151
|
Pistollato F, Iglesias RC, Ruiz R, Aparicio S, Crespo J, Lopez LD, Manna PP, Giampieri F, Battino M. Nutritional patterns associated with the maintenance of neurocognitive functions and the risk of dementia and Alzheimer’s disease: A focus on human studies. Pharmacol Res 2018; 131:32-43. [DOI: 10.1016/j.phrs.2018.03.012] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/15/2018] [Accepted: 03/15/2018] [Indexed: 11/25/2022]
|
152
|
Paik S, Somvanshi RK, Kumar U. Somatostatin Maintains Permeability and Integrity of Blood-Brain Barrier in β-Amyloid Induced Toxicity. Mol Neurobiol 2018; 56:292-306. [PMID: 29700775 DOI: 10.1007/s12035-018-1045-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 03/27/2018] [Indexed: 12/11/2022]
Abstract
In Alzheimer's disease (AD), the impaired clearance of β-amyloid peptide (Aβ) due to disrupted tight junction and transporter proteins is the prominent cause of disease progression. Somatostatin (SST) blocks the aggregation of Aβ and inflammation whereas reduction of SST levels in the CSF and brain tissue is associated with impaired cognitive function and memory loss. However, the role of SST in preservation of blood-brain barrier (BBB) integrity and functionality in Aβ-induced toxicity is not known. In the present study using human CMEC/D3 cells, we demonstrate that SST prevents Aβ-induced BBB permeability by regulating LRP1 and RAGE expression and improving the disrupted tight junction proteins. Furthermore, SST abrogates Aβ-induced JNK phosphorylation and expression of MMP2. Taken together, results presented here suggest that SST might serve as a therapeutic intervention in AD via targeting multiple pathways responsible for neurotoxicity, impaired BBB function, and disease progression.
Collapse
Affiliation(s)
- Seungil Paik
- Faculty of Pharmaceutical Sciences, The University of British Columbia, V6T1Z3, Vancouver, BC, Canada
| | - Rishi K Somvanshi
- Faculty of Pharmaceutical Sciences, The University of British Columbia, V6T1Z3, Vancouver, BC, Canada
| | - Ujendra Kumar
- Faculty of Pharmaceutical Sciences, The University of British Columbia, V6T1Z3, Vancouver, BC, Canada.
| |
Collapse
|
153
|
Etchegoyen M, Nobile MH, Baez F, Posesorski B, González J, Lago N, Milei J, Otero-Losada M. Metabolic Syndrome and Neuroprotection. Front Neurosci 2018; 12:196. [PMID: 29731703 PMCID: PMC5919958 DOI: 10.3389/fnins.2018.00196] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/12/2018] [Indexed: 12/17/2022] Open
Abstract
Introduction: Over the years the prevalence of metabolic syndrome (MetS) has drastically increased in developing countries as a major byproduct of industrialization. Many factors, such as the consumption of high-calorie diets and a sedentary lifestyle, bolster the spread of this disorder. Undoubtedly, the massive and still increasing incidence of MetS places this epidemic as an important public health issue. Hereon we revisit another outlook of MetS beyond its classical association with cardiovascular disease (CVD) and Diabetes Mellitus Type 2 (DM2), for MetS also poses a risk factor for the nervous tissue and threatens neuronal function. First, we revise a few essential concepts of MetS pathophysiology. Second, we explore some neuroprotective approaches in MetS pertaining brain hypoxia. The articles chosen for this review range from the years 1989 until 2017; the selection criteria was based on those providing data and exploratory information on MetS as well as those that studied innovative therapeutic approaches. Pathophysiology: The characteristically impaired metabolic pathways of MetS lead to hyperglycemia, insulin resistance (IR), inflammation, and hypoxia, all closely associated with an overall pro-oxidative status. Oxidative stress is well-known to cause the wreckage of cellular structures and tissue architecture. Alteration of the redox homeostasis and oxidative stress alter the macromolecular array of DNA, lipids, and proteins, in turn disrupting the biochemical pathways necessary for normal cell function. Neuroprotection: Different neuroprotective strategies are discussed involving lifestyle changes, medication aimed to mitigate MetS cardinal symptoms, and treatments targeted toward reducing oxidative stress. It is well-known that the routine practice of physical exercise, aerobic activity in particular, and a complete and well-balanced nutrition are key factors to prevent MetS. Nevertheless, pharmacological control of MetS as a whole and pertaining hypertension, dyslipidemia, and endothelial injury contribute to neuronal health improvement. Conclusion: The development of MetS has risen as a risk factor for neurological disorders. The therapeutic strategies include multidisciplinary approaches directed to address different pathological pathways all in concert.
Collapse
Affiliation(s)
- Melisa Etchegoyen
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Mariana H Nobile
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Francisco Baez
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Barbara Posesorski
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Julian González
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Néstor Lago
- Institute of Cardiovascular Pathophysiology, School of Medicine, University of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina
| | - José Milei
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
154
|
Ruan Q, Yu Z, Zhang W, Ruan J, Liu C, Zhang R. Cholinergic Hypofunction in Presbycusis-Related Tinnitus With Cognitive Function Impairment: Emerging Hypotheses. Front Aging Neurosci 2018; 10:98. [PMID: 29681847 PMCID: PMC5897739 DOI: 10.3389/fnagi.2018.00098] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 03/22/2018] [Indexed: 01/21/2023] Open
Abstract
Presbycusis (age-related hearing loss) is a potential risk factor for tinnitus and cognitive deterioration, which result in poor life quality. Presbycusis-related tinnitus with cognitive impairment is a common phenotype in the elderly population. In these individuals, the central auditory system shows similar pathophysiological alterations as those observed in Alzheimer's disease (AD), including cholinergic hypofunction, epileptiform-like network synchronization, chronic inflammation, and reduced GABAergic inhibition and neural plasticity. Observations from experimental rodent models indicate that recovery of cholinergic function can improve memory and other cognitive functions via acetylcholine-mediated GABAergic inhibition enhancement, nicotinic acetylcholine receptor (nAChR)-mediated anti-inflammation, glial activation inhibition and neurovascular protection. The loss of cholinergic innervation of various brain structures may provide a common link between tinnitus seen in presbycusis-related tinnitus and age-related cognitive impairment. We hypothesize a key component of the condition is the withdrawal of cholinergic input to a subtype of GABAergic inhibitory interneuron, neuropeptide Y (NPY) neurogliaform cells. Cholinergic denervation might not only cause the degeneration of NPY neurogliaform cells, but may also result in decreased AChR activation in GABAergic inhibitory interneurons. This, in turn, would lead to reduced GABA release and inhibitory regulation of neural networks. Reduced nAChR-mediated anti-inflammation due to the loss of nicotinic innervation might lead to the transformation of glial cells and release of inflammatory mediators, lowering the buffering of extracellular potassium and glutamate metabolism. Further research will provide evidence for the recovery of cholinergic function with the use of cholinergic input enhancement alone or in combination with other rehabilitative interventions to reestablish inhibitory regulation mechanisms of involved neural networks for presbycusis-related tinnitus with cognitive impairment.
Collapse
Affiliation(s)
- Qingwei Ruan
- Shanghai Institute of Geriatrics and Gerontology, Shanghai Key Laboratory of Clinical Geriatrics, Huadong Hospital, and Research Center of Aging and Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhuowei Yu
- Shanghai Institute of Geriatrics and Gerontology, Shanghai Key Laboratory of Clinical Geriatrics, Huadong Hospital, and Research Center of Aging and Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weibin Zhang
- Shanghai Institute of Geriatrics and Gerontology, Shanghai Key Laboratory of Clinical Geriatrics, Huadong Hospital, and Research Center of Aging and Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Ruan
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunhui Liu
- Department of Otolaryngology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruxin Zhang
- Department of Otolaryngology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
155
|
Van Skike CE, Jahrling JB, Olson AB, Sayre NL, Hussong SA, Ungvari Z, Lechleiter JD, Galvan V. Inhibition of mTOR protects the blood-brain barrier in models of Alzheimer's disease and vascular cognitive impairment. Am J Physiol Heart Circ Physiol 2018; 314:H693-H703. [PMID: 29351469 PMCID: PMC5966773 DOI: 10.1152/ajpheart.00570.2017] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/29/2017] [Accepted: 12/13/2017] [Indexed: 01/05/2023]
Abstract
An intact blood-brain barrier (BBB) limits entry of proinflammatory and neurotoxic blood-derived factors into the brain parenchyma. The BBB is damaged in Alzheimer's disease (AD), which contributes significantly to the progression of AD pathologies and cognitive decline. However, the mechanisms underlying BBB breakdown in AD remain elusive, and no interventions are available for treatment or prevention. We and others recently established that inhibition of the mammalian/mechanistic target of rapamycin (mTOR) pathway with rapamycin yields significant neuroprotective effects, improving cerebrovascular and cognitive function in mouse models of AD. To test whether mTOR inhibition protects the BBB in neurological diseases of aging, we treated hAPP(J20) mice modeling AD and low-density lipoprotein receptor-null (LDLR-/-) mice modeling vascular cognitive impairment with rapamycin. We found that inhibition of mTOR abrogates BBB breakdown in hAPP(J20) and LDLR-/- mice. Experiments using an in vitro BBB model indicated that mTOR attenuation preserves BBB integrity through upregulation of specific tight junction proteins and downregulation of matrix metalloproteinase-9 activity. Together, our data establish mTOR activity as a critical mediator of BBB breakdown in AD and, potentially, vascular cognitive impairment and suggest that rapamycin and/or rapalogs could be used for the restoration of BBB integrity. NEW & NOTEWORTHY This report establishes mammalian/mechanistic target of rapamycin as a critical mediator of blood-brain barrier breakdown in models of Alzheimer's disease and vascular cognitive impairment and suggests that drugs targeting the target of rapamycin pathway could be used for the restoration of blood-brain barrier integrity in disease states.
