251
|
Ferreira LK, Tamashiro-Duran JH, Squarzoni P, Duran FL, Alves TC, Buchpiguel CA, Busatto GF. The link between cardiovascular risk, Alzheimer's disease, and mild cognitive impairment: support from recent functional neuroimaging studies. ACTA ACUST UNITED AC 2014; 36:344-57. [PMID: 24918525 DOI: 10.1590/1516-4446-2013-1275] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 01/03/2014] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To review functional neuroimaging studies about the relationship between cardiovascular risk factors (CVRFs), Alzheimer's disease (AD), and mild cognitive impairment (MCI). METHODS We performed a comprehensive literature search to identify articles in the neuroimaging field addressing CVRF in AD and MCI. We included studies that used positron emission tomography (PET), single photon emission computerized tomography (SPECT), or functional magnetic resonance imaging (fMRI). RESULTS CVRFs have been considered risk factors for cognitive decline, MCI, and AD. Patterns of AD-like changes in brain function have been found in association with several CVRFs (both regarding individual risk factors and also composite CVRF measures). In vivo assessment of AD-related pathology with amyloid imaging techniques provided further evidence linking CVRFs and AD, but there is still limited information resulting from this new technology. CONCLUSION There is a large body of evidence from functional neuroimaging studies supporting the hypothesis that CVRFs may play a causal role in the pathophysiology of AD. A major limitation of most studies is their cross-sectional design; future longitudinal studies using multiple imaging modalities are expected to better document changes in CVRF-related brain function patterns and provide a clearer picture of the complex relationship between aging, CVRFs, and AD.
Collapse
Affiliation(s)
- Luiz K Ferreira
- Laboratory of Psychiatric Neuroimaging (LIM-21), Department and Institute of Psychiatry, School of Medicine, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Jaqueline H Tamashiro-Duran
- Laboratory of Psychiatric Neuroimaging (LIM-21), Department and Institute of Psychiatry, School of Medicine, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Paula Squarzoni
- Laboratory of Psychiatric Neuroimaging (LIM-21), Department and Institute of Psychiatry, School of Medicine, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Fabio L Duran
- Laboratory of Psychiatric Neuroimaging (LIM-21), Department and Institute of Psychiatry, School of Medicine, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Tania C Alves
- Laboratory of Psychiatric Neuroimaging (LIM-21), Department and Institute of Psychiatry, School of Medicine, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Carlos A Buchpiguel
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), USP, São Paulo, SP, Brazil
| | - Geraldo F Busatto
- Laboratory of Psychiatric Neuroimaging (LIM-21), Department and Institute of Psychiatry, School of Medicine, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| |
Collapse
|
252
|
McLarnon JG. Correlated inflammatory responses and neurodegeneration in peptide-injected animal models of Alzheimer's disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:923670. [PMID: 24822221 PMCID: PMC4005142 DOI: 10.1155/2014/923670] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/26/2014] [Accepted: 02/27/2014] [Indexed: 12/15/2022]
Abstract
Animal models of Alzheimer's disease (AD) which emphasize activation of microglia may have particular utility in correlating proinflammatory activity with neurodegeneration. This paper reviews injection of amyloid- β (A β ) into rat brain as an alternative AD animal model to the use of transgenic animals. In particular, intrahippocampal injection of Aβ 1-42 peptide demonstrates prominent microglial mobilization and activation accompanied by a significant loss of granule cell neurons. Furthermore, pharmacological inhibition of inflammatory reactivity is demonstrated by a broad spectrum of drugs with a common endpoint in conferring neuroprotection in peptide-injected animals. Peptide-injection models provide a focus on glial cell responses to direct peptide injection in rat brain and offer advantages in the study of the mechanisms underlying neuroinflammation in AD brain.
Collapse
Affiliation(s)
- James G. McLarnon
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada V6T 1W3
| |
Collapse
|
253
|
An apolipoprotein E modified liposomal nanoparticle: Ligand dependent efficiency as a siRNA delivery carrier for mouse-derived brain endothelial cells. Int J Pharm 2014; 465:77-82. [DOI: 10.1016/j.ijpharm.2014.02.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/21/2013] [Accepted: 02/08/2014] [Indexed: 12/26/2022]
|
254
|
Valproic acid suppresses lipopolysaccharide-induced cyclooxygenase-2 expression via MKP-1 in murine brain microvascular endothelial cells. Biochem Pharmacol 2014; 88:372-83. [DOI: 10.1016/j.bcp.2014.02.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/07/2014] [Accepted: 02/07/2014] [Indexed: 02/06/2023]
|
255
|
Abstract
Cerebrovascular dysfunction significantly contributes to the clinical presentation and pathoetiology of Alzheimer's disease (AD). Deposition and aggregation of β-amyloid (Aβ) within vascular smooth muscle cells leads to inflammation, oxidative stress, impaired vasorelaxation, and disruption of blood-brain barrier integrity. Midlife vascular risk factors, such as hypertension, cardiovascular disease, diabetes, and dyslipidemia, increase the relative risk for AD. These comorbidities are all characterized by low and/or dysfunctional high-density lipoproteins (HDL), which itself is a risk factor for AD. HDL performs a wide variety of critical functions in the periphery and CNS. In addition to lipid transport, HDL regulates vascular health via mediating vasorelaxation, inflammation, and oxidative stress and promotes endothelial cell survival and integrity. Here, we summarize clinical and preclinical data examining the involvement of HDL, originating from the circulation and from within the CNS, on AD and hypothesize potential synergistic actions between the two lipoprotein pools.
