microglial cells, and in the presence of CASP4, more microglia were clustered around amyloid plaques. Furthermore, our data indicated that caspase-4 modulates microglial cells in a manner that increases proinflammatory processes. We propose that microglial caspase-4 expression contributes to the cognitive impairments in AD, and that further study of caspase-4 will enhance our understanding of AD pathogenesis and may lead to novel therapeutic targets in AD.
Collapse
Affiliation(s)
| | - Andrew McKenzie
- Department of Genetics and Genomic Sciences.,Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, USA
| | | | | | - Gregory Elder
- Department of Psychiatry.,Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA.,Department of Neurology
| | | | - Dara L Dickstein
- Department of Neuroscience.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Bin Zhang
- Department of Genetics and Genomic Sciences.,Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Joseph D Buxbaum
- Department of Psychiatry .,Department of Genetics and Genomic Sciences.,Department of Neuroscience.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
32
|
Virk SA, Eslick GD. Aluminum Levels in Brain, Serum, and Cerebrospinal Fluid are Higher in Alzheimer's Disease Cases than in Controls: A Series of Meta-Analyses. J Alzheimers Dis 2016; 47:629-38. [PMID: 26401698 DOI: 10.3233/jad-150193] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Aluminum is the most studied environmental agent linked with Alzheimer's disease (AD). However, it remains unclear whether levels are significantly elevated in AD sufferers. OBJECTIVE To systematically assess levels of aluminum in brain, serum, and cerebrospinal fluid (CSF) of AD cases and controls. METHODS Electronic searches of Medline, Embase, PubMed, and Cochrane Library were conducted up to June 2015. Studies reporting brain, serum, or CSF aluminum levels in individuals with AD and non-demented controls were included. Meta-analyses were performed using random-effects models and the pooled standardized mean difference (SMD) reported with 95% confidence intervals (CI). RESULTS Overall, 34 studies involving 1,208 participants and 613 AD cases met the criteria for inclusion. Aluminum was measured in brain tissue in 20 studies (n = 386), serum in 12 studies (n = 698), and CSF in 4 studies (n = 124). Compared to control subjects, AD sufferers had significantly higher levels of brain (SMD 0.88; 95% CI, 0.25-1.51), serum (SMD 0.28; 95% CI, 0.03-0.54), and CSF (SMD 0.48; 95% CI, 0.03-0.93) aluminum. Sensitivity analyses excluding studies without age-matched controls did not impact upon these results. CONCLUSIONS The findings of the present meta-analyses demonstrate that aluminum levels are significantly elevated in brain, serum, and CSF of patients with AD. These findings suggest that elevated aluminum levels, particularly in serum, may serve as an early marker of AD and/or play a role in the development of the disease. These results substantially clarify the existing evidence examining the link between chronic aluminum exposure and the development of AD.
Collapse
|
33
|
Di Marco LY, Farkas E, Martin C, Venneri A, Frangi AF. Is Vasomotion in Cerebral Arteries Impaired in Alzheimer's Disease? J Alzheimers Dis 2016; 46:35-53. [PMID: 25720414 PMCID: PMC4878307 DOI: 10.3233/jad-142976] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A substantial body of evidence supports the hypothesis of a vascular component in the pathogenesis of Alzheimer’s disease (AD). Cerebral hypoperfusion and blood-brain barrier dysfunction have been indicated as key elements of this pathway. Cerebral amyloid angiopathy (CAA) is a cerebrovascular disorder, frequent in AD, characterized by the accumulation of amyloid-β (Aβ) peptide in cerebral blood vessel walls. CAA is associated with loss of vascular integrity, resulting in impaired regulation of cerebral circulation, and increased susceptibility to cerebral ischemia, microhemorrhages, and white matter damage. Vasomotion— the spontaneous rhythmic modulation of arterial diameter, typically observed in arteries/arterioles in various vascular beds including the brain— is thought to participate in tissue perfusion and oxygen delivery regulation. Vasomotion is impaired in adverse conditions such as hypoperfusion and hypoxia. The perivascular and glymphatic pathways of Aβ clearance are thought to be driven by the systolic pulse. Vasomotion produces diameter changes of comparable amplitude, however at lower rates, and could contribute to these mechanisms of Aβ clearance. In spite of potential clinical interest, studies addressing cerebral vasomotion in the context of AD/CAA are limited. This study reviews the current literature on vasomotion, and hypothesizes potential paths implicating impaired cerebral vasomotion in AD/CAA. Aβ and oxidative stress cause vascular tone dysregulation through direct effects on vascular cells, and indirect effects mediated by impaired neurovascular coupling. Vascular tone dysregulation is further aggravated by cholinergic deficit and results in depressed cerebrovascular reactivity and (possibly) impaired vasomotion, aggravating regional hypoperfusion and promoting further Aβ and oxidative stress accumulation.