Collapse
MESH Headings
- Alzheimer Disease/drug therapy
- Alzheimer Disease/enzymology
- Alzheimer Disease/pathology
- Alzheimer Disease/psychology
- Animals
- Behavior, Animal
- Blood-Brain Barrier/drug effects
- Blood-Brain Barrier/enzymology
- Blood-Brain Barrier/pathology
- Cell Line
- Cognition
- Dementia, Vascular/drug therapy
- Dementia, Vascular/enzymology
- Dementia, Vascular/pathology
- Dementia, Vascular/psychology
- Disease Models, Animal
- Female
- Male
- Matrix Metalloproteinase 9/metabolism
- Mechanistic Target of Rapamycin Complex 1/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Protein Kinase Inhibitors/pharmacology
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Sirolimus/pharmacology
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/metabolism
- Tight Junction Proteins/metabolism
- Tight Junctions/drug effects
- Tight Junctions/enzymology
- Tight Junctions/pathology
Collapse
Affiliation(s)
- Candice E Van Skike
- Department of Cellular and Integrative Physiology and Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio , San Antonio, Texas
| | - Jordan B Jahrling
- Department of Cellular and Integrative Physiology and Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio , San Antonio, Texas
| | - Angela B Olson
- Department of Cellular and Integrative Physiology and Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio , San Antonio, Texas
| | - Naomi L Sayre
- Department of Neurosurgery, University of Texas Health San Antonio , San Antonio, Texas
- Department of Veterans Affairs, South Texas Veterans Health Care System, San Antonio, Texas
| | - Stacy A Hussong
- Department of Cellular and Integrative Physiology and Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio , San Antonio, Texas
- Department of Veterans Affairs, South Texas Veterans Health Care System, San Antonio, Texas
| | - Zoltan Ungvari
- Department of Geriatric Medicine and Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| | - James D Lechleiter
- Department of Cellular and Structural Biology, South Texas Research Facility Neuroscience Center, University of Texas Health San Antonio, San Antonio, Texas
| | - Veronica Galvan
- Department of Cellular and Integrative Physiology and Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio , San Antonio, Texas
- Department of Veterans Affairs, South Texas Veterans Health Care System, San Antonio, Texas
| |
Collapse
|
156
|
Monzón M, Hernández RS, Garcés M, Sarasa R, Badiola JJ. Glial alterations in human prion diseases: A correlative study of astroglia, reactive microglia, protein deposition, and neuropathological lesions. Medicine (Baltimore) 2018; 97:e0320. [PMID: 29642165 PMCID: PMC5908569 DOI: 10.1097/md.0000000000010320] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Neuroinflammation has recently been proposed to be a major component of neurodegenerative diseases. The aim of this study was to determine how the interaction between microglia and astroglia, which are the primary immune cell populations in the brain, and pathological prion protein (PrPsc) could influence the development and propagation of this neurodegenerative disease. Because a relevant role for glial response in prion disease has been clearly demonstrated in our previous studies using the natural animal model, a similar approach has been taken here using the natural human model. METHODS A morphological approach has been developed to analyze cerebellar samples from patients with Creutzfeldt-Jakob disease (CJD) in comparison with healthy control cases. Histopathological lesions were assessed, and PrPsc, glial fibrillary acidic protein (GFAP) and reactive microglia were immunolabelled by specific antibodies. Furthermore, co-location studies using confocal microscopy were performed to determine the possible relationships between both types of glial cells in all samples. RESULTS The results presented in this study support the involvement of both types of glial cells in CJD. Evidence of increased astrocyte and microglia reactivity can be observed in all CJD cases, and a close relationship between the types of glia is demonstrated by co-location studies. CONCLUSION Proteinopathies such as Alzheimer, Parkinson, and Huntington diseases, where aberrant proteins spread throughout the brain during disease progression, may share a molecular basis and mechanisms of propagation. Therefore, studies elucidating the interaction between gliosis and prion propagation may be relevant to these other neurodegenerative diseases and may provide new targets for therapeutic intervention.
Collapse
|
157
|
Huang Y, Miao Z, Hu Y, Yuan Y, Zhou Y, Wei L, Zhao K, Guo Q, Lu N. Baicalein reduces angiogenesis in the inflammatory microenvironment via inhibiting the expression of AP-1. Oncotarget 2018; 8:883-899. [PMID: 27903990 PMCID: PMC5352204 DOI: 10.18632/oncotarget.13669] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 11/12/2016] [Indexed: 01/20/2023] Open
Abstract
Increasing clinical and experimental studies have demonstrated that refractory chronic inflammation will result in malignant tumor and anti-angiogenic therapy may be an effective way to thwart the progression. Baicalein, one of the major active flavanoids found in Scutellaria baicalensis Georgi, has been exhibited potent anti-inflammation and anti-tumor effects by reducing angiogenesis. However, the exact mechanism of baicalein on endothelial cells in inflammatory microenvironment was not clear yet. Here, we investigated the anti-angiogenic effect of baicalein by incubating human umbilical vein endothelial cells (HUVECs) with THP-1 conditioned medium in vitro. The tube formation of HUVECs and microvessel outgrowth of rat aorta were attenuated, as well as the number of newly formed blood vessels in chicken chorioallantoic membrane (CAM) was reduced by baicalein. This anti-angiogenic effect was mainly on account of the inhibited motility, migration and invasion of HUVECs. In addition, mechanistic studies showed that baicalein could bind to AP-1 directly and the expression of c-Jun and c-Fos in HUVECs was reduced, accompanied by their increased proteasomal degradation. Besides, baicalein suppressed the nuclear translation, heterodimer formation and DNA binding affinity of c-Jun and c-Fos. What's more, the anti-angiogenic effect of baicalein was further confirmed by matrigel plug assay in vivo. Taken together, our study demonstrated that baicalein could exert its anti-angiogenic effect in the inflammation microenvironment via inhibiting the transcriptional activity of AP-1, which suggested that baicalein might be an alternative treatment against refractory chronic inflammation.
Collapse
Affiliation(s)
- Yujie Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Zhaorui Miao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yang Hu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yang Yuan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yuxin Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Libin Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Kai Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| |
Collapse
|
158
|
Rius-Pérez S, Tormos A, Pérez S, Taléns-Visconti R. Patología vascular: ¿causa o efecto en la enfermedad de Alzheimer? Neurologia 2018; 33:112-120. [DOI: 10.1016/j.nrl.2015.07.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 07/28/2015] [Indexed: 10/23/2022] Open
|
159
|
Rius-Pérez S, Tormos A, Pérez S, Taléns-Visconti R. Vascular pathology: Cause or effect in Alzheimer disease? NEUROLOGÍA (ENGLISH EDITION) 2018. [DOI: 10.1016/j.nrleng.2015.07.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
160
|
Zhan X, Stamova B, Sharp FR. Lipopolysaccharide Associates with Amyloid Plaques, Neurons and Oligodendrocytes in Alzheimer's Disease Brain: A Review. Front Aging Neurosci 2018. [PMID: 29520228 PMCID: PMC5827158 DOI: 10.3389/fnagi.2018.00042] [Citation(s) in RCA: 237] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
This review proposes that lipopolysaccharide (LPS, found in the wall of all Gram-negative bacteria) could play a role in causing sporadic Alzheimer’s disease (AD). This is based in part upon recent studies showing that: Gram-negative E. coli bacteria can form extracellular amyloid; bacterial-encoded 16S rRNA is present in all human brains with over 70% being Gram-negative bacteria; ultrastructural analyses have shown microbes in erythrocytes of AD patients; blood LPS levels in AD patients are 3-fold the levels in control; LPS combined with focal cerebral ischemia and hypoxia produced amyloid-like plaques and myelin injury in adult rat cortex. Moreover, Gram-negative bacterial LPS was found in aging control and AD brains, though LPS levels were much higher in AD brains. In addition, LPS co-localized with amyloid plaques, peri-vascular amyloid, neurons, and oligodendrocytes in AD brains. Based upon the postulate LPS caused oligodendrocyte injury, degraded Myelin Basic Protein (dMBP) levels were found to be much higher in AD compared to control brains. Immunofluorescence showed that the dMBP co-localized with β amyloid (Aβ) and LPS in amyloid plaques in AD brain, and dMBP and other myelin molecules were found in the walls of vesicles in periventricular White Matter (WM). These data led to the hypothesis that LPS acts on leukocyte and microglial TLR4-CD14/TLR2 receptors to produce NFkB mediated increases of cytokines which increase Aβ levels, damage oligodendrocytes and produce myelin injury found in AD brain. Since Aβ1–42 is also an agonist for TLR4 receptors, this could produce a vicious cycle that accounts for the relentless progression of AD. Thus, LPS, the TLR4 receptor complex, and Gram-negative bacteria might be treatment or prevention targets for sporadic AD.
Collapse
Affiliation(s)
- Xinhua Zhan
- Department of Neurology, MIND Institute, University of California, Davis, Davis, CA, United States
| | - Boryana Stamova
- Department of Neurology, MIND Institute, University of California, Davis, Davis, CA, United States
| | - Frank R Sharp
- Department of Neurology, MIND Institute, University of California, Davis, Davis, CA, United States
| |
Collapse
|
161
|
Eve DJ, Steiner G, Mahendrasah A, Sanberg PR, Kurien C, Thomson A, Borlongan CV, Garbuzova-Davis S. Reduction of microhemorrhages in the spinal cord of symptomatic ALS mice after intravenous human bone marrow stem cell transplantation accompanies repair of the blood-spinal cord barrier. Oncotarget 2018; 9:10621-10634. [PMID: 29535831 PMCID: PMC5828209 DOI: 10.18632/oncotarget.24360] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 01/20/2018] [Indexed: 12/13/2022] Open
Abstract
Blood-spinal cord barrier (BSCB) alterations, including capillary rupture, have been demonstrated in animal models of amyotrophic lateral sclerosis (ALS) and ALS patients. To date, treatment to restore BSCB in ALS is underexplored. Here, we evaluated whether intravenous transplantation of human bone marrow CD34+ (hBM34+) cells into symptomatic ALS mice leads to restoration of capillary integrity in the spinal cord as determined by detection of microhemorrhages. Three different doses of hBM34+ cells (5 × 104, 5 × 105 or 1 × 106) or media were intravenously injected into symptomatic G93A SOD1 mice at 13 weeks of age. Microhemorrhages were determined in the cervical and lumbar spinal cords of mice at 4 weeks post-treatment, as revealed by Perls' Prussian blue staining for ferric iron. Numerous microhemorrhages were observed in the gray and white matter of the spinal cords in media-treated mice, with a greater number of capillary ruptures within the ventral horn of both segments. In cell-treated mice, microhemorrhage numbers in the cervical and lumbar spinal cords were inversely related to administered cell doses. In particular, the pervasive microvascular ruptures determined in the spinal cords in late symptomatic ALS mice were significantly decreased by the highest cell dose, suggestive of BSCB repair by grafted hBM34+ cells. The study results provide translational outcomes supporting transplantation of hBM34+ cells at an optimal dose as a potential therapeutic strategy for BSCB repair in ALS patients.
Collapse
Affiliation(s)
- David J. Eve
- Center of Excellence for Aging and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - George Steiner
- Center of Excellence for Aging and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Ajay Mahendrasah
- Center of Excellence for Aging and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Paul R. Sanberg
- Center of Excellence for Aging and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
- Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
- Department of Psychiatry, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Crupa Kurien
- Center of Excellence for Aging and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Avery Thomson
- Center of Excellence for Aging and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Svitlana Garbuzova-Davis
- Center of Excellence for Aging and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
- Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
- Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
162
|
Ariga T. The Pathogenic Role of Ganglioside Metabolism in Alzheimer's Disease-Cholinergic Neuron-Specific Gangliosides and Neurogenesis. Mol Neurobiol 2018; 54:623-638. [PMID: 26748510 DOI: 10.1007/s12035-015-9641-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia with clinical symptoms that include deficits in memory, judgment, thinking, and behavior. Gangliosides are present on the outer surface of plasma membranes and are especially abundant in the nervous tissues of vertebrates. Ganglioside metabolism, especially the cholinergic neuron-specific gangliosides, GQ1bα and GT1aα, is altered in mouse model of AD and patients with AD. Thus, alterations in ganglioside metabolism may participate in several events related to the pathogenesis of AD. Increased expressions of GT1aα may reflect cholinergic neurogenesis. Most changes in ganglioside metabolism occur in the specific brain areas and their lipid rafts. Targeting ganglioside metabolism in lipid rafts may represent an underexploited opportunity to design novel therapeutic strategies for AD.