Collapse
|
256
|
Engelhardt S, Patkar S, Ogunshola OO. Cell-specific blood-brain barrier regulation in health and disease: a focus on hypoxia. Br J Pharmacol 2014; 171:1210-30. [PMID: 24641185 PMCID: PMC3952799 DOI: 10.1111/bph.12489] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/02/2013] [Accepted: 10/16/2013] [Indexed: 01/16/2023] Open
Abstract
The blood-brain barrier (BBB) is a complex vascular structure consisting of microvascular endothelial cells that line the vessel wall, astrocyte end-feet, pericytes, as well as the basal lamina. BBB cells act in concert to maintain the characteristic impermeable and low paracellular flux of the brain vascular network, thus ensuring a homeostatic neuronal environment. Alterations in BBB stability that occur during injury have dire consequences on disease progression and it is clear that BBB cell-specific responses, positive or negative, must make a significant contribution to injury outcome. Reduced oxygenation, or hypoxia, is a characteristic of many brain diseases that significantly increases barrier permeability. Recent data suggest that hypoxia-inducible factor (HIF-1), the master regulator of the hypoxic response, probably mediates many hypoxic effects either directly or indirectly via its target genes. This review discusses current knowledge of physiological cell-specific regulation of barrier function, their responses to hypoxia as well as consequences of hypoxic- and HIF-1-mediated mechanisms on barrier integrity during select brain diseases. In the final sections, the potential of current advances in targeting HIF-1 as a therapeutic strategy will be overviewed.
Collapse
Affiliation(s)
- S Engelhardt
- Institute of Veterinary Physiology, University of ZurichZurich, Switzerland
| | - S Patkar
- Institute of Veterinary Physiology, University of ZurichZurich, Switzerland
| | - O O Ogunshola
- Institute of Veterinary Physiology, University of ZurichZurich, Switzerland
| |
Collapse
|
257
|
Lim DC, Pack AI. Obstructive sleep apnea and cognitive impairment: addressing the blood-brain barrier. Sleep Med Rev 2014; 18:35-48. [PMID: 23541562 PMCID: PMC3758447 DOI: 10.1016/j.smrv.2012.12.003] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 12/21/2012] [Accepted: 12/24/2012] [Indexed: 12/14/2022]
Abstract
Increasing data support a connection between obstructive sleep apnea (OSA) and cognitive impairment but a causal link has yet to be established. Although neuronal loss has been linked to cognitive impairment, emerging theories propose that changes in synaptic plasticity can cause cognitive impairment. Studies demonstrate that disruption to the blood-brain barrier (BBB), which is uniquely structured to tightly maintain homeostasis inside the brain, leads to changes in the brain's microenvironment and affects synaptic plasticity. Cyclical intermittent hypoxia is a stressor that could disrupt the BBB via molecular responses already known to occur in either OSA patients or animal models of intermittent hypoxia. However, we do not yet know if or how intermittent hypoxia can cause cognitive impairment by mechanisms operating at the BBB. Therefore, we propose that initially, adaptive homeostatic responses at the BBB occur in response to increased oxygen and nutrient demand, specifically through regulation of influx and efflux BBB transporters that alter microvessel permeability. We further hypothesize that although these responses are initially adaptive, these changes in BBB transporters can have long-term consequences that disrupt the brain's microenvironment and alter synaptic plasticity leading to cognitive impairment.
Collapse
Affiliation(s)
- Diane C Lim
- Department of Medicine, Division of Sleep Medicine, and Center for Sleep and Circadian Neurobiology, University of Pennsylvania, 125 South 31st Street, Suite 2100, Philadelphia, PA 19104, USA.
| | - Allan I Pack
- Department of Medicine, Division of Sleep Medicine, and Center for Sleep and Circadian Neurobiology, University of Pennsylvania, 125 South 31st Street, Suite 2100, Philadelphia, PA 19104, USA.