Collapse
Affiliation(s)
- Luigi Yuri Di Marco
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Electronic and Electrical Engineering, University of Sheffield, Sheffield, UK
| | - Eszter Farkas
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Chris Martin
- Department of Psychology, University of Sheffield, Sheffield, UK
| | - Annalena Venneri
- Department of Neuroscience, University of Sheffield, Sheffield, UK.,IRCCS, Fondazione Ospedale S. Camillo, Venice, Italy
| | - Alejandro F Frangi
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Electronic and Electrical Engineering, University of Sheffield, Sheffield, UK
| |
Collapse
|
34
|
Minter MR, Taylor JM, Crack PJ. The contribution of neuroinflammation to amyloid toxicity in Alzheimer's disease. J Neurochem 2015; 136:457-74. [PMID: 26509334 DOI: 10.1111/jnc.13411] [Citation(s) in RCA: 317] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 10/11/2015] [Accepted: 10/22/2015] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and the most common cause of dementia. Deposition of amyloid-β (Aβ) remains a hallmark feature of the disease, yet the precise mechanism(s) by which this peptide induces neurotoxicity remain unknown. Neuroinflammation has long been implicated in AD pathology, yet its contribution to disease progression is still not understood. Recent evidence suggests that various Aβ complexes interact with microglial and astrocytic expressed pattern recognition receptors that initiate innate immunity. This process involves secretion of pro-inflammatory cytokines, chemokines and generation of reactive oxygen species that, in excess, drive a dysregulated immune response that contributes to neurodegeneration. The mechanisms by which a neuroinflammatory response can influence Aβ production, aggregation and eventual clearance are now becoming key areas where future therapeutic intervention may slow progression of AD. This review will focus on evidence supporting the combined neuroinflammatory-amyloid hypothesis for pathogenesis of AD, describing the key cell types, pathways and mediators involved. Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the leading cause of dementia worldwide. Deposition of intracellular plaques containing amyloid-beta (Aβ) is a hallmark proteinopathy of the disease yet the precise mechanisms by which this peptide induces neurotoxicity remains unknown. A neuroinflammatory response involving polarized microglial activity, enhanced astrocyte reactivity and elevated pro-inflammatory cytokine and chemokine load has long been implicated in AD and proposed to facilitate neurodegeneration. In this issue we discuss key receptor systems of innate immunity that detect Aβ, drive pro-inflammatory cytokine and chemokine production and influence Aβ aggregation and clearance. Evidence summarized in this review supports the combined neuroinflammatory-amyloid hypothesis for pathogenesis of AD and highlights the potential of immunomodulatory agents as potential future therapies for AD patients.
Collapse
Affiliation(s)
- Myles R Minter
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| | - Juliet M Taylor
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
35
|
Garwood CJ, Simpson JE, Al Mashhadi S, Axe C, Wilson S, Heath PR, Shaw PJ, Matthews FE, Brayne C, Ince PG, Wharton SB. DNA damage response and senescence in endothelial cells of human cerebral cortex and relation to Alzheimer's neuropathology progression: a population-based study in the Medical Research Council Cognitive Function and Ageing Study (MRC-CFAS) cohort. Neuropathol Appl Neurobiol 2015; 40:802-14. [PMID: 24861546 DOI: 10.1111/nan.12156] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 04/27/2014] [Indexed: 12/15/2022]
Abstract
AIMS Abnormalities of the brain microvasculature in Alzheimer's disease have led to the vascular hypothesis of the disease, which predicts that vascular changes precede neuronal dysfunction and degeneration. To determine the spectrum of endothelial injury in the elderly and its relation to Alzheimer-type neuropathology we investigated DNA damage in a population-based sample derived from the Medical Research Council Cognitive Function and Ageing Study. METHODS We examined endothelial damage in frontal and temporal cortex (n = 97) using immunohistochemistry for γH2AX and DNA-protein kinase (DNA-PKcs). To determine the effects of endothelial DNA damage at the earliest stages of Alzheimer's pathology we further focused our analysis on cases classified as Braak 0-II and examined endothelial senescence using histochemistry for β-galactosidase and the expression of genes related to DNA damage and senescence using quantitative polymerase chain reaction (qPCR). RESULTS We demonstrated large variation in endothelial DNA damage which was not associated with Alzheimer's neuropathology. Endothelial DNA-PKcs correlated with neuronal and glial DNA-PKcs counts. Focusing our further analysis on Braak 0-II cases, qPCR analysis demonstrated a trend to increased TP53 (P = 0.064) in cases with high compared with low endothelial DNA damage which was supported by immunohistochemical analysis of p53. Endothelial β-galactosidase expression was associated with increased neuronal (P = 0.033) and glial (P = 0.038), but not endothelial DNA-PKcs expression. CONCLUSIONS Damage to brain endothelial cells occurs early in relation to, or independently of, Alzheimer pathology, and parallels that in neurones and glia. Endothelial DNA damage and senescence are a brain ageing process that may contribute to dysfunction of the neurovascular unit in some elderly individuals.