Collapse
Affiliation(s)
- Toshio Ariga
- Department of Neuroscience and Regenerative Medicine, Institute of Neuroscience, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA. .,Department of Materials and Applied Chemistry, College of Science and Technology, Nihon University, Chiyoda-ku, Tokyo, 101-8308, Japan.
| |
Collapse
|
163
|
Gauberti M, Fournier AP, Docagne F, Vivien D, Martinez de Lizarrondo S. Molecular Magnetic Resonance Imaging of Endothelial Activation in the Central Nervous System. Theranostics 2018; 8:1195-1212. [PMID: 29507614 PMCID: PMC5835930 DOI: 10.7150/thno.22662] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 01/12/2018] [Indexed: 01/01/2023] Open
Abstract
Endothelial cells of the central nervous system over-express surface proteins during neurological disorders, either as a cause, or a consequence, of the disease. Since the cerebral vasculature is easily accessible by large contrast-carrying particles, it constitutes a target of choice for molecular magnetic resonance imaging (MRI). In this review, we highlight the most recent advances in molecular MRI of brain endothelial activation and focus on the development of micro-sized particles of iron oxide (MPIO) targeting adhesion molecules including intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), P-Selectin and E-Selectin. We also discuss the perspectives and challenges for the clinical application of this technology in neurovascular disorders (ischemic stroke, intracranial hemorrhage, subarachnoid hemorrhage, diabetes mellitus), neuroinflammatory disorders (multiple sclerosis, brain infectious diseases, sepsis), neurodegenerative disorders (Alzheimer's disease, vascular dementia, aging) and brain cancers (primitive neoplasms, metastasis).
Collapse
Affiliation(s)
- Maxime Gauberti
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging for Neurological Disorders (PhIND), Cyceron, 14000 Caen, France
- CHU Caen, Department of diagnostic imaging and interventional radiology, CHU de Caen Côte de Nacre, Caen, France
| | - Antoine P. Fournier
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging for Neurological Disorders (PhIND), Cyceron, 14000 Caen, France
| | - Fabian Docagne
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging for Neurological Disorders (PhIND), Cyceron, 14000 Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging for Neurological Disorders (PhIND), Cyceron, 14000 Caen, France
- CHU Caen, Clinical Research Department, CHU de Caen Côte de Nacre, Caen, France
| | - Sara Martinez de Lizarrondo
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging for Neurological Disorders (PhIND), Cyceron, 14000 Caen, France
| |
Collapse
|
164
|
Momtaz S, Hassani S, Khan F, Ziaee M, Abdollahi M. Cinnamon, a promising prospect towards Alzheimer's disease. Pharmacol Res 2017; 130:241-258. [PMID: 29258915 DOI: 10.1016/j.phrs.2017.12.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/10/2017] [Accepted: 12/10/2017] [Indexed: 12/25/2022]
Abstract
Over the last decades, an exponential increase of efforts concerning the treatment of Alzheimer's disease (AD) has been practiced. Phytochemicals preparations have a millenary background to combat various pathological conditions. Various cinnamon species and their biologically active ingredients have renewed the interest towards the treatment of patients with mild-to-moderate AD through the inhibition of tau protein aggregation and prevention of the formation and accumulation of amyloid-β peptides into the neurotoxic oligomeric inclusions, both of which are considered to be the AD trademarks. In this review, we presented comprehensive data on the interactions of a number of cinnamon polyphenols (PPs) with oxidative stress and pro-inflammatory signaling pathways in the brain. In addition, we discussed the potential association between AD and diabetes mellitus (DM), vis-à-vis the effluence of cinnamon PPs. Further, an upcoming prospect of AD epigenetic pathophysiological conditions and cinnamon has been sighted. Data was retrieved from the scientific databases such as PubMed database of the National Library of Medicine, Scopus and Google Scholar without any time limitation. The extract of cinnamon efficiently inhibits tau accumulations, Aβ aggregation and toxicity in vivo and in vitro models. Indeed, cinnamon possesses neuroprotective effects interfering multiple oxidative stress and pro-inflammatory pathways. Besides, cinnamon modulates endothelial functions and attenuates the vascular cell adhesion molecules. Cinnamon PPs may induce AD epigenetic modifications. Cinnamon and in particular, cinnamaldehyde seem to be effective and safe approaches for treatment and prevention of AD onset and/or progression. However, further molecular and translational research studies as well as prolonged clinical trials are required to establish the therapeutic safety and efficacy in different cinnamon spp.
Collapse
Affiliation(s)
- Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran; Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shokoufeh Hassani
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fazlullah Khan
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; International Campus, Tehran University of Medical Sciences (IC-TUMS), Tehran Iran
| | - Mojtaba Ziaee
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran; Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; International Campus, Tehran University of Medical Sciences (IC-TUMS), Tehran Iran.
| |
Collapse
|
165
|
Dhull DK, Kumar A. Tramadol ameliorates behavioural, biochemical, mitochondrial and histological alterations in ICV-STZ-induced sporadic dementia of Alzheimer's type in rats. Inflammopharmacology 2017; 26:925-938. [PMID: 29249049 DOI: 10.1007/s10787-017-0431-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 12/06/2017] [Indexed: 12/23/2022]
Abstract
Alzheimer disease represents a major public health issue with limited therapeutic interventions. We explored the possibility of therapeutic approach by repurposing of tramadol in a sporadic animal model of Alzheimer's type. Streptozocin (STZ 3 mg/kg; bilaterally) was injected to male SD rats through intracerebroventricular (ICV) route. Drug treatment was started just after streptozocin administration and continued for 3 weeks. The rats were killed on the 21st day following the last behavioral test, and cytoplasmic fractions of the hippocampus and pre-frontal cortex were prepared for the quantification of acetylcholinesterase, oxidative stress parameter, mitochondrial enzymes activity and histological examination. Tramadol (5, 10 and 20 mg/kg, i.p.) was used as a treatment drug, and memantine (10 mg/kg, i.p.) was used as a standard. Tramadol significantly attenuated behavioral, biochemical, mitochondrial and histological alterations at low (5 mg/kg) and intermediate (10 mg/kg) dose, suggesting its neuroprotective potential in ICV-STZ-treated rats. Further, the neuroprotective effect of tramadol (10 mg/kg) was comparable to memantine (10 mg/kg). In conclusion, our results indicate the effectiveness of tramadol in preventing ICV-STZ-induced cognitive impairment as well as mito-oxidative stress. Further, these findings reveal the possibility of MOR agonist as a therapeutic approach for sporadic Alzheimer disease.
Collapse
Affiliation(s)
- Dinesh K Dhull
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
166
|
A Cross-Sectional Analysis of Late-Life Cardiovascular Factors and Their Relation to Clinically Defined Neurodegenerative Diseases. Alzheimer Dis Assoc Disord 2017; 30:223-9. [PMID: 26756386 DOI: 10.1097/wad.0000000000000138] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Studies have demonstrated associations between cardiovascular factors and Alzheimer disease (AD) with minimal focus on other neurodegenerative diseases. Utilizing cross-sectional data from 17,532 individuals in the National Alzheimer's Coordinating Center, Uniform Data Set, we compared the presence of cardiovascular factors [body mass index (BMI), atrial fibrillation, hypertension, hyperlipidemia, and diabetes] in individuals carrying a diagnosis of Probable AD (ProbAD), Possible AD, vascular dementia, dementia with Lewy bodies (DLB), frontotemporal dementia, Parkinson disease, progressive supranuclear palsy, or corticobasal degeneration, with that of normals. Generalized linear mixed models were fitted with age at visit, gender, and cardiovascular factors as fixed effects and Alzheimer's Disease Centers as random effects. In late life, only BMI of ProbAD and DLB patients was statistically significantly lower than that in normals (P-values <0.001). When accounting for colinearity within cardiovascular factors, a low BMI was a comorbidity of certain dementia etiologies as compared with normals. These data support a concept of disease-specific associations with certain cardiovascular factors.
Collapse
|
167
|
de Wit NM, Vanmol J, Kamermans A, Hendriks JJA, de Vries HE. Inflammation at the blood-brain barrier: The role of liver X receptors. Neurobiol Dis 2017; 107:57-65. [DOI: 10.1016/j.nbd.2016.09.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 09/11/2016] [Accepted: 09/17/2016] [Indexed: 02/05/2023] Open
|
168
|
Yamazaki Y, Kanekiyo T. Blood-Brain Barrier Dysfunction and the Pathogenesis of Alzheimer's Disease. Int J Mol Sci 2017; 18:ijms18091965. [PMID: 28902142 PMCID: PMC5618614 DOI: 10.3390/ijms18091965] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/06/2017] [Accepted: 09/07/2017] [Indexed: 01/22/2023] Open
Abstract
Brain capillary endothelial cells form the blood-brain barrier (BBB), which is covered with basement membranes and is also surrounded by pericytes and astrocyte end-feet in the neurovascular unit. The BBB tightly regulates the molecular exchange between the blood flow and brain parenchyma, thereby regulating the homeostasis of the central nervous system (CNS). Thus, dysfunction of the BBB is likely involved in the pathogenesis of several neurological diseases, including Alzheimer’s disease (AD). While amyloid-β (Aβ) deposition and neurofibrillary tangle formation in the brain are central pathological hallmarks in AD, cerebrovascular lesions and BBB alteration have also been shown to frequently coexist. Although further clinical studies should clarify whether BBB disruption is a specific feature of AD pathogenesis, increasing evidence indicates that each component of the neurovascular unit is significantly affected in the presence of AD-related pathologies in animal models and human patients. Conversely, since some portions of Aβ are eliminated along the neurovascular unit and across the BBB, disturbing the pathways may result in exacerbated Aβ accumulation in the brain. Thus, current evidence suggests that BBB dysfunction may causatively and consequently contribute to AD pathogenesis, forming a vicious cycle between brain Aβ accumulation and neurovascular unit impairments during disease progression.