| |
Collapse
|
258
|
Heringa SM, van den Berg E, Reijmer YD, Nijpels G, Stehouwer CDA, Schalkwijk CG, Teerlink T, Scheffer PG, van den Hurk K, Kappelle LJ, Dekker JM, Biessels GJ. Markers of low-grade inflammation and endothelial dysfunction are related to reduced information processing speed and executive functioning in an older population - the Hoorn Study. Psychoneuroendocrinology 2014; 40:108-18. [PMID: 24485482 DOI: 10.1016/j.psyneuen.2013.11.011] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 10/26/2013] [Accepted: 11/11/2013] [Indexed: 11/29/2022]
Abstract
Low-grade inflammation and endothelial dysfunction are related to cognitive decline and dementia, in a complex interplay with vascular factors and aging. We investigated, in an older population, low-grade inflammation and endothelial dysfunction in relation to detailed assessment of cognitive functioning. Furthermore, we explored this association within the context of vascular factors. 377 participants (73 ± 6 years) of the population-based Hoorn Study were included. In plasma samples of 2000-2001 (n=363) and/or 2005-2008 (n=323), biomarkers were determined of low-grade inflammation (CRP, TNF-alpha, IL-6, IL-8, SAA, MPO, and sICAM-1) and endothelial dysfunction (vWF, sICAM-1, sVCAM-1, sTM, sE-selectin). In 2005-2008, all participants underwent neuropsychological examination. Composite z-scores were computed for low-grade inflammation and endothelial dysfunction at both time points, and for six domains of cognitive functioning (abstract reasoning, memory, information processing speed, attention and executive functioning, visuoconstruction, and language). The association between low-grade inflammation and endothelial dysfunction, and cognitive functioning was evaluated with linear regression analysis. In secondary analyses, we explored the relation with vascular risk factors and cardiovascular disease. Low-grade inflammation and endothelial dysfunction were associated with worse performance on information processing speed and attention and executive functioning, in prospective and cross-sectional analyses (standardized betas ranging from -0.20 to -0.10). No significant relation with other cognitive domains was observed. Adjusting for vascular factors slightly attenuated the associations. Low-grade inflammation and endothelial dysfunction accounted for only 2.6% explained variance in cognitive functioning, on top of related vascular risk factors and cardiovascular disease. Bootstrapping analyses show that low-grade inflammation and endothelial dysfunction mediate the relation between vascular risk factors and cognitive functioning. This study shows that low-grade inflammation and endothelial dysfunction contribute to reduced information processing speed and executive functioning in an older population.
Collapse
Affiliation(s)
- S M Heringa
- University Medical Center Utrecht, Brain Center Rudolf Magnus, Department of Neurology, Utrecht, The Netherlands.
| | - E van den Berg
- University Medical Center Utrecht, Brain Center Rudolf Magnus, Department of Neurology, Utrecht, The Netherlands; Utrecht University, Helmholtz Institute, Department of Experimental Psychology, Utrecht, The Netherlands
| | - Y D Reijmer
- Massachusetts General Hospital, J. Philip Kistler Stroke Research Center, Boston, United States
| | - G Nijpels
- VU University Medical Center, Institute for Research in Extramural Medicine (EMGO Institute), Amsterdam, The Netherlands; VU University Medical Center, Department of General Practice, Amsterdam, The Netherlands
| | - C D A Stehouwer
- Maastricht University Medical Centre, Department of Internal Medicine, Maastricht, The Netherlands; Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - C G Schalkwijk
- Maastricht University Medical Centre, Department of Internal Medicine, Maastricht, The Netherlands; Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - T Teerlink
- VU University Medical Center, Department of Clinical Chemistry, Amsterdam, The Netherlands
| | - P G Scheffer
- VU University Medical Center, Department of Clinical Chemistry, Amsterdam, The Netherlands
| | - K van den Hurk
- Sanquin Blood Supply, Department of Donor Studies, Amsterdam, The Netherlands
| | - L J Kappelle
- University Medical Center Utrecht, Brain Center Rudolf Magnus, Department of Neurology, Utrecht, The Netherlands
| | - J M Dekker
- VU University Medical Center, Institute for Research in Extramural Medicine (EMGO Institute), Amsterdam, The Netherlands
| | - G J Biessels
- University Medical Center Utrecht, Brain Center Rudolf Magnus, Department of Neurology, Utrecht, The Netherlands
| |
Collapse
|
259
|
KIF6 719Arg Carrier Status Association with Homocysteine and C-Reactive Protein in Amnestic Mild Cognitive Impairment and Alzheimer's Disease Patients. Int J Alzheimers Dis 2014; 2013:242303. [PMID: 24455405 PMCID: PMC3884607 DOI: 10.1155/2013/242303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 10/11/2013] [Accepted: 10/12/2013] [Indexed: 12/02/2022] Open
Abstract
Recent research has demonstrated associations between statin use, KIF6 719Arg carrier status, and cholesterol levels and amnestic mild cognitive impairment (aMCI) and Alzheimer's disease (AD) patients. The association between 719Arg carrier status with homocysteine (tHcy) and c-reactive protein (CRP) levels in aMCI and AD has not been previously investigated. Data from 175 aMCI and AD patients were used for the analysis. 719Arg carriers had significantly lower levels of tHcy than noncarriers (P = 0.02). No significant difference in CRP levels between 719Arg carriers and noncarriers was present (P = 0.37). Logistic regression yielded no significant effect for 719Arg status on CRP [OR = 1.79 (0.85, 3.83), P = 0.13] but did demonstrate a significant effect for tHcy [OR = 0.44 (0.23, 0.83), P = 0.01] after adjusting for ApoE ε4 carrier status, age, gender, and statin use. This study is the first to explore the relationship between KIF6 719Arg carrier status with tHcy and CRP levels. 719Arg carriers were more likely to have normal tHcy levels after adjusting for ApoE ε4 status, age, gender, and statin use. These results suggest that the KIF6 gene might influence cardiovascular pathways associated with AD.