Collapse
Affiliation(s)
- Claire J Garwood
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Zenaro E, Pietronigro E, Della Bianca V, Piacentino G, Marongiu L, Budui S, Turano E, Rossi B, Angiari S, Dusi S, Montresor A, Carlucci T, Nanì S, Tosadori G, Calciano L, Catalucci D, Berton G, Bonetti B, Constantin G. Neutrophils promote Alzheimer's disease-like pathology and cognitive decline via LFA-1 integrin. Nat Med 2015. [PMID: 26214837 DOI: 10.1038/nm.3913] [Citation(s) in RCA: 609] [Impact Index Per Article: 60.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammation is a pathological hallmark of Alzheimer's disease, and innate immune cells have been shown to contribute to disease pathogenesis. In two transgenic models of Alzheimer's disease (5xFAD and 3xTg-AD mice), neutrophils extravasated and were present in areas with amyloid-β (Aβ) deposits, where they released neutrophil extracellular traps (NETs) and IL-17. Aβ42 peptide triggered the LFA-1 integrin high-affinity state and rapid neutrophil adhesion to integrin ligands. In vivo, LFA-1 integrin controlled neutrophil extravasation into the CNS and intraparenchymal motility. In transgenic Alzheimer's disease models, neutrophil depletion or inhibition of neutrophil trafficking via LFA-1 blockade reduced Alzheimer's disease-like neuropathology and improved memory in mice already showing cognitive dysfunction. Temporary depletion of neutrophils for 1 month at early stages of disease led to sustained improvements in memory. Transgenic Alzheimer's disease model mice lacking LFA-1 were protected from cognitive decline and had reduced gliosis. In humans with Alzheimer's disease, neutrophils adhered to and spread inside brain venules and were present in the parenchyma, along with NETs. Our results demonstrate that neutrophils contribute to Alzheimer's disease pathogenesis and cognitive impairment and suggest that the inhibition of neutrophil trafficking may be beneficial in Alzheimer's disease.