Collapse
Affiliation(s)
- Yu Yamazaki
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| |
Collapse
|
169
|
Robert J, Button EB, Stukas S, Boyce GK, Gibbs E, Cowan CM, Gilmour M, Cheng WH, Soo SK, Yuen B, Bahrabadi A, Kang K, Kulic I, Francis G, Cashman N, Wellington CL. High-density lipoproteins suppress Aβ-induced PBMC adhesion to human endothelial cells in bioengineered vessels and in monoculture. Mol Neurodegener 2017; 12:60. [PMID: 28830501 PMCID: PMC5568306 DOI: 10.1186/s13024-017-0201-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/07/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Alzheimer's Disease (AD), characterized by accumulation of beta-amyloid (Aβ) plaques in the brain, can be caused by age-related failures to clear Aβ from the brain through pathways that involve the cerebrovasculature. Vascular risk factors are known to increase AD risk, but less is known about potential protective factors. We hypothesize that high-density lipoproteins (HDL) may protect against AD, as HDL have vasoprotective properties that are well described for peripheral vessels. Epidemiological studies suggest that HDL is associated with reduced AD risk, and animal model studies support a beneficial role for HDL in selectively reducing cerebrovascular amyloid deposition and neuroinflammation. However, the mechanism by which HDL may protect the cerebrovascular endothelium in the context of AD is not understood. METHODS We used peripheral blood mononuclear cell adhesion assays in both a highly novel three dimensional (3D) biomimetic model of the human vasculature composed of primary human endothelial cells (EC) and smooth muscle cells cultured under flow conditions, as well as in monolayer cultures of ECs, to study how HDL protects ECs from the detrimental effects of Aβ. RESULTS Following Aβ addition to the abluminal (brain) side of the vessel, we demonstrate that HDL circulated within the lumen attenuates monocyte adhesion to ECs in this biofidelic vascular model. The mechanism by which HDL suppresses Aβ-mediated monocyte adhesion to ECs was investigated using monotypic EC cultures. We show that HDL reduces Aβ-induced PBMC adhesion to ECs independent of nitric oxide (NO) production, miR-233 and changes in adhesion molecule expression. Rather, HDL acts through scavenger receptor (SR)-BI to block Aβ uptake into ECs and, in cell-free assays, can maintain Aβ in a soluble state. We confirm the role of SR-BI in our bioengineered human vessel. CONCLUSION Our results define a novel activity of HDL that suppresses Aβ-mediated monocyte adhesion to the cerebrovascular endothelium.
Collapse
Affiliation(s)
- Jérôme Robert
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
| | - Emily B. Button
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
| | - Sophie Stukas
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
| | - Guilaine K. Boyce
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
| | - Ebrima Gibbs
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
- Department of Neurology, University of British Columbia, Vancouver, BC V6T 2B5 Canada
| | - Catherine M. Cowan
- Department of Zoology, University of British Columbia, Vancouver, BC V6T 2B5 Canada
| | - Megan Gilmour
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
| | - Wai Hang Cheng
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
| | - Sonja K. Soo
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
| | - Brian Yuen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
| | - Arvin Bahrabadi
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
| | - Kevin Kang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
| | - Iva Kulic
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
| | - Gordon Francis
- Department of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6 Canada
| | - Neil Cashman
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
- Department of Neurology, University of British Columbia, Vancouver, BC V6T 2B5 Canada
| | - Cheryl L. Wellington
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3 Canada
| |
Collapse
|
170
|
Jiang L, Hu M, Lu Y, Cao Y, Chang Y, Dai Z. The protective effects of dexmedetomidine on ischemic brain injury: A meta-analysis. J Clin Anesth 2017. [DOI: 10.1016/j.jclinane.2017.04.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
171
|
Polymicrobial Infections In Brain Tissue From Alzheimer's Disease Patients. Sci Rep 2017; 7:5559. [PMID: 28717130 PMCID: PMC5514053 DOI: 10.1038/s41598-017-05903-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/05/2017] [Indexed: 01/10/2023] Open
Abstract
Several studies have advanced the idea that the etiology of Alzheimer’s disease (AD) could be microbial in origin. In the present study, we tested the possibility that polymicrobial infections exist in tissue from the entorhinal cortex/hippocampus region of patients with AD using immunohistochemistry (confocal laser scanning microscopy) and highly sensitive (nested) PCR. We found no evidence for expression of early (ICP0) or late (ICP5) proteins of herpes simplex virus type 1 (HSV-1) in brain sections. A polyclonal antibody against Borrelia detected structures that appeared not related to spirochetes, but rather to fungi. These structures were not found with a monoclonal antibody. Also, Borrelia DNA was undetectable by nested PCR in the ten patients analyzed. By contrast, two independent Chlamydophila antibodies revealed several structures that resembled fungal cells and hyphae, and prokaryotic cells, but most probably were unrelated to Chlamydophila spp. Finally, several structures that could belong to fungi or prokaryotes were detected using peptidoglycan and Clostridium antibodies, and PCR analysis revealed the presence of several bacteria in frozen brain tissue from AD patients. Thus, our results show that polymicrobial infections consisting of fungi and bacteria can be revealed in brain tissue from AD patients.
Collapse
|
172
|
Basavarajappa BS, Shivakumar M, Joshi V, Subbanna S. Endocannabinoid system in neurodegenerative disorders. J Neurochem 2017; 142:624-648. [PMID: 28608560 DOI: 10.1111/jnc.14098] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/22/2017] [Accepted: 06/02/2017] [Indexed: 12/19/2022]
Abstract
Most neurodegenerative disorders (NDDs) are characterized by cognitive impairment and other neurological defects. The definite cause of and pathways underlying the progression of these NDDs are not well-defined. Several mechanisms have been proposed to contribute to the development of NDDs. These mechanisms may proceed concurrently or successively, and they differ among cell types at different developmental stages in distinct brain regions. The endocannabinoid system, which involves cannabinoid receptors type 1 (CB1R) and type 2 (CB2R), endogenous cannabinoids and the enzymes that catabolize these compounds, has been shown to contribute to the development of NDDs in several animal models and human studies. In this review, we discuss the functions of the endocannabinoid system in NDDs and converse the therapeutic efficacy of targeting the endocannabinoid system to rescue NDDs.
Collapse
Affiliation(s)
- Balapal S Basavarajappa
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, USA.,New York State Psychiatric Institute, New York City, New York, USA.,Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York City, New York, USA.,Department of Psychiatry, New York University Langone Medical Center, New York City, New York, USA
| | - Madhu Shivakumar
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, USA
| | - Vikram Joshi
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, USA
| | - Shivakumar Subbanna
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, USA
| |
Collapse
|
173
|
Endothelial cell-oligodendrocyte interactions in small vessel disease and aging. Clin Sci (Lond) 2017; 131:369-379. [PMID: 28202749 PMCID: PMC5310718 DOI: 10.1042/cs20160618] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/28/2016] [Accepted: 12/14/2016] [Indexed: 12/11/2022]
Abstract
Cerebral small vessel disease (SVD) is a prevalent, neurological disease that significantly increases the risk of stroke and dementia. The main pathological changes are vascular, in the form of lipohyalinosis and arteriosclerosis, and in the white matter (WM), in the form of WM lesions. Despite this, it is unclear to what extent the key cell types involved–the endothelial cells (ECs) of the vasculature and the oligodendrocytes of the WM–interact. Here, we describe the work that has so far been carried out suggesting an interaction between ECs and oligodendrocytes in SVD. As these interactions have been studied in more detail in other disease states and in development, we explore these systems and discuss the role these mechanisms may play in SVD.
Collapse
|
174
|
|
175
|
Angiotensin IV Receptors Mediate the Cognitive and Cerebrovascular Benefits of Losartan in a Mouse Model of Alzheimer's Disease. J Neurosci 2017; 37:5562-5573. [PMID: 28476949 DOI: 10.1523/jneurosci.0329-17.2017] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/30/2017] [Accepted: 04/22/2017] [Indexed: 12/21/2022] Open
Abstract
The use of angiotensin receptor blockers (ARBs) correlates with reduced onset and progression of Alzheimer's disease (AD). The mechanism depicting how ARBs such as losartan restore cerebrovascular and cognitive deficits in AD is unknown. Here, we propose a mechanism underlying losartan's benefits by selectively blocking the effects of angiotensin IV (AngIV) at its receptor (AT4R) with divalinal in mice overexpressing the AD-related Swedish and Indiana mutations of the human amyloid precursor protein (APP mice) and WT mice. Young (3-month-old) mice were treated with losartan (∼10 mg/kg/d, 4 months), followed by intracerebroventricular administration of vehicle or divalinal in the final month of treatment. Spatial learning and memory were assessed using Morris water mazes at 3 and 4 months of losartan treatment. Cerebrovascular reactivity and whisker-evoked neurovascular coupling responses were measured at end point (∼7 months of age), together with biomarkers related to neuronal and vascular oxidative stress (superoxide dismutase-2), neuroinflammation (astroglial and microglial activation), neurogenesis (BrdU-labeled newborn cells), and amyloidosis [soluble amyloid-β (Aβ) species and Aβ plaque load]. Divalinal countered losartan's capacity to rescue spatial learning and memory and blocked losartan's benefits on dilatory function and baseline nitric oxide bioavailability. Divalinal reverted losartan's anti-inflammatory effects, but failed to modify losartan-mediated reductions in oxidative stress. Neither losartan nor divalinal affected arterial blood pressure or significantly altered the amyloid pathology in APP mice. Our findings identify activation of the AngIV/AT4R cascade as the underlying mechanism in losartan's benefits and a target that could restore Aβ-related cognitive and cerebrovascular deficits in AD.SIGNIFICANCE STATEMENT Antihypertensive medications that target the renin angiotensin system, such as angiotensin receptor blockers (ARBs), have been associated with lower incidence and progression of Alzheimer's disease (AD) in cohort studies. However, the manner by which ARBs mediate their beneficial effects is unknown. Here, the angiotensin IV receptor (AT4R) was identified as mediating the cognitive and cerebrovascular rescue of losartan, a commonly prescribed ARB, in a mouse model of AD. The AT4R was further implicated in mediating anti-inflammatory benefits. AT4R-mediated effects were independent from changes in blood pressure, amyloidosis, and oxidative stress. Overall, our results implicate the angiotensin IV/AT4R cascade as a promising candidate for AD intervention.
Collapse
|
176
|
Peripheral complement interactions with amyloid β peptide: Erythrocyte clearance mechanisms. Alzheimers Dement 2017; 13:1397-1409. [PMID: 28475854 DOI: 10.1016/j.jalz.2017.03.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 02/28/2017] [Accepted: 03/27/2017] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Although amyloid β peptide (Aβ) is cleared from the brain to cerebrospinal fluid and the peripheral circulation, mechanisms for its removal from blood remain unresolved. Primates have uniquely evolved a highly effective peripheral clearance mechanism for pathogens, immune adherence, in which erythrocyte complement receptor 1 (CR1) plays a major role. METHODS Multidisciplinary methods were used to demonstrate immune adherence capture of Aβ by erythrocytes and its deficiency in Alzheimer's disease (AD). RESULTS Aβ was shown to be subject to immune adherence at every step in the pathway. Aβ dose-dependently activated serum complement. Complement-opsonized Aβ was captured by erythrocytes via CR1. Erythrocytes, Aβ, and hepatic Kupffer cells were colocalized in the human liver. Significant deficits in erythrocyte Aβ levels were found in AD and mild cognitive impairment patients. DISCUSSION CR1 polymorphisms elevate AD risk, and >80% of human CR1 is vested in erythrocytes to subserve immune adherence. The present results suggest that this pathway is pathophysiologically relevant in AD.