Collapse
|
260
|
Burstein AH, Grimes I, Galasko DR, Aisen PS, Sabbagh M, Mjalli AMM. Effect of TTP488 in patients with mild to moderate Alzheimer's disease. BMC Neurol 2014; 14:12. [PMID: 24423155 PMCID: PMC4021072 DOI: 10.1186/1471-2377-14-12] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 01/06/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND TTP488, an antagonist at the Receptor for Advanced Glycation End products, was evaluated as a potential treatment for patients with mild-to-moderate Alzheimer's disease (AD). A previous report describes decreased decline in ADAS-cog (delta = 3.1, p = 0.008 at 18 months, ANCOVA with multiple imputation), relative to placebo, following a 5 mg/day dose of TTP488. Acute, reversible cognitive worsening was seen with a 20 mg/day dose. The present study further evaluates the efficacy of TTP488 by subgroup analyses based on disease severity and concentration effect analysis. METHODS 399 patients were randomized to one of two oral TTP488 doses (60 mg for 6 days followed by 20 mg/day; 15 mg for 6 days followed by 5 mg/day) or placebo for 18 months. Pre-specified primary analysis, using an ITT population, was on the ADAS-cog11. Secondary analyses included as a key secondary variable the Clinical Dementia Rating-Sum of Boxes (CDR-SB), and another secondary variable of the ADCS-ADL. RESULTS On-treatment analysis demonstrated numerical differences favoring 5 mg/day over placebo, with nominal significance at Month 18 (delta = 2.7, p = 0.03). Patients with mild AD, whether defined by MMSE or ADAS-cog, demonstrated significant differences favoring 5 mg/day on ADAS-cog and trends on CDR-sb and ADCS-ADL at Month 18. TTP488 plasma concentrations of 7.6-16.8 ng/mL were associated with a decreased decline in ADAS-cog over time compared to placebo. Worsening on the ADAS-cog relative to placebo was evident at 46.8-167.0 ng/mL. CONCLUSIONS Results of these analyses support further investigation of 5 mg/day in future Phase 3 trials in patients with mild AD.
Collapse
Affiliation(s)
| | | | - Douglas R Galasko
- Department of Neurosciences, University of California, San Diego, CA, USA
| | - Paul S Aisen
- Department of Neurosciences, University of California, San Diego, CA, USA
| | - Marwan Sabbagh
- Banner Sun Health Research Institute, Sun City, AZ, USA
- University of Arizona, Tucson, Arizona, AZ, USA
| | | |
Collapse
|
261
|
PINK1 and its familial Parkinson's disease-associated mutation regulate brain vascular endothelial inflammation. J Mol Neurosci 2014; 53:109-16. [PMID: 24385196 DOI: 10.1007/s12031-013-0207-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 12/08/2013] [Indexed: 12/18/2022]
Abstract
Parkinson's disease (PD) is a debilitating disorder that affects movement. Inflammation-mediated endothelial dysfunction has been found to be involved in neurodegenerative diseases, including PD. More than 40 PTEN-induced putative kinase 1 (PINK1) mutations have been found in PD patients. The effects of PINK1 in vascular inflammation are as yet unknown. In this study, our findings revealed that PINK1 can be increased by the inflammatory cytokine tumor necrosis factor-α in primary human brain microvascular endothelial cells (HBMECs). We found that wild-type PINK1 prevents expression of the adhesion molecule vascular cell adhesion molecule-1 (VCAM-1), thus inhibiting the attachment of monocytes to brain endothelial cells. However, PINK1G309D, the loss-of-function mutation associated with early-onset familial PD, promotes expression of VCAM-1 and exacerbates attachment of monocytes to brain endothelial cells. Mechanism studies revealed that overexpression of wild-type PINK1 inhibits the VCAM-1 promoter by inhibiting the transcriptional activity of interferon regulatory factor 1 (IRF-1). However, PINK1G309D promotes the VCAM-1 promoter by increasing the transcriptional activity of IRF-1.