Collapse
Affiliation(s)
- Elena Zenaro
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Enrica Pietronigro
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | | | - Gennj Piacentino
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Laura Marongiu
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Simona Budui
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Ermanna Turano
- Department of Neurological and Movement Sciences, Neurology Section, University of Verona, Verona, Italy
| | - Barbara Rossi
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Stefano Angiari
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Silvia Dusi
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Alessio Montresor
- 1] Department of Pathology and Diagnostics, University of Verona, Verona, Italy. [2] The Center for Biomedical Computing (CBMC), University of Verona, Verona, Italy
| | - Tommaso Carlucci
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Sara Nanì
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Gabriele Tosadori
- 1] Department of Pathology and Diagnostics, University of Verona, Verona, Italy. [2] The Center for Biomedical Computing (CBMC), University of Verona, Verona, Italy
| | - Lucia Calciano
- Department of Public Health and Community Medicine, University of Verona, Verona, Italy
| | - Daniele Catalucci
- National Research Council (CNR), Institute of Genetic and Biomedical Research (IRGB), and Humanitas Research Hospital, Milan, Italy
| | - Giorgio Berton
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Bruno Bonetti
- Department of Neurological and Movement Sciences, Neurology Section, University of Verona, Verona, Italy
| | - Gabriela Constantin
- 1] Department of Pathology and Diagnostics, University of Verona, Verona, Italy. [2] The Center for Biomedical Computing (CBMC), University of Verona, Verona, Italy
| |
Collapse
|
37
|
de Jager M, Boot MV, Bol JGJM, Brevé JJP, Jongenelen CAM, Drukarch B, Wilhelmus MMM. The blood clotting Factor XIIIa forms unique complexes with amyloid-beta (Aβ) and colocalizes with deposited Aβ in cerebral amyloid angiopathy. Neuropathol Appl Neurobiol 2015; 42:255-72. [PMID: 25871449 DOI: 10.1111/nan.12244] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 04/09/2015] [Indexed: 12/11/2022]
Abstract
AIMS Cerebral amyloid angiopathy (CAA) is a key pathological hallmark of Alzheimer's disease (AD) characterized by accumulation of amyloid-beta (Aβ) protein in blood vessel walls. CAA impairs vessel functioning, affects blood brain barrier integrity and accelerates cognitive decline of AD patients. Unfortunately, mechanisms underlying Aβ deposition in the vessel wall remain largely unknown. Factor XIIIa (FXIIIa) is a blood-derived transglutaminase crucial in blood coagulation by cross-linking fibrin molecules. Evidence is mounting that blood-derived factors are present in CAA and may play a role in protein deposition in the vessel wall. We therefore investigated whether FXIIIa is present in CAA and if FXIIIa cross-link activity affects Aβ aggregation. METHODS Using immunohistochemistry, we investigated the distribution of FXIIIa, its activator thrombin and in situ FXIIIa activity in CAA in post-mortem AD tissue. We used surface plasmon resonance and Western blot analysis to study binding of FXIIIa to Aβ and the formation of FXIIIa-Aβ complexes, respectively. In addition, we studied cytotoxicity of FXIIIa-Aβ complexes to cerebrovascular cells. RESULTS FXIIIa, thrombin and in situ FXIIIa activity colocalize with the Aβ deposition in CAA. Furthermore, FXIIIa binds to Aβ with a higher binding affinity for Aβ1-42 compared with Aβ1-40 . Moreover, highly stable FXIIIa-Aβ complexes are formed independently of FXIIIa cross-linking activity that protected cerebrovascular cells from Aβ-induced toxicity in vitro. CONCLUSIONS Our data showed that FXIIIa colocalizes with Aβ in CAA and that FXIIIa forms unique protein complexes with Aβ that might play an important role in Aβ deposition and persistence in the vessel wall.
Collapse
Affiliation(s)
- M de Jager
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - M V Boot
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - J G J M Bol
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - J J P Brevé
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - C A M Jongenelen
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - B Drukarch
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - M M M Wilhelmus
- Department of Anatomy and Neurosciences, Cellular Neuropharmacology Section, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
38
|
Combined Alzheimer's disease and cerebrovascular staging explains advanced dementia cognition. Alzheimers Dement 2015; 11:1358-66. [DOI: 10.1016/j.jalz.2015.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 12/15/2014] [Accepted: 01/30/2015] [Indexed: 01/07/2023]
|
39
|
Seals DR, Justice JN, LaRocca TJ. Physiological geroscience: targeting function to increase healthspan and achieve optimal longevity. J Physiol 2015; 594:2001-24. [PMID: 25639909 DOI: 10.1113/jphysiol.2014.282665] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/27/2015] [Indexed: 12/17/2022] Open
Abstract
Most nations of the world are undergoing rapid and dramatic population ageing, which presents great socio-economic challenges, as well as opportunities, for individuals, families, governments and societies. The prevailing biomedical strategy for reducing the healthcare impact of population ageing has been 'compression of morbidity' and, more recently, to increase healthspan, both of which seek to extend the healthy period of life and delay the development of chronic diseases and disability until a brief period at the end of life. Indeed, a recently established field within biological ageing research, 'geroscience', is focused on healthspan extension. Superimposed on this background are new attitudes and demand for 'optimal longevity' - living long, but with good health and quality of life. A key obstacle to achieving optimal longevity is the progressive decline in physiological function that occurs with ageing, which causes functional limitations (e.g. reduced mobility) and increases the risk of chronic diseases, disability and mortality. Current efforts to increase healthspan centre on slowing the fundamental biological processes of ageing such as inflammation/oxidative stress, increased senescence, mitochondrial dysfunction, impaired proteostasis and reduced stress resistance. We propose that optimization of physiological function throughout the lifespan should be a major emphasis of any contemporary biomedical policy addressing global ageing. Effective strategies should delay, reduce in magnitude or abolish reductions in function with ageing (primary prevention) and/or improve function or slow further declines in older adults with already impaired function (secondary prevention). Healthy lifestyle practices featuring regular physical activity and ideal energy intake/diet composition represent first-line function-preserving strategies, with pharmacological agents, including existing and new pharmaceuticals and novel 'nutraceutical' compounds, serving as potential complementary approaches. Future research efforts should focus on defining the temporal patterns of functional declines with ageing, identifying the underlying mechanisms and modulatory factors involved, and establishing the most effective lifestyle practices and pharmacological options for maintaining function. Continuing development of effective behavioural approaches for enhancing adherence to healthy ageing practices in diverse populations, and ongoing analysis of the socio-economic costs and benefits of healthspan extension will be important supporting goals. To meet the demands created by rapid population ageing, a new emphasis in physiological geroscience is needed, which will require the collaborative, interdisciplinary efforts of investigators working throughout the translational research continuum from basic science to public health.