Collapse
|
177
|
Markers of neuroinflammation associated with Alzheimer's disease pathology in older adults. Brain Behav Immun 2017; 62:203-211. [PMID: 28161476 DOI: 10.1016/j.bbi.2017.01.020] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/15/2017] [Accepted: 01/30/2017] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND In vitro and animal studies have linked neuroinflammation to Alzheimer's disease (AD) pathology. Studies on markers of inflammation in subjects with mild cognitive impairment or AD dementia provided inconsistent results. We hypothesized that distinct blood and cerebrospinal fluid (CSF) inflammatory markers are associated with biomarkers of amyloid and tau pathology in older adults without cognitive impairment or with beginning cognitive decline. OBJECTIVE To identify blood-based and CSF neuroinflammation marker signatures associated with AD pathology (i.e. an AD CSF biomarker profile) and to investigate associations of inflammation markers with CSF biomarkers of amyloid, tau pathology, and neuronal injury. DESIGN/METHODS Cross-sectional analysis was performed on data from 120 older community-dwelling adults with normal cognition (n=48) or with cognitive impairment (n=72). CSF Aβ1-42, tau and p-tau181, and a panel of 37 neuroinflammatory markers in both CSF and serum were quantified. Least absolute shrinkage and selection operator (LASSO) regression was applied to determine a reference model that best predicts an AD CSF biomarker profile defined a priori as p-tau181/Aβ1-42 ratio >0.0779. It was then compared to a second model that included the inflammatory markers from either serum or CSF. In addition, the correlations between inflammatory markers and CSF Aβ1-42, tau and p-tau181 levels were assessed. RESULTS Forty-two subjects met criteria for having an AD CSF biomarker profile. The best predictive models included 8 serum or 3 CSF neuroinflammatory markers related to cytokine mediated inflammation, vascular injury, and angiogenesis. Both models improved the accuracy to predict an AD biomarker profile when compared to the reference model. In analyses separately performed in the subgroup of participants with cognitive impairment, adding the serum or the CSF neuroinflammation markers also improved the accuracy of the diagnosis of AD pathology. None of the inflammatory markers correlated with the CSF Aβ1-42 levels. Six CSF markers (IL-15, MCP-1, VEGFR-1, sICAM1, sVCAM-1, and VEGF-D) correlated with the CSF tau and p-tau181 levels, and these associations remained significant after controlling for age, sex, cognitive impairment, and APOEε4 status. CONCLUSIONS The identified serum and CSF neuroinflammation biomarker signatures improve the accuracy of classification for AD pathology in older adults. Our results suggest that inflammation, vascular injury, and angiogenesis as reflected by CSF markers are closely related to cerebral tau pathology.
Collapse
|
178
|
Role of thrombin-PAR1-PKCθ/δ axis in brain pericytes in thrombin-induced MMP-9 production and blood-brain barrier dysfunction in vitro. Neuroscience 2017; 350:146-157. [PMID: 28344073 DOI: 10.1016/j.neuroscience.2017.03.026] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 03/09/2017] [Accepted: 03/14/2017] [Indexed: 02/08/2023]
Abstract
Thrombin, an essential component in the coagulation cascade, participates in the pathogenesis of brain diseases, such as ischemic stroke, intracerebral hemorrhage, Alzheimer's disease and Parkinson's disease through blood-brain barrier (BBB) dysfunction. It is thought that the thrombin-matrix metalloproteinase (MMP)-9 axis is an important process in the pathogenesis of neurovascular disease, such as BBB dysfunction. We recently reported that brain pericytes are the most MMP-9-releasing cells in response to thrombin stimulation among the BBB-constituting cells. This thrombin-induced MMP-9 release is partially due to protease-activated receptor (PAR1), one of the specific thrombin receptors. Then, we evaluated the intracellular signaling pathways involved in MMP-9 release and the contribution of thrombin-reactive brain pericytes to BBB dysfunction. PKC activator evoked MMP-9 release from brain pericytes. The thrombin-induced MMP-9 release was inhibited by U0126, LY294002, Go6976, and Go6983. However, Go6976 decreased phosphorylation levels of PKCθ and Akt, and Go6983 decreased phosphorylation levels of PKCδ and extracellular signal-regulated kinase (ERK). Additionally, treatment of pericytes with thrombin or PAR1-activating peptide stimulated PKCδ/θ signaling. These substances impaired brain endothelial barrier function in the presence of brain pericytes. Brain pericytes function through two independent downstream signaling pathways via PAR1 activation to release MMP-9 in response to thrombin - the PKCθ-Akt pathway and the PKCδ-ERK1/2 pathway. These pathways participate in PAR1-mediated MMP-9 release from pericytes, which leads to BBB dysfunction. Brain pericytes and their specific signaling pathways could provide novel therapeutic targets for thrombin-induced neurovascular diseases.
Collapse
|
179
|
Bagyinszky E, Giau VV, Shim K, Suk K, An SSA, Kim S. Role of inflammatory molecules in the Alzheimer's disease progression and diagnosis. J Neurol Sci 2017; 376:242-254. [PMID: 28431620 DOI: 10.1016/j.jns.2017.03.031] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/14/2017] [Accepted: 03/20/2017] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is a complex disorder and the most common form of neurodegenerative dementia. Several genetic, environmental, and physiological factors, including inflammations and metabolic influences, are involved in the progression of AD. Inflammations are composed of complicated networks of many chemokines and cytokines with diverse cells. Inflammatory molecules are needed for the protection against pathogens, and maintaining their balances is important for normal physiological function. Recent studies demonstrated that inflammation may be involved in neurodegenerative dementia. Cellular immune components, such as microglia or astrocytes, mediate the release of inflammatory molecules, including tumor necrosis factor, growth factors, adhesion molecules, or chemokines. Over- and underexpression of pro- and anti-inflammatory molecules, respectively, may result in neuroinflammation and thus disease initiation and progression. In addition, levels of several inflammatory factors were reported to be altered in the brain or bodily fluids of patients with AD, reflecting their neuropathological changes. Therefore, simultaneous detection of several inflammatory molecules in the early or pre-symptomatic stage may improve the early diagnosis of AD. Further studies are needed to determine, how induction or inhibition of inflammatory factors could be used for AD therapies. This review summarizes the role or possible role of immune cells and inflammatory molecules in disease progression or prevention.
Collapse
Affiliation(s)
- Eva Bagyinszky
- Department of Bionano Technology, Gachon University, Gyeonggi-do, Republic of Korea
| | - Vo Van Giau
- Department of Bionano Technology, Gachon University, Gyeonggi-do, Republic of Korea
| | - Kyuhwan Shim
- Department of Bionano Technology, Gachon University, Gyeonggi-do, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Seong Soo A An
- Department of Bionano Technology, Gachon University, Gyeonggi-do, Republic of Korea.
| | - SangYun Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Gyeonggi-do, Republic of Korea
| |
Collapse
|
180
|
Baldacci F, Lista S, Cavedo E, Bonuccelli U, Hampel H. Diagnostic function of the neuroinflammatory biomarker YKL-40 in Alzheimer’s disease and other neurodegenerative diseases. Expert Rev Proteomics 2017; 14:285-299. [DOI: 10.1080/14789450.2017.1304217] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- AXA Research Fund UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du cerveau et de la moelle (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Simone Lista
- AXA Research Fund UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du cerveau et de la moelle (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Enrica Cavedo
- AXA Research Fund UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du cerveau et de la moelle (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
- IRCCS Istituto Centro San Giovanni di Dio-Fatebenefratelli, Brescia, Italy
| | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Harald Hampel
- AXA Research Fund UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du cerveau et de la moelle (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| |
Collapse
|
181
|
Cai W, Zhang K, Li P, Zhu L, Xu J, Yang B, Hu X, Lu Z, Chen J. Dysfunction of the neurovascular unit in ischemic stroke and neurodegenerative diseases: An aging effect. Ageing Res Rev 2017; 34:77-87. [PMID: 27697546 PMCID: PMC5384332 DOI: 10.1016/j.arr.2016.09.006] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/15/2016] [Accepted: 09/26/2016] [Indexed: 12/23/2022]
Abstract
Current understanding on the mechanisms of brain injury and neurodegeneration highlights an appreciation of multicellular interactions within the neurovascular unit (NVU), which include the evolution of blood-brain barrier (BBB) damage, neuronal cell death or degeneration, glial reaction, and immune cell infiltration. Aging is an important factor that influences the integrity of the NVU. The age-related physiological or pathological changes in the cellular components of the NVU have been shown to increase the vulnerability of the NVU to ischemia/reperfusion injury or neurodegeneration, and to result in deteriorated brain damage. This review describes the impacts of aging on each NVU component and discusses the mechanisms by which aging increases NVU sensitivity to stroke and neurodegenerative diseases. Prophylactic or therapeutic perspectives that may delay or diminish aging and thus prevent the incidence of these neurological disorders will also be reviewed.
Collapse
Affiliation(s)
- Wei Cai
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA; Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, China; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Kai Zhang
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Peiying Li
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Ling Zhu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA; Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jing Xu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Boyu Yang
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Xiaoming Hu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Zhengqi Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, China.
| | - Jun Chen
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
182
|
Lourenço CF, Ledo A, Barbosa RM, Laranjinha J. Neurovascular uncoupling in the triple transgenic model of Alzheimer's disease: Impaired cerebral blood flow response to neuronal-derived nitric oxide signaling. Exp Neurol 2017; 291:36-43. [PMID: 28161255 DOI: 10.1016/j.expneurol.2017.01.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/05/2017] [Accepted: 01/27/2017] [Indexed: 11/16/2022]
Abstract
Nitric oxide (NO)-dependent pathways and cerebrovascular dysfunction have been shown to contribute to the cognitive decline and neurodegeneration observed in Alzheimer's disease (AD) but whether they represent initial factors or later changes of the disease is still a matter of debate. In this work, we aimed at investigating whether and to what extent neuronal-derived NO signaling and related neurovascular coupling are impaired along aging in the hippocampus of the triple transgenic mouse model of Alzheimer's Disease (3xTg-AD). We performed a longitudinal study combining behavior studies, in vivo simultaneous measurements of NO concentration gradients and cerebral blood flow (CBF), along with detection of NO synthase (NOS) and markers of nitroxidative stress. Our results revealed an impairment in the neurovascular coupling along aging in the 3xTg-AD mice which preceded obvious cognitive decline. This impairment was characterized by diminished CBF changes in response to normal or even increased NO signals and associated with markers of nitroxidative stress. The results suggest that impairment in neurovascular coupling is primarily due to cerebrovascular dysfunction, rather than due to dysfunctional NO signaling from neurons to blood vessels. Overall, this work supports cerebrovascular dysfunction as a fundamental underlying process in AD pathology.