Collapse
|
262
|
Ghiso J, Fossati S, Rostagno A. Amyloidosis associated with cerebral amyloid angiopathy: cell signaling pathways elicited in cerebral endothelial cells. J Alzheimers Dis 2014; 42 Suppl 3:S167-76. [PMID: 24670400 PMCID: PMC4467213 DOI: 10.3233/jad-140027] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Substantial genetic, biochemical, and in vivo data indicate that progressive accumulation of amyloid-β (Aβ) plays a central role in the pathogenesis of Alzheimer's disease (AD). Historically centered in the importance of parenchymal plaques, the role of cerebral amyloid angiopathy (CAA)--a frequently neglected amyloid deposit present in >80% of AD cases--for the mechanism of disease pathogenesis is now starting to emerge. CAA consistently associates with microvascular modifications, ischemic lesions, micro- and macro-hemorrhages, and dementia, progressively affecting cerebral blood flow, altering blood-brain barrier permeability, interfering with brain clearance mechanisms and triggering a cascade of deleterious pro-inflammatory and metabolic events that compromise the integrity of the neurovascular unit. New evidence highlights the contribution of pre-fibrillar Aβ in the induction of cerebral endothelial cell dysfunction. The recently discovered interaction of oligomeric Aβ species with TRAIL DR4 and DR5 cell surface death receptors mediates the engagement of mitochondrial pathways and sequential activation of multiple caspases, eliciting a cascade of cell death mechanisms while unveiling an opportunity for exploring mechanistic-based therapeutic interventions to preserve the integrity of the neurovascular unit.
Collapse
Affiliation(s)
- Jorge Ghiso
- Department of Pathology, New York University School of Medicine, New York, NY, USA Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Silvia Fossati
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Agueda Rostagno
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
263
|
Shrestha R, Millington O, Brewer J, Dev KK, Bushell TJ. Lymphocyte-mediated neuroprotection in in vitro models of excitotoxicity involves astrocytic activation and the inhibition of MAP kinase signalling pathways. Neuropharmacology 2014; 76 Pt A:184-93. [DOI: 10.1016/j.neuropharm.2013.06.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 06/18/2013] [Accepted: 06/25/2013] [Indexed: 12/14/2022]
|
264
|
Edwards M, Balldin VH, Hall J, O'Bryant S. Combining select neuropsychological assessment with blood-based biomarkers to detect mild Alzheimer's disease: a molecular neuropsychology approach. J Alzheimers Dis 2014; 42:635-40. [PMID: 24916542 PMCID: PMC4416413 DOI: 10.3233/jad-140852] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Current work has sought to establish a rapid and cost effective means of screening for Alzheimer's disease (AD) with the most recent findings showing utility of integrating blood-based biomarkers with cognitive measures. OBJECTIVE The current project sought to create a combined biomarker-cognitive profile to detect mild AD. METHODS Data was analyzed from 266 participants (129 AD cases [Early AD n = 93; Very Early AD n = 36]; 137 controls) enrolled in the Texas Alzheimer's Research and Care Consortium (TARCC). Non-fasting serum samples were collected from each participant and assayed via a multi-plex biomarker assay platform using electrochemiluminescence. Logistic Regression was utilized to detect early AD using two serum biomarkers (TNFα and IL7), demographic information (age), and one neuropsychological measure (Clock 4-point) as predictor variable. Disease severity was determined via Clinical Dementia Rating (CDR) scale global scores. RESULTS In the total sample (all levels of CDR scores), the combination of biomarkers, cognitive test score, and demographics yielded the obtained sensitivity (SN) of 0.94, specificity (SP) of 0.90, and an overall accuracy of 0.92. When examining early AD cases (i.e.m CDR = 0.5-1), the biomarker-cognitive profile yielded SN of 0.94, SP of 0.85, and an overall accuracy of 0.91. When restricted to very early AD cases (i.e., CDR = 0.5), the biomarker-cognitive profile yielded SN of 0.97 and SP of 0.72, with an overall accuracy of 0.91. CONCLUSIONS The combination of demographics, two biomarkers, and one cognitive test created a biomarker-cognitive profile that was highly accurate in detecting the presence of AD, even in the very early stages.
Collapse
Affiliation(s)
- Melissa Edwards
- Department of Psychology, University of North Texas, Denton, TX, USA
| | | | - James Hall
- Department of Psychiatry, University of North Texas Health Science Center, Fort Worth, TX, USA Institute for Aging & Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Sid O'Bryant
- Institute for Aging & Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX, USA Department of Internal Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
265
|
Welser-Alves JV, Boroujerdi A, Milner R. Isolation and culture of primary mouse brain endothelial cells. Methods Mol Biol 2014; 1135:345-56. [PMID: 24510877 DOI: 10.1007/978-1-4939-0320-7_28] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Blood vessels in the central nervous system (CNS) are unique in forming the blood-brain barrier (BBB), which confers high electrical resistance and low permeability properties, thus protecting neural cells from potentially harmful blood components. Endothelial cells, which form the inner cellular lining of all blood vessels, play a critical role in this process by forming tight adhesive interactions between each other. To study the properties of primary brain endothelial cells (BECs), a number of different methods have been described. In this chapter, we present a relatively simple method that produces high numbers of primary mouse BECs that are highly pure (greater than 99 % CD31-positive). In addition, we also describe an immunocytochemical approach to demonstrate the endothelial purity of these cultures.