Collapse
Affiliation(s)
- Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Jamie N Justice
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Thomas J LaRocca
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA
| |
Collapse
|
40
|
Mauro C, De Rosa V, Marelli-Berg F, Solito E. Metabolic syndrome and the immunological affair with the blood-brain barrier. Front Immunol 2015; 5:677. [PMID: 25601869 PMCID: PMC4283608 DOI: 10.3389/fimmu.2014.00677] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/16/2014] [Indexed: 12/29/2022] Open
Abstract
Epidemiological studies reveal an increased incidence of obesity worldwide, which is associated with increased prevalence and severity of cognitive disorders. The blood–brain barrier (BBB) represents the interface between the peripheral circulation and the brain, and plays a fundamental role in the cross-talk between these two compartments. The homeostatic function of the BBB is the protection of the brain from peripheral insult/inflammation. Alterations in the function of the BBB lead to pathologies of the central nervous system. Recently, metabolic imbalance has been shown to be an important risk factor associated with the decline of BBB integrity and function. This has direct etiological consequences on a variety of cerebrovascular and neurodegenerative pathologies with great impact to society. Priority areas for future preclinical research include strategies to improve clinicians’ ability to diagnose, prevent, and manage BBB abnormalities. In sharp contrast with epidemiological studies and clinical needs, little is known about the mechanisms that link metabolic syndrome to BBB functionality and cognitive disorders. Our view is that immune responses caused by metabolic stress might play a major role in this conundrum.
Collapse
Affiliation(s)
- Claudio Mauro
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London , London , UK
| | - Veronica De Rosa
- Istituto Per L'Endocrinologia e L'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (IEOS-CNR) , Naples , Italy
| | - Federica Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London , London , UK
| | - Egle Solito
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London , London , UK
| |
Collapse
|
41
|
Sohrabji F. Estrogen-IGF-1 interactions in neuroprotection: ischemic stroke as a case study. Front Neuroendocrinol 2015; 36:1-14. [PMID: 24882635 PMCID: PMC4247812 DOI: 10.1016/j.yfrne.2014.05.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 12/25/2022]
Abstract
The steroid hormone 17b-estradiol and the peptide hormone insulin-like growth factor (IGF)-1 independently exert neuroprotective actions in neurologic diseases such as stroke. Only a few studies have directly addressed the interaction between the two hormone systems, however, there is a large literature that indicates potentially greater interactions between the 17b-estradiol and IGF-1 systems. The present review focuses on key issues related to this interaction including IGF-1 and sex differences and common activation of second messenger systems. Using ischemic stroke as a case study, this review also focuses on independent and cooperative actions of estrogen and IGF-1 on neuroprotection, blood brain barrier integrity, angiogenesis, inflammation and post-stroke epilepsy. Finally, the review also focuses on the astrocyte, a key mediator of post stroke repair, as a local source of 17b-estradiol and IGF-1. This review thus highlights areas where significant new research is needed to clarify the interactions between these two neuroprotectants.