Collapse
Affiliation(s)
- Cátia F Lourenço
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal.
| | - Ana Ledo
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Rui M Barbosa
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - João Laranjinha
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
183
|
Malek-Ahmadi M, Lu S, Chan Y, Perez SE, Chen K, Mufson EJ. Cognitive Domain Dispersion Association with Alzheimer's Disease Pathology. J Alzheimers Dis 2017; 58:575-583. [PMID: 28453479 PMCID: PMC6314665 DOI: 10.3233/jad-161233] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Within neuropsychology, the term dispersion refers to the degree of variation in performance between different cognitive domains for an individual. Previous studies have demonstrated that cognitively normal individuals with higher dispersion are at an increased risk for progressing to mild cognitive impairment (MCI) and Alzheimer's disease (AD). Therefore, we determined 1) whether increased dispersion in older adults was associated with amyloid plaques and neurofibrillary tangles (NFTs) and 2) whether increased cognitive dispersion accurately differentiated MCI and AD from non-cognitively impaired (NCI) individuals. The intra-subject standard deviation (ISD) was used to quantify cognitive dispersion, and receiver operator characteristic (ROC) analysis determined whether ISD differentiated MCI and AD from NCI. Neuropathological scores for diffuse plaques (DPs), neuritic plaques (NPs), and NFTs were used as outcome measures in a series of negative binomial regression models. Regression analyses found that increased ISD was associated with increased NFT pathology (β= 10.93, SE = 3.82, p = 0.004), but not with DPs (β= 1.33, SE = 8.85, p = 0.88) or NPs (β= 14.64, SE = 8.45, p = 0.08) after adjusting for age at death, gender, education, APOE ɛ4 status, and clinical diagnosis. An interaction term of ISD with age at death also showed a significant negative association (β= -0.13, SE = 0.04, p = 0.004), revealing an age-dependent association between ISD with NFTs. The ISD failed to show an acceptable level of diagnostic accuracy for MCI (AUC = 0.60). These findings suggest that increased cognitive dispersion is related to NFT pathology where age significantly affects this association.
Collapse
Affiliation(s)
| | - Sophie Lu
- Williams College, Williamstown, MA, USA
| | | | - Sylvia E. Perez
- Department of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Kewei Chen
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
| | - Elliott J. Mufson
- Department of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
184
|
Bhat SA, Goel R, Shukla R, Hanif K. Platelet CD40L induces activation of astrocytes and microglia in hypertension. Brain Behav Immun 2017; 59:173-189. [PMID: 27658543 DOI: 10.1016/j.bbi.2016.09.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 09/16/2016] [Accepted: 09/17/2016] [Indexed: 12/20/2022] Open
Abstract
Studies have demonstrated separately that hypertension is associated with platelet activation in the periphery (resulting in accumulation and localized inflammatory response) and glial activation in the brain. We investigated the contribution of platelets in brain inflammation, particularly glial activation in vitro and in a rat model of hypertension. We found that HTN increased the expression of adhesion molecules like JAM-1, ICAM-1, and VCAM-1 on brain endothelium and resulted in the deposition of platelets in the brain. Platelet deposition in hypertensive rats was associated with augmented CD40 and CD40L and activation of astrocytes (GFAP expression) and microglia (Iba-1 expression) in the brain. Platelets isolated from hypertensive rats had significantly higher sCD40L levels and induced more prominent glial activation than platelets from normotensive rats. Activation of platelets with ADP induced sCD40L release and activation of astrocytes and microglia. Moreover, CD40L induced glial (astrocytes and microglia) activation, NFкB and MAPK inflammatory signaling, culminating in neuroinflammation and neuronal injury (increased apoptotic cells). Importantly, injection of ADP-activated platelets into normotensive rats strongly induced activation of astrocytes and microglia and increased plasma sCD40L levels compared with control platelets. On the contrary, inhibition of platelet activation by Clopidogrel or disruption of CD40 signaling prevented astrocyte and microglial activation and provided neuroprotection in both in vivo and in vitro conditions. Thus, we have identified platelet CD40L as a key inflammatory molecule for the induction of astrocyte and microglia activation, the major contributors to inflammation-mediated injury in the brain.
Collapse
Affiliation(s)
- Shahnawaz Ali Bhat
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., India
| | - Ruby Goel
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., India
| | - Rakesh Shukla
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., India
| | - Kashif Hanif
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., India; National Institute of Pharmaceutical Education and Research, Rae Bareli, India.
| |
Collapse
|
185
|
Chi LM, Wang X, Nan GX. In silico analyses for molecular genetic mechanism and candidate genes in patients with Alzheimer's disease. Acta Neurol Belg 2016; 116:543-547. [PMID: 26935318 DOI: 10.1007/s13760-016-0613-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/29/2016] [Indexed: 12/14/2022]
Abstract
This study aimed to identify candidate genes and explore the molecular pathogenesis of Alzheimer's disease (AD). Exon microarray data composed by of three human entorhinal cortex samples of AD patients and three non-demented controls (NDC) were analyzed, then expression profile data were preprocessed with the Oligo package and differentially expressed genes (DEGs) were identified by limma package in R/Bioconductor. In addition, protein-protein interaction (PPI) network was predicted and constructed using the STRING database. Finally, gene ontology (GO)-biological processes (BP) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways enriched by DEGs were recognized. A total of 124 up-regulated and 218 down-regulated genes were identified. TGF-beta-activated kinase 1/MAP3K7 binding protein 2 (TAB 2) and chromogranin B (secretogranin 1) (CHGB) were the significantly up- and down-regulated genes, respectively. In addition, DEGs of DnaJ (Hsp40) homolog, subfamily B, member 1 (DNAJB1) and heat shock 70 kDa protein 1A (HSPA1A) were in the up-regulated network, while synaptophysin (SYP) and somatostatin (SST) were in the down-regulated network. Furthermore, the up-regulated genes were enriched in GO-BP terms of protein stimulus, unfolding and organic substance, etc., and pathways of ECM-receptor interaction, etc. The down-regulated genes were mainly associated with nerve-related transmission and neuroactive substances transportation. Protein folding abnormality and altered synaptic transmission could have a synergistic effect on the pathomechanism of AD. DEGs including DNAJB1 and HSPA1A may be involved in both the processes, while CHGB, SYP and SST may be important for the regulation of synaptic transmission to contribute to the progress and development of AD.
Collapse
Affiliation(s)
- Lu-Mei Chi
- Department of Neurology, China-Japan Union Hospital of Jilin University, No 126 Xintai Street, Changchun, 130000, Jilin, China
| | - Xu Wang
- Cancer Center, First Affiliated Hospital, Jilin University, Changchun, 130061, Jilin, China
| | - Guang-Xian Nan
- Department of Neurology, China-Japan Union Hospital of Jilin University, No 126 Xintai Street, Changchun, 130000, Jilin, China.
| |
Collapse
|
186
|
Di Giovanni G, Svob Strac D, Sole M, Unzeta M, Tipton KF, Mück-Šeler D, Bolea I, Della Corte L, Nikolac Perkovic M, Pivac N, Smolders IJ, Stasiak A, Fogel WA, De Deurwaerdère P. Monoaminergic and Histaminergic Strategies and Treatments in Brain Diseases. Front Neurosci 2016; 10:541. [PMID: 27932945 PMCID: PMC5121249 DOI: 10.3389/fnins.2016.00541] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 11/07/2016] [Indexed: 12/18/2022] Open
Abstract
The monoaminergic systems are the target of several drugs for the treatment of mood, motor and cognitive disorders as well as neurological conditions. In most cases, advances have occurred through serendipity, except for Parkinson's disease where the pathophysiology led almost immediately to the introduction of dopamine restoring agents. Extensive neuropharmacological studies first showed that the primary target of antipsychotics, antidepressants, and anxiolytic drugs were specific components of the monoaminergic systems. Later, some dramatic side effects associated with older medicines were shown to disappear with new chemical compounds targeting the origin of the therapeutic benefit more specifically. The increased knowledge regarding the function and interaction of the monoaminergic systems in the brain resulting from in vivo neurochemical and neurophysiological studies indicated new monoaminergic targets that could achieve the efficacy of the older medicines with fewer side-effects. Yet, this accumulated knowledge regarding monoamines did not produce valuable strategies for diseases where no monoaminergic drug has been shown to be effective. Here, we emphasize the new therapeutic and monoaminergic-based strategies for the treatment of psychiatric diseases. We will consider three main groups of diseases, based on the evidence of monoamines involvement (schizophrenia, depression, obesity), the identification of monoamines in the diseases processes (Parkinson's disease, addiction) and the prospect of the involvement of monoaminergic mechanisms (epilepsy, Alzheimer's disease, stroke). In most cases, the clinically available monoaminergic drugs induce widespread modifications of amine tone or excitability through neurobiological networks and exemplify the overlap between therapeutic approaches to psychiatric and neurological conditions. More recent developments that have resulted in improved drug specificity and responses will be discussed in this review.
Collapse
Affiliation(s)
| | | | - Montse Sole
- Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Institut de Neurociències, Universitat Autònoma de BarcelonaBarcelona, Spain
| | - Mercedes Unzeta
- Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Institut de Neurociències, Universitat Autònoma de BarcelonaBarcelona, Spain
| | - Keith F. Tipton
- School of Biochemistry and Immunology, Trinity College DublinDublin, Ireland
| | - Dorotea Mück-Šeler
- Division of Molecular Medicine, Rudjer Boskovic InstituteZagreb, Croatia
| | - Irene Bolea
- Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Institut de Neurociències, Universitat Autònoma de BarcelonaBarcelona, Spain
| | | | | | - Nela Pivac
- Division of Molecular Medicine, Rudjer Boskovic InstituteZagreb, Croatia
| | - Ilse J. Smolders
- Department of Pharmaceutical Chemistry and Drug Analysis, Vrije Universiteit BrusselBrussels, Belgium
| | - Anna Stasiak
- Department of Hormone Biochemistry, Medical University of LodzLodz, Poland
| | - Wieslawa A. Fogel
- Department of Hormone Biochemistry, Medical University of LodzLodz, Poland
| | - Philippe De Deurwaerdère
- Centre National de la Recherche Scientifique (Unité Mixte de Recherche 5293), Institut of Neurodegenerative DiseasesBordeaux Cedex, France
| |
Collapse
|
187
|
Reduced Cerebral Perfusion Pressure during Lung Transplant Surgery Is Associated with Risk, Duration, and Severity of Postoperative Delirium. Ann Am Thorac Soc 2016; 13:180-7. [PMID: 26731642 DOI: 10.1513/annalsats.201507-454oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
RATIONALE Delirium is common following lung transplant and is associated with poorer clinical outcomes. The extent to which intraoperative hemodynamic alterations may contribute to postoperative delirium among lung transplant recipients has not been examined. OBJECTIVES To examine the impact of intraoperative hemodynamic changes on neurobehavioral outcomes among lung transplant recipients. METHODS Intraoperative hemodynamic function during lung transplant was assessed in a consecutive series of patients between March and November 2013. Intraoperative cerebral perfusion pressure was assessed every minute in all patients. Following lung transplant, patients were monitored for the presence and severity of delirium using the Confusion Assessment Method and the Delirium Rating Scale until hospital discharge. MEASUREMENTS AND MAIN RESULTS Sixty-three patients received lung transplants, of whom 23 (37%) subsequently developed delirium. Lower cerebral perfusion pressure was associated with increased risk of delirium (odds ratio [OR], 2.08 per 10-mm Hg decrease; 95% confidence interval [CI], 1.02-4.24; P = 0.043), longer duration of delirium (OR, 1.7 d longer per 10-mm Hg decrease; 95% CI, 1.1-2.7; P = 0.022), and greater delirium severity (b = -0.81; 95% CI, -1.47 to -0.15; P = 0.017). CONCLUSIONS Poorer cerebral perfusion pressure during lung transplant is associated with greater risk for delirium following transplant, as well as greater duration and severity of delirium, independent of demographic and medical predictors.