Collapse
Affiliation(s)
- Jennifer V Welser-Alves
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | | | |
Collapse
|
266
|
Najjar S, Pearlman DM, Devinsky O, Najjar A, Zagzag D. Neurovascular unit dysfunction with blood-brain barrier hyperpermeability contributes to major depressive disorder: a review of clinical and experimental evidence. J Neuroinflammation 2013; 10:142. [PMID: 24289502 PMCID: PMC4220803 DOI: 10.1186/1742-2094-10-142] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 11/15/2013] [Indexed: 12/27/2022] Open
Abstract
About one-third of people with major depressive disorder (MDD) fail at least two antidepressant drug trials at 1 year. Together with clinical and experimental evidence indicating that the pathophysiology of MDD is multifactorial, this observation underscores the importance of elucidating mechanisms beyond monoaminergic dysregulation that can contribute to the genesis and persistence of MDD. Oxidative stress and neuroinflammation are mechanistically linked to the presence of neurovascular dysfunction with blood-brain barrier (BBB) hyperpermeability in selected neurological disorders, such as stroke, epilepsy, multiple sclerosis, traumatic brain injury, and Alzheimer’s disease. In contrast to other major psychiatric disorders, MDD is frequently comorbid with such neurological disorders and constitutes an independent risk factor for morbidity and mortality in disorders characterized by vascular endothelial dysfunction (cardiovascular disease and diabetes mellitus). Oxidative stress and neuroinflammation are implicated in the neurobiology of MDD. More recent evidence links neurovascular dysfunction with BBB hyperpermeability to MDD without neurological comorbidity. We review this emerging literature and present a theoretical integration between these abnormalities to those involving oxidative stress and neuroinflammation in MDD. We discuss our hypothesis that alterations in endothelial nitric oxide levels and endothelial nitric oxide synthase uncoupling are central mechanistic links in this regard. Understanding the contribution of neurovascular dysfunction with BBB hyperpermeability to the pathophysiology of MDD may help to identify novel therapeutic and preventative approaches.
Collapse
Affiliation(s)
- Souhel Najjar
- Department of Neurology, Neuroinflammation Research Group, Epilepsy Center Division, NYU School of Medicine, New York, NY 10016, USA.
| | | | | | | | | |
Collapse
|
267
|
Falsafi SK, Roßner S, Ghafari M, Groessl M, Morawski M, Gerner C, Lubec G. Changes of several brain receptor complexes in the cerebral cortex of patients with Alzheimer disease: probable new potential pharmaceutical targets. Amino Acids 2013; 46:223-33. [PMID: 24292102 DOI: 10.1007/s00726-013-1623-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/04/2013] [Indexed: 12/31/2022]
Abstract
Although Alzheimer disease (AD) has been linked to defects in major brain receptors, studies thus far have been limited to the determination of receptor subunits or specific ligand binding studies. However, the availability of current technology enables the determination and quantification of brain receptor complexes. Thus, we examined levels of native receptor complexes in the brains of patients with AD. Cortical tissue was obtained from control subjects (n = 12 females and 12 males) and patients with AD (n = 12 females and 12 males) within a 3-h postmortem time period. The tissues were kept frozen until further biochemical analyses. Membrane proteins were extracted and subsequently enriched by ultracentrifugation using a sucrose gradient. Membrane proteins were then electrophoresed onto native gels and immunoblotted using antibodies against individual brain receptors. We found that the levels were comparable for complexes containing GluR2, GluR3 and GluR4 as well as 5-HT1A. Moreover, the levels of complexes containing muscarinic AChR M1, NR1 and GluR1 were significantly increased in male patients with AD. Nicotinic AChRs 4 and 7 as well as dopaminergic receptors D1 and D2 were also increased in males and females with AD. These findings reveal a pattern of altered receptor complex levels that may contribute to the deterioration of the concerted activity of these receptors and thus result in cognitive deficits observed in patients with AD. It should be emphasised that receptor complexes function as working units rather than individual subunits. Thus, the receptor deficits identified may be relevant for the design of experimental therapies. Therefore, specific pharmacological modulation of these receptors is within the pharmaceutical repertoire.