Collapse
Affiliation(s)
- Farida Sohrabji
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, TAMHSC College of Medicine, Bryan, TX 77807, United States.
| |
Collapse
|
42
|
van de Haar HJ, Burgmans S, Hofman PAM, Verhey FRJ, Jansen JFA, Backes WH. Blood-brain barrier impairment in dementia: current and future in vivo assessments. Neurosci Biobehav Rev 2014; 49:71-81. [PMID: 25524876 DOI: 10.1016/j.neubiorev.2014.11.022] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 10/20/2014] [Accepted: 11/21/2014] [Indexed: 01/14/2023]
Abstract
Increasing evidence indicates that blood-brain barrier (BBB) impairment may play a role in the pathophysiology of cognitive decline and dementia. In vivo imaging studies are needed to quantify and localize the BBB defects during life, contemplating the circulatory properties. We reviewed the literature for imaging studies investigating BBB impairment in patients suffering from dementia. After selection, 11 imaging studies were included, of which 6 used contrast-enhanced magnetic resonance imaging (MRI), 2 used contrast-enhanced computed tomography (CT), and 3 positron emission tomography (PET). Primarily the MRI studies hint at a subtle increasing permeability of the BBB, particularly in patients already exhibiting cerebrovascular pathology. More elaborate studies are required to provide convincing evidence on BBB impairment in patients with various stages of dementia with and without obvious cerebrovascular pathology. In the future, dynamic contrast enhanced MRI techniques and transport specific imaging using PET may further detail the research on the molecular nature of BBB defects.
Collapse
Affiliation(s)
- Harm J van de Haar
- School for Mental Health and Neuroscience, Maastricht University, Dr. Tanslaan 12, 6229 ET, Maastricht, The Netherlands; Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, Maastricht University Medical Center, Dr. Tanslaan 12, 6229 ET Maastricht, The Netherlands; Departments of Radiology and Nuclear Medicine, Maastricht University Medical Center, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Saartje Burgmans
- School for Mental Health and Neuroscience, Maastricht University, Dr. Tanslaan 12, 6229 ET, Maastricht, The Netherlands; Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, Maastricht University Medical Center, Dr. Tanslaan 12, 6229 ET Maastricht, The Netherlands
| | - Paul A M Hofman
- School for Mental Health and Neuroscience, Maastricht University, Dr. Tanslaan 12, 6229 ET, Maastricht, The Netherlands; Departments of Radiology and Nuclear Medicine, Maastricht University Medical Center, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Frans R J Verhey
- School for Mental Health and Neuroscience, Maastricht University, Dr. Tanslaan 12, 6229 ET, Maastricht, The Netherlands; Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, Maastricht University Medical Center, Dr. Tanslaan 12, 6229 ET Maastricht, The Netherlands
| | - Jacobus F A Jansen
- School for Mental Health and Neuroscience, Maastricht University, Dr. Tanslaan 12, 6229 ET, Maastricht, The Netherlands; Departments of Radiology and Nuclear Medicine, Maastricht University Medical Center, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Walter H Backes
- School for Mental Health and Neuroscience, Maastricht University, Dr. Tanslaan 12, 6229 ET, Maastricht, The Netherlands; Departments of Radiology and Nuclear Medicine, Maastricht University Medical Center, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands; CARIM School for Cardiovascular Disease, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands.
| |
Collapse
|
43
|
Avila-Muñoz E, Arias C. When astrocytes become harmful: functional and inflammatory responses that contribute to Alzheimer's disease. Ageing Res Rev 2014; 18:29-40. [PMID: 25078115 DOI: 10.1016/j.arr.2014.07.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 07/03/2014] [Accepted: 07/21/2014] [Indexed: 12/24/2022]
Abstract
A growing body of research suggests that astrocytes play roles as contributors to the pathophysiology of Alzheimer's disease (AD). Several lines of evidence propose that activated astrocytes produce and release proinflammatory molecules that may be critical for the generation of amyloid-β peptide (Aβ). However, accumulating evidence indicates that Aβ may activate astrocytes, which leads to an increase in cytokines that has been suggested to be a causative factor in the cognitive dysfunction of AD; thus, a vicious circle may be created. Intrinsic inflammatory mechanisms may provide a regulatory system that is capable of influencing the neuronal microenvironment that affects neuronal survival. In this article, we address the evidence surrounding the interactions of dysfunctional astrocytes with neighboring neurons that may initiate a cascade of events that culminates with neuronal injury and the expression of the hallmark lesions of AD. Comprehensive knowledge of the molecular mechanisms underlying the participation of astrocytes in neurodegeneration could aid the development of therapies to restore proper astrocyte function that can be used in AD patients to prevent or alleviate the progression of the disease in a more efficient and comprehensive manner.