Collapse
|
188
|
Daugherty AM, Raz N. A virtual water maze revisited: Two-year changes in navigation performance and their neural correlates in healthy adults. Neuroimage 2016; 146:492-506. [PMID: 27659539 DOI: 10.1016/j.neuroimage.2016.09.044] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 09/12/2016] [Accepted: 09/17/2016] [Indexed: 11/30/2022] Open
Abstract
Age-related declines in spatial navigation are associated with deficits in procedural and episodic memory and deterioration of their neural substrates. For the lack of longitudinal evidence, the pace and magnitude of these declines and their neural mediators remain unclear. Here we examined virtual navigation in healthy adults (N=213, age 18-77 years) tested twice, two years apart, with complementary indices of navigation performance (path length and complexity) measured over six learning trials at each occasion. Slopes of skill acquisition curves and longitudinal change therein were estimated in structural equation modeling, together with change in regional brain volumes and iron content (R2* relaxometry). Although performance on the first trial did not differ between occasions separated by two years, the slope of path length improvement over trials was shallower and end-of-session performance worse at follow-up. Advanced age, higher pulse pressure, smaller cerebellar and caudate volumes, and greater caudate iron content were associated with longer search paths, i.e. poorer navigation performance. In contrast, path complexity diminished faster over trials at follow-up, albeit less so in older adults. Improvement in path complexity after two years was predicted by lower baseline hippocampal iron content and larger parahippocampal volume. Thus, navigation path length behaves as an index of perceptual-motor skill that is vulnerable to age-related decline, whereas path complexity may reflect cognitive mapping in episodic memory that improves with repeated testing, although not enough to overcome age-related deficits.
Collapse
Affiliation(s)
- Ana M Daugherty
- Institute of Gerontology, Wayne State University, Detroit, MI, USA; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Champaign, IL, USA.
| | - Naftali Raz
- Institute of Gerontology, Wayne State University, Detroit, MI, USA; Department of Psychology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
189
|
Navarro-Dorado J, Villalba N, Prieto D, Brera B, Martín-Moreno AM, Tejerina T, de Ceballos ML. Vascular Dysfunction in a Transgenic Model of Alzheimer's Disease: Effects of CB1R and CB2R Cannabinoid Agonists. Front Neurosci 2016; 10:422. [PMID: 27695396 PMCID: PMC5025475 DOI: 10.3389/fnins.2016.00422] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/29/2016] [Indexed: 01/21/2023] Open
Abstract
There is evidence of altered vascular function, including cerebrovascular, in Alzheimer's disease (AD) and transgenic models of the disease. Indeed vasoconstrictor responses are increased, while vasodilation is reduced in both conditions. β-Amyloid (Aβ) appears to be responsible, at least in part, of alterations in vascular function. Cannabinoids, neuroprotective and anti-inflammatory agents, induce vasodilation both in vivo and in vitro. We have demonstrated a beneficial effect of cannabinoids in models of AD by preventing glial activation. In this work we have studied the effects of these compounds on vessel density in amyloid precursor protein (APP) transgenic mice, line 2576, and on altered vascular responses in aortae isolated ring. First we showed increased collagen IV positive vessels in AD brain compared to control subjects, with a similar increase in TgAPP mice, which was normalized by prolonged oral treatment with the CB1/CB2 mixed agonist WIN 55,212-2 (WIN) and the CB2 selective agonist JWH-133 (JWH). In Tg APP mice the vasoconstriction induced by phenylephrine and the thromboxane agonist U46619 was significantly increased, and no change in the vasodilation to acetylcholine (ACh) was observed. Tg APP displayed decreased vasodilation to both cannabinoid agonists, which were able to prevent decreased ACh relaxation in the presence of Aβ. In summary, we have confirmed and extended the existence of altered vascular responses in Tg APP mice. Moreover, our results suggest that treatment with cannabinoids may ameliorate the vascular responses in AD-type pathology.
Collapse
Affiliation(s)
- Jorge Navarro-Dorado
- Department of Pharmacology, School of Medicine, Complutense University of Madrid Madrid, Spain
| | - Nuria Villalba
- Department of Physiology, Faculty of Pharmacy, Complutense University of Madrid Madrid, Spain
| | - Dolores Prieto
- Department of Physiology, Faculty of Pharmacy, Complutense University of Madrid Madrid, Spain
| | - Begoña Brera
- Neurodegeneration Group, Cellular, Molecular and Developmental Neurobiology and CIBERNED, Cajal Institute, CSIC Madrid, Spain
| | - Ana M Martín-Moreno
- Neurodegeneration Group, Cellular, Molecular and Developmental Neurobiology and CIBERNED, Cajal Institute, CSIC Madrid, Spain
| | - Teresa Tejerina
- Department of Pharmacology, School of Medicine, Complutense University of Madrid Madrid, Spain
| | - María L de Ceballos
- Neurodegeneration Group, Cellular, Molecular and Developmental Neurobiology and CIBERNED, Cajal Institute, CSIC Madrid, Spain
| |
Collapse
|
190
|
Devassy JG, Leng S, Gabbs M, Monirujjaman M, Aukema HM. Omega-3 Polyunsaturated Fatty Acids and Oxylipins in Neuroinflammation and Management of Alzheimer Disease. Adv Nutr 2016; 7:905-16. [PMID: 27633106 PMCID: PMC5015035 DOI: 10.3945/an.116.012187] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Alzheimer disease (AD) is becoming one of the most prevalent neurodegenerative conditions worldwide. Although the disease progression is becoming better understood, current medical interventions can only ameliorate some of the symptoms but cannot slow disease progression. Neuroinflammation plays an important role in the advancement of this disorder, and n-3 (ω-3) polyunsaturated fatty acids (PUFAs) are involved in both the reduction in and resolution of inflammation. These effects may be mediated by the anti-inflammatory and proresolving effects of bioactive lipid mediators (oxylipins) derived from n-3 PUFAs [eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)] in fish oil. Although interventions have generally used fish oil containing both EPA and DHA, several studies that used either EPA or DHA alone or specific oxylipins derived from these fatty acids indicate that they have distinct effects. Both DHA and EPA can reduce neuroinflammation and cognitive decline, but EPA positively influences mood disorders, whereas DHA maintains normal brain structure. Fewer studies with a plant-derived n-3 PUFA, α-linolenic acid, suggest that other n-3 PUFAs and their oxylipins also may positively affect AD. Further research identifying the unique anti-inflammatory and proresolving properties of oxylipins from individual n-3 PUFAs will enable the discovery of novel disease-management strategies in AD.
Collapse
Affiliation(s)
| | | | | | | | - Harold M Aukema
- Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Canada; and Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, Canada
| |
Collapse
|
191
|
Hutter-Schmid B, Humpel C. Alpha-Smooth Muscle Actin mRNA and Protein Are Increased in Isolated Brain Vessel Extracts of Alzheimer Mice. Pharmacology 2016; 98:251-260. [PMID: 27463512 DOI: 10.1159/000448007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 06/24/2016] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disorder of the brain, characterized by extracellular beta-amyloid (Aβ) plaques, intracellular tau pathology, neurodegeneration and inflammation. There is clear evidence that the blood-brain barrier is damaged in AD and that vessel function is impaired. Alpha-smooth muscle actin (αSMA) is a prominent protein expressed on brain vessels, especially in cells located closer to the arteriole end of the capillaries, which possibly influences the blood vessel contraction. The aim of the present study was to observe αSMA protein and mRNA expression in isolated brain vessel extracts and cortex in an Alzheimer mouse model with strong Aβ plaque deposition. Our data revealed a prominent expression of αSMA protein in isolated brain vessel extracts of AD mice by Western blot analysis. Immunostaining showed that these vessels were associated with Aβ plaques. Quantitative real-time PCR analysis confirmed this increase at the mRNA expression level and showed a significant increase of transforming growth factor beta-1 mRNA expression in AD mice. In situ hybridization demonstrated a strong expression pattern of αSMA mRNA in the whole cortex and hippocampus. In conclusion, our data provide evidence that αSMA protein and mRNA are enhanced in vessels in an AD mouse model, possibly counteracting vessel malfunction in AD.
Collapse
Affiliation(s)
- Bianca Hutter-Schmid
- Department of Psychiatry, Psychotherapy and Psychosomatics, Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
192
|
Malek-Ahmadi M, Perez SE, Chen K, Mufson EJ. Neuritic and Diffuse Plaque Associations with Memory in Non-Cognitively Impaired Elderly. J Alzheimers Dis 2016; 53:1641-52. [PMID: 27540968 PMCID: PMC6314669 DOI: 10.3233/jad-160365] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The presence of Alzheimer's disease (AD)-related neuropathology among cognitively normal individuals has been well documented. It has been proposed that these individuals may represent a pre-clinical AD population. Previous studies have demonstrated a negative association between the presence of both amyloid-β (Aβ) plaques and neurofibrillary tangles with ante-mortem cognitive performance, a relationship which is likely influenced by a number of factors including age and APOE ɛ4 carrier status. The present study determined whether the presence of neuritic plaques (NPs) and diffuse plaques (DPs) are associated with performance in a number of cognitive domains after accounting for APOE ɛ4 carrier status and neurofibrillary tangle presence in a cohort of 123 older participants from the Rush Religious Order Study who died with a premortem clinical diagnosis of no cognitive impairment (NCI). After adjusting for age at death, education, gender, Braak stage, and APOE ɛ4 carrier status, the presence of NPs was associated with lower performance in the cognitive domains of Global Cognition (p = 0.002), Episodic Memory (p = 0.03), Semantic Memory (p = 0.009), and Visuospatial performance (p = 0.006), while DPs showed no association with any cognitive domain examined. These results suggest that decreases in cognition in elderly NCI individuals are associated with an increase in NPs and not DPs when age at death, education, gender, APOE ɛ4 status, and Braak stage are taken into consideration.