Collapse
Affiliation(s)
- Soheil Keihan Falsafi
- Department of Pediatrics, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
268
|
Bungart BL, Dong L, Sobek D, Sun GY, Yao G, Lee JCM. Nanoparticle-emitted light attenuates amyloid-β-induced superoxide and inflammation in astrocytes. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 10:15-7. [PMID: 24200521 DOI: 10.1016/j.nano.2013.10.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 10/23/2013] [Accepted: 10/25/2013] [Indexed: 01/16/2023]
Abstract
UNLABELLED Alzheimer's disease (AD) is the sixth leading cause of age-related death with no effective intervention yet available. Our previous studies have demonstrated the potential efficacy of Low Level Laser Therapy (LLLT) in AD cell models by mitigating amyloid-β peptide (Aβ)-induced oxidative stress and inflammation. However, the penetration depth of light is still the major challenge for implementing LLLT in animal models and in the clinical settings. In this study, we present the potential of applying Bioluminescence Resonance Energy Transfer to Quantum Dots (BRET-Qdots) as an alternative near infrared (NIR) light source for LLLT. Our results show that BRET-Qdot-emitted NIR suppresses Aβ-induced oxidative stress and inflammatory responses in primary rat astrocytes. These data provide a proof of concept for a nanomedicine platform for LLLT. FROM THE CLINICAL EDITOR Low Level Laser Therapy has already been demonstrated to mitigate amyloid-β peptide induced oxidative stress and inflammation, a key driver of Alzheimer's disease. The major issue in moving this forward from cell cultures to live animals and potentially to human subjects is light penetration depth. In this novel study, BRET-Qdots were used as an alternative near infrared light source with good efficacy, paving the way to the development of a nanomedicine platform.
Collapse
Affiliation(s)
- Brittani L Bungart
- Department of Biological Engineering, University of Missouri, Columbia, MO
| | - Li Dong
- Department of Biological Engineering, University of Missouri, Columbia, MO
| | | | - Grace Y Sun
- Department of Biochemistry, University of Missouri, Columbia, MO
| | - Gang Yao
- Department of Biological Engineering, University of Missouri, Columbia, MO
| | - James C-M Lee
- Department of Biological Engineering, University of Missouri, Columbia, MO.
| |
Collapse
|
269
|
Hosseinzadeh S, Zahmatkesh M, Zarrindast MR, Hassanzadeh GR, Karimian M, Sarrafnejad A. Elevated CSF and plasma microparticles in a rat model of streptozotocin-induced cognitive impairment. Behav Brain Res 2013; 256:503-11. [DOI: 10.1016/j.bbr.2013.09.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 09/04/2013] [Accepted: 09/07/2013] [Indexed: 01/23/2023]
|
270
|
Mortazavian SM, Parsaee H, Mousavi SH, Tayarani-Najaran Z, Ghorbani A, Sadeghnia HR. Acetylcholinesterase inhibitors promote angiogenesis in chick chorioallantoic membrane and inhibit apoptosis of endothelial cells. Int J Alzheimers Dis 2013; 2013:121068. [PMID: 24159418 PMCID: PMC3789489 DOI: 10.1155/2013/121068] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 08/12/2013] [Accepted: 08/18/2013] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most common causes of dementia in the elderly. Recently, a great attention has been paid to the possible role of vascular changes in the pathogenesis of AD. Reduced microvascular density and degeneration of the endothelium are of structural cerebrovascular changes in AD. Acetylcholinesterase (AChE) inhibitors are widely used for the improvement of AD symptoms. Until now, however, the effects of AChE inhibitors on vascular changes including angiogenesis and endothelial cell apoptosis are not fully understood. In the present work, the effects of three AChE inhibitors (donepezil, rivastigmine, and galantamine) were tested on H2O2-induced apoptosis in human umbilical vein endothelial cells (HUVECs) and on angiogenesis in chicken chorioallantoic membrane model. Incubation of HUVEC with H2O2 led to a significant decrease in cell viability and an increase in the percentage of apoptotic cells. The tested drugs, at concentrations of 1-100 μ M, significantly inhibited the H2O2-induced toxicity. Also, all donepezil, rivastigmine and galantamine significantly increased the number of vessels in the chorioallantoic membrane when injected into fertilized eggs. In conclusion, AChE inhibitors possess angiogenesis-accelerating properties and have antiapoptotic effects on endothelial cells. These effects of AChE inhibitors may be involved in their beneficial effects on AD.