Collapse
|
44
|
Drug Access to the Central Nervous System in Alzheimer’s Disease: Preclinical and Clinical Insights. Pharm Res 2014; 32:819-39. [DOI: 10.1007/s11095-014-1522-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/12/2014] [Indexed: 12/12/2022]
|
45
|
Aβ1-42 reduces P-glycoprotein in the blood-brain barrier through RAGE-NF-κB signaling. Cell Death Dis 2014; 5:e1299. [PMID: 24967961 PMCID: PMC4611731 DOI: 10.1038/cddis.2014.258] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 04/23/2014] [Accepted: 04/28/2014] [Indexed: 01/09/2023]
Abstract
The reduced clearance of amyloid-β peptide (Aβ) from the brain partly accounts for the neurotoxic accumulation of Aβ in Alzheimer's disease (AD). Recently, it has been suggested that P-glycoprotein (P-gp), which is an efflux transporter expressed on the luminal membrane of the brain capillary endothelium, is capable of transporting Aβ out of the brain. Although evidence has shown that restoring P-gp reduces brain Aβ in a mouse model of AD, the molecular mechanisms underlying the decrease in P-gp expression in AD is largely unknown. We found that Aβ1–42 reduced P-gp expression in the murine brain endothelial cell line bEnd.3, which was consistent with our in vivo data that P-gp expression was significantly reduced, especially near amyloid plaques in the brains of five familial AD mutations (5XFAD) mice that are used as an animal model for AD. A neutralizing antibody against the receptor for advanced glycation end products (RAGE) and an inhibitor of nuclear factor-kappa B (NF-κB) signaling prevented the decrease in Aβ1–42-induced P-gp expression, suggesting that Aβ reduced P-gp expression through NF-κB signaling by interacting with RAGE. In addition, we observed that the P-gp reduction by Aβ was rescued in bEnd.3 cells receiving inductive signals or factors from astrocytes making contacts with endothelial cells (ECs). These results support that alterations of astrocyte–EC contacts were closely associated with P-gp expression. This suggestion was further supported by the observation of a loss of astrocyte polarity in the brains of 5XFAD mice. Taken together, we found that P-gp downregulation by Aβ was mediated through RAGE–NF-κB signaling pathway in ECs and that the contact between astrocytes and ECs was an important factor in the regulation of P-gp expression.
Collapse
|
46
|
Abstract
The Aluminum Hypothesis, the idea that aluminum exposure is involved in the etiology of Alzheimer disease, dates back to a 1965 demonstration that aluminum causes neurofibrillary tangles in the brains of rabbits. Initially the focus of intensive research, the Aluminum Hypothesis has gradually been abandoned by most researchers. Yet, despite this current indifference, the Aluminum Hypothesis continues to attract the attention of a small group of scientists and aluminum continues to be viewed with concern by some of the public. This review article discusses reasons that mainstream science has largely abandoned the Aluminum Hypothesis and explores a possible reason for some in the general public continuing to view aluminum with mistrust.
Collapse
|
47
|
High-dose of vitamin C supplementation reduces amyloid plaque burden and ameliorates pathological changes in the brain of 5XFAD mice. Cell Death Dis 2014; 5:e1083. [PMID: 24577081 PMCID: PMC3944243 DOI: 10.1038/cddis.2014.26] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 12/22/2013] [Accepted: 01/07/2014] [Indexed: 11/20/2022]
Abstract
Blood–brain barrier (BBB) breakdown and mitochondrial dysfunction have been implicated in the pathogenesis of Alzheimer's disease (AD), a neurodegenerative disease characterized by cognitive deficits and neuronal loss. Besides vitamin C being as one of the important antioxidants, recently, it has also been reported as a modulator of BBB integrity and mitochondria morphology. Plasma levels of vitamin C are decreased in AD patients, which can affect disease progression. However, investigation using animal models on the role of vitamin C in the AD pathogenesis has been hampered because rodents produce with no dependence on external supply. Therefore, to identify the pathogenic importance of vitamin C in an AD mouse model, we cross-bred 5 familial Alzheimer's disease mutation (5XFAD) mice (AD mouse model) with ι-gulono-γ-lactone oxidase (Gulo) knockout (KO) mice, which are unable to synthesize their own vitamin C, and produced Gulo KO mice with 5XFAD mice background (KO-Tg). These mice were maintained on either low (0.66 g/l) or high (3.3 g/l) supplementation of vitamin C. We found that the higher supplementation of vitamin C had reduced amyloid plaque burden in the cortex and hippocampus in KO-Tg mice, resulting in amelioration of BBB disruption and mitochondrial alteration. These results suggest that intake of a larger amount of vitamin C could be protective against AD-like pathologies.