Collapse
Affiliation(s)
| | - Sylvia E. Perez
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Kewei Chen
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
| | - Elliott J. Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
193
|
Kwan JSK, Myint PK, Wong A, Mok V, Lau GKK, Mak KFH. Antithrombotic therapy to prevent cognitive decline in people with small vessel disease on neuroimaging but without dementia. Hippokratia 2016. [DOI: 10.1002/14651858.cd012269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Joseph SK Kwan
- The University of Hong Kong, Queen Mary Hospital; Department of Medicine; 102 Pok Fu Lam Road Hong Kong China
| | - Phyo K Myint
- University of Aberdeen; Division of Applied Health Sciences, School of Medicine & Dentistry; Room 4:013 Polwarth Building Foresterhill Aberdeen UK AB25 2ZD
| | - Adrian Wong
- The Chinese University of Hong Kong; Medicine and Therapeutics; 10/F Lui Che Woo Clinical Sciences Building, Prince of Wales Hospital Shatin, N.T. Hong Kong
| | - Vincent Mok
- The Chinese University of Hong Kong; Medicine and Therapeutics; 10/F Lui Che Woo Clinical Sciences Building, Prince of Wales Hospital Shatin, N.T. Hong Kong
| | - Gary KK Lau
- The University of Hong Kong; Division of Neurology, Department of Medicine, LKS Faculty of Medicine; Hong Kong Hong Kong
| | - Ka-Fung Henry Mak
- The University of Hong Kong; Diagnostic Radiology; Room 406, Block K, Queen Mary Hospital, 102 Pokfulam Road Hong Kong Hong Kong
| |
Collapse
|
194
|
Beydoun MA, Beydoun HA, Gamaldo AA, Rostant OS, Dore GA, Zonderman AB, Eid SM. Nationwide Inpatient Prevalence, Predictors, and Outcomes of Alzheimer's Disease among Older Adults in the United States, 2002-2012. J Alzheimers Dis 2016; 48:361-75. [PMID: 26402000 DOI: 10.3233/jad-150228] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In the inpatient setting, prevalence, predictors, and outcomes [mortality risk (MR), length of stay (LOS), and total charges (TC)] of Alzheimer's disease (AD) are largely unknown. We used data on older adults (60+ y) from the Nationwide Inpatient Sample (NIS) 2002-2012. AD prevalence was ∼3.12% in 2012 (total weighted discharges with AD ± standard error: 474, 410 ± 6,276). Co-morbidities prevailing more in AD inpatient admissions included depression (OR = 1.67, 95% CI: 1.63-1.71, p < 0.001), fluid/electrolyte disorders (OR = 1.25, 95% CI: 1.22-1.27, p < 0.001), weight loss (OR = 1.26, 95% CI: 1.22-1.30, p < 0.001), and psychosis (OR = 2.59, 95% CI: 2.47-2.71, p < 0.001), with mean total co-morbidities increasing over time. AD was linked to higher MR and longer LOS, but lower TC. TC rose in AD, while MR and LOS dropped markedly over time. In AD, co-morbidities predicting simultaneously higher MR, TC, and LOS (2012) included congestive heart failure, chronic pulmonary disease, coagulopathy, fluid/electrolyte disorders, metastatic cancer, paralysis, pulmonary circulatory disorders, and weight loss. In sum, co-morbidities and TC increased over time in AD, while MR and LOS dropped. Few co-morbidities predicted occurrence of AD or adverse outcomes in AD.
Collapse
Affiliation(s)
- May A Beydoun
- National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, USA
| | - Hind A Beydoun
- Graduate Program in Public Health, Eastern Virginia Medical School, Norfolk, VA, USA
| | | | - Ola S Rostant
- National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, USA
| | - Greg A Dore
- National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, USA
| | | | - Shaker M Eid
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
195
|
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
|
196
|
Bou Khalil R, Khoury E, Koussa S. Linking multiple pathogenic pathways in Alzheimer's disease. World J Psychiatry 2016; 6:208-214. [PMID: 27354962 PMCID: PMC4919259 DOI: 10.5498/wjp.v6.i2.208] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/24/2016] [Accepted: 05/10/2016] [Indexed: 02/05/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder presenting as progressive cognitive decline with dementia that does not, to this day, benefit from any disease-modifying drug. Multiple etiologic pathways have been explored and demonstrate promising solutions. For example, iron ion chelators, such as deferoxamine, are a potential therapeutic solution around which future studies are being directed. Another promising domain is related to thrombin inhibitors. In this minireview, a common pathophysiological pathway is suggested for the pathogenesis of AD to prove that all these mechanisms converge onto the same cascade of neuroinflammatory events. This common pathway is initiated by the presence of vascular risk factors that induce brain tissue hypoxia, which leads to endothelial cell activation. However, the ensuing hypoxia stimulates the production and release of reactive oxygen species and pro-inflammatory proteins. Furthermore, the endothelial activation may become excessive and dysfunctional in predisposed individuals, leading to thrombin activation and iron ion decompartmentalization. The oxidative stress that results from these modifications in the neurovascular unit will eventually lead to neuronal and glial cell death, ultimately leading to the development of AD. Hence, future research in this field should focus on conducting trials with combinations of potentially efficient treatments, such as the combination of intranasal deferoxamine and direct thrombin inhibitors.
Collapse
|
197
|
Wes PD, Sayed FA, Bard F, Gan L. Targeting microglia for the treatment of Alzheimer's Disease. Glia 2016; 64:1710-32. [DOI: 10.1002/glia.22988] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/22/2016] [Accepted: 03/24/2016] [Indexed: 12/19/2022]
Affiliation(s)
- Paul D. Wes
- Neuroinflammation Department; Lundbeck Research USA; Paramus New Jersey
| | - Faten A. Sayed
- Gladstone Institute for Neurodegeneration; San Francisco California
| | | | - Li Gan
- Gladstone Institute for Neurodegeneration; San Francisco California
| |
Collapse
|
198
|
Aung HH, Altman R, Nyunt T, Kim J, Nuthikattu S, Budamagunta M, Voss JC, Wilson D, Rutledge JC, Villablanca AC. Lipotoxic brain microvascular injury is mediated by activating transcription factor 3-dependent inflammatory and oxidative stress pathways. J Lipid Res 2016; 57:955-68. [PMID: 27087439 DOI: 10.1194/jlr.m061853] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Indexed: 01/10/2023] Open
Abstract
Dysfunction of the cerebrovasculature plays an important role in vascular cognitive impairment (VCI). Lipotoxic injury of the systemic endothelium in response to hydrolyzed triglyceride-rich lipoproteins (TGRLs; TGRL lipolysis products) or a high-fat Western diet (WD) suggests similar mechanisms may be present in brain microvascular endothelium. We investigated the hypothesis that TGRL lipolysis products cause lipotoxic injury to brain microvascular endothelium by generating increased mitochondrial superoxide radical generation, upregulation of activating transcription factor 3 (ATF3)-dependent inflammatory pathways, and activation of cellular oxidative stress and apoptotic pathways. Human brain microvascular endothelial cells were treated with human TGRL lipolysis products that induced intracellular lipid droplet formation, mitochondrial superoxide generation, ATF3-dependent transcription of proinflammatory, stress response, and oxidative stress genes, as well as activation of proapoptotic cascades. Male apoE knockout mice were fed a high-fat/high-cholesterol WD for 2 months, and brain microvessels were isolated by laser capture microdissection. ATF3 gene transcription was elevated 8-fold in the hippocampus and cerebellar brain region of the WD-fed animals compared with chow-fed control animals. The microvascular injury phenotypes observed in vitro and in vivo were similar. ATF3 plays an important role in mediating brain microvascular responses to acute and chronic lipotoxic injury and may be an important preventative and therapeutic target for endothelial dysfunction in VCI.
Collapse
Affiliation(s)
- Hnin Hnin Aung
- Division of Cardiovascular Medicine, Department of Internal Medicine School of Medicine
| | - Robin Altman
- Division of Cardiovascular Medicine, Department of Internal Medicine School of Medicine
| | - Tun Nyunt
- Division of Cardiovascular Medicine, Department of Internal Medicine School of Medicine
| | - Jeffrey Kim
- Division of Cardiovascular Medicine, Department of Internal Medicine School of Medicine
| | | | - Madhu Budamagunta
- Department of Biochemistry and Molecular Medicine, School of Medicine
| | - John C Voss
- Department of Biochemistry and Molecular Medicine, School of Medicine
| | - Dennis Wilson
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616
| | - John C Rutledge
- Division of Cardiovascular Medicine, Department of Internal Medicine School of Medicine
| | - Amparo C Villablanca
- Division of Cardiovascular Medicine, Department of Internal Medicine School of Medicine
| |
Collapse
|
199
|
Soto I, Grabowska WA, Onos KD, Graham LC, Jackson HM, Simeone SN, Howell GR. Meox2 haploinsufficiency increases neuronal cell loss in a mouse model of Alzheimer's disease. Neurobiol Aging 2016; 42:50-60. [PMID: 27143421 DOI: 10.1016/j.neurobiolaging.2016.02.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 02/10/2016] [Accepted: 02/20/2016] [Indexed: 10/22/2022]
Abstract
Evidence suggests that multiple genetic and environmental factors conspire together to increase susceptibility to Alzheimer's disease (AD). The amyloid cascade hypothesis states that deposition of the amyloid-β (Aβ) peptide is central to AD; however, evidence in humans and animals suggests that Aβ buildup alone is not sufficient to cause neuronal cell loss and cognitive decline. Mouse models that express high levels of mutant forms of amyloid precursor protein and/or cleaving enzymes deposit amyloid but do not show neuron loss. Therefore, a double-hit hypothesis for AD has been proposed whereby vascular dysfunction precedes and promotes Aβ toxicity. In support of this, copy number variations in mesenchyme homeobox 2 (MEOX2), a gene involved in vascular development, are associated with severe forms of AD. However, the role of MEOX2 in AD has not been studied. Here, we tested Meox2 haploinsufficiency in B6.APP/PS1 (B6.APB(Tg)) mice, a mouse model of AD. Despite no overt differences in plaque deposition or glial activation, B6.APB(Tg) mice that carry only one copy of Meox2 (B6.APB(Tg).Mx(-/+)) show increased neuronal cell loss, particularly in regions containing plaques, compared with B6.APB(Tg) mice. Neuronal cell loss corresponds with a significant decrease in plaque-associated microvessels, further supporting a synergistic effect of vascular compromise and amyloid deposition on neuronal cell dysfunction in AD.
Collapse
Affiliation(s)
- Ileana Soto
- The Jackson Laboratory, Bar Harbor, ME, USA; Department of Biological Science, Rowan University, Glassboro, NJ, USA; Department of Biomedical and Translational Sciences, Rowan University, Glassboro, NJ, USA
| | - Weronika A Grabowska
- The Jackson Laboratory, Bar Harbor, ME, USA; College of the Atlantic, Bar Harbor, ME, USA
| | | | - Leah C Graham
- The Jackson Laboratory, Bar Harbor, ME, USA; Graduate Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA
| | | | | | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME, USA; Graduate Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA.
| |
Collapse
|
200
|
Daugherty AM, Raz N. Accumulation of iron in the putamen predicts its shrinkage in healthy older adults: A multi-occasion longitudinal study. Neuroimage 2016; 128:11-20. [PMID: 26746579 PMCID: PMC4762718 DOI: 10.1016/j.neuroimage.2015.12.045] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/16/2015] [Accepted: 12/23/2015] [Indexed: 10/22/2022] Open
Abstract
Accumulation of non-heme iron is believed to play a major role in neurodegeneration of the basal ganglia. In healthy aging, however, the temporal relationship between change in brain iron content and age-related volume loss is unclear. Here, we present the first long-term longitudinal multi-occasion investigation of changes in iron content and volume in the neostriatum in a sample of healthy middle-aged and older adults (N=32; ages 49-83years at baseline). Iron content, estimated via R2* relaxometry, increased in the putamen, but not the caudate nucleus. In the former, the rate of accumulation was coupled with change in volume. Moreover, greater baseline iron content predicted faster shrinkage and smaller volumes seven years later. Older age partially accounted for individual differences in neostriatal iron content and volume, but vascular risk did not. Thus, brain iron content may be a promising biomarker of impending decline in normal aging.
Collapse
Affiliation(s)
- Ana M Daugherty
- Institute of Gerontology, Wayne State University, Detroit, MI, USA.
| | - Naftali Raz
- Institute of Gerontology, Wayne State University, Detroit, MI, USA; Psychology Department, Wayne State University, Detroit, MI, USA
| |
Collapse
|