Collapse
|
48
|
Grimaldi LME, Zappalà G, Iemolo F, Castellano AE, Ruggieri S, Bruno G, Paolillo A. A pilot study on the use of interferon beta-1a in early Alzheimer's disease subjects. J Neuroinflammation 2014; 11:30. [PMID: 24524367 PMCID: PMC3931325 DOI: 10.1186/1742-2094-11-30] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 01/26/2014] [Indexed: 11/10/2022] Open
Abstract
Despite the fact that multiple sclerosis (MS) and Alzheimer’s disease (AD) share common neuroimmunological features, interferon beta 1a (IFNβ1a), the well-established treatment for the prevention of disease progression and cognitive decline in MS patients, has never been used in AD. We evaluated the safety and efficacy of IFNβ1a in subjects affected by mild-to-moderate AD in a double-blind, randomized, placebo-controlled, multicenter pilot study. Forty-two early Alzheimer’s patients were randomized to receive either a 22 mcg subcutaneous injection of IFNβ1a or placebo three times per week. A treatment period of 28 weeks was followed by 24 weeks of observation. IFNβ1a was well tolerated and adverse events were infrequent and mild to moderate. Although not statistically significant, a reduction in disease progression during follow-up was measured in IFNβ1a-treated patients by the Alzheimer’s Disease Assessment Scale cognitive subscale. Interestingly, the treatment group showed significant improvements in the Instrumental Activities of Daily Living and Physical Self-maintenance Scale. This study suggests that IFNβ1a is safe and well tolerated in early AD patients, and its possible beneficial role should be further investigated in larger studies.
Collapse
Affiliation(s)
- Luigi Maria Edoardo Grimaldi
- U,O, di Neurologia, Fondazione Istituto San Raffaele "G, Giglio" di Cefalù, Contrada Pietrapollastra, 90015 Cefalù, PA, Italy.
| | | | | | | | | | | | | |
Collapse
|
49
|
Park CH, Kim YS. Effects of Salviae Miltiorrhizae Radix on Blood-Brain Barrier Impairment of ICH-Induced Rats. ACTA ACUST UNITED AC 2014. [DOI: 10.6116/kjh.2014.29.1.19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
50
|
Chen X, Hui L, Geiger JD. Role of LDL cholesterol and endolysosomes in amyloidogenesis and Alzheimer's disease. ACTA ACUST UNITED AC 2014; 5. [PMID: 26413387 DOI: 10.4172/2155-9562.1000236] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The pathogenesis of late-onset sporadic Alzheimer's disease (AD) is believed to result from complex interactions between nutritional, environmental, epigenetic and genetic factors. Among those factors, altered circulating cholesterol homeostasis, independent of the APOE genotype, continues to be implicated in brain deposition of amyloid beta protein (Aβ) and the pathogenesis of AD. It is believed that trafficking of amyloid beta precursor protein (AβPP) into endolysosomes appears to play a critical role in determining amyloidogenic processing of AβPP because this is precisely where two enzymes critically important in AβPP metabolism are located; beta amyloid converting enzyme (BACE-1) and gamma secretase enzyme. We have shown that elevated levels of LDL cholesterol promote AβPP internalization, disturb neuronal endolysosome structure and function, and increase Aβ accumulation in neuronal endolysosomes. Here, we will further discuss the linkage between elevated levels of LDL cholesterol and AD pathogenesis, and explore the underlying mechanisms whereby elevated levels of plasma LDL cholesterol promote amyloidogenesis.
Collapse
Affiliation(s)
- Xuesong Chen
- Department of Basic Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Liang Hui
- Department of Basic Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Jonathan D Geiger
- Department of Basic Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203
| |
Collapse
|