Collapse
Affiliation(s)
- Seyed Mohsen Mortazavian
- Department of Pharmacology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran
| | - Heydar Parsaee
- Department of Pharmacology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran
- Pharmacological Research Center of Medicinal Plants, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran
| | - Seyed Hadi Mousavi
- Department of Pharmacology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran
- Pharmacological Research Center of Medicinal Plants, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran
| | - Zahra Tayarani-Najaran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91775-1365, Iran
| | - Ahmad Ghorbani
- Pharmacological Research Center of Medicinal Plants, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran
| | - Hamid Reza Sadeghnia
- Department of Pharmacology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran
- Pharmacological Research Center of Medicinal Plants, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran
- Neurocognitive Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran
| |
Collapse
|
271
|
Currais A, Maher P. Functional consequences of age-dependent changes in glutathione status in the brain. Antioxid Redox Signal 2013; 19:813-22. [PMID: 23249101 DOI: 10.1089/ars.2012.4996] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
SIGNIFICANCE A decline in both cognitive and motor functions is one of the characteristics of aging. This results in changes in learning and memory, as well as deficits in balance and coordination that significantly impact the quality of life. Importantly, age is the greatest risk factor for a number of neurodegenerative diseases. Alterations in redox homeostasis, protein modification and processing, mitochondrial function, and the immune response have all been implicated in the decline of the aging brain. RECENT ADVANCES Brain glutathione (GSH) decreases with age in humans, and a loss of GSH can impact cognitive function. Decreases in GSH are also associated with microglial activation and endothelial dysfunction, both of which can contribute to impairments in brain function. Changes in redox homeostasis can also potentiate the accumulation of advanced glycation endproducts, resulting in defects in protein processing and function as well as a further increase in inflammation. CRITICAL ISSUES We argue here that many of the changes in brain function associated with age are linked through GSH metabolism. FUTURE DIRECTIONS Further research focused on better understanding how age affects GSH homeostasis with a particular emphasis on the key transcription factors involved in GSH metabolism is needed.
Collapse
Affiliation(s)
- Antonio Currais
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | | |
Collapse
|
272
|
Raha AA, Vaishnav RA, Friedland RP, Bomford A, Raha-Chowdhury R. The systemic iron-regulatory proteins hepcidin and ferroportin are reduced in the brain in Alzheimer's disease. Acta Neuropathol Commun 2013; 1:55. [PMID: 24252754 PMCID: PMC3893417 DOI: 10.1186/2051-5960-1-55] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 08/25/2013] [Indexed: 12/11/2022] Open
Abstract
Background The pathological features of the common neurodegenerative conditions, Alzheimer’s disease (AD), Parkinson’s disease and multiple sclerosis are all known to be associated with iron dysregulation in regions of the brain where the specific pathology is most highly expressed. Iron accumulates in cortical plaques and neurofibrillary tangles in AD where it participates in redox cycling and causes oxidative damage to neurons. To understand these abnormalities in the distribution of iron the expression of proteins that maintain systemic iron balance was investigated in human AD brains and in the APP-transgenic (APP-tg) mouse. Results Protein levels of hepcidin, the iron-homeostatic peptide, and ferroportin, the iron exporter, were significantly reduced in hippocampal lysates from AD brains. By histochemistry, hepcidin and ferroportin were widely distributed in the normal human brain and co-localised in neurons and astrocytes suggesting a role in regulating iron release. In AD brains, hepcidin expression was reduced and restricted to the neuropil, blood vessels and damaged neurons. In the APP-tg mouse immunoreactivity for ferritin light-chain, the iron storage isoform, was initially distributed throughout the brain and as the disease progressed accumulated in the core of amyloid plaques. In human and mouse tissues, extensive AD pathology with amyloid plaques and severe vascular damage with loss of pericytes and endothelial disruption was seen. In AD brains, hepcidin and ferroportin were associated with haem-positive granular deposits in the region of damaged blood vessels. Conclusion Our results suggest that the reduction in ferroportin levels are likely associated with cerebral ischaemia, inflammation, the loss of neurons due to the well-characterised protein misfolding, senile plaque formation and possibly the ageing process itself. The reasons for the reduction in hepcidin levels are less clear but future investigation could examine circulating levels of the peptide in AD and a possible reduction in the passage of hepcidin across damaged vascular endothelium. Imbalance in the levels and distribution of ferritin light-chain further indicate a failure to utilize and release iron by damaged and degenerating neurons.
Collapse
|
273
|
Endothelial dysfunction associated with mild cognitive impairment in elderly population. Aging Clin Exp Res 2013; 25:247-55. [PMID: 23740584 DOI: 10.1007/s40520-013-0043-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 12/14/2012] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND AIMS According to the original Petersen criteria, we investigated the association between endothelial dysfunction and mild cognitive impairment (MCI) by flow-mediated dilation (FMD). We aimed to verify if endothelial dysfunction occurs in MCI and whether vascular factors are implicated in the MCI pathogenesis. METHODS This is a cross-sectional study performed on 34 subjects with clinical diagnosis of MCI and 37 controls, older than 60 years. Patients were enrolled from a geriatric outpatient clinic. All the recognized cardiovascular risk factors and an objective state of cognitive impairment were used as exclusion criteria. Cognitive function was evaluated using a scientific-validated neuropsychological battery, whereas MCI was recognized according to the Petersen criteria. Endothelial function was evaluated according to FMD from the brachial artery. The association between FMD and MCI was evaluated both by using a multivariate analysis and a correlation test. Finally, using the ANOVA analysis of variance, we tested the differences in flow-mediated dilation among MCI subgroups. RESULTS Brachial FMD was significantly associated with MCI (p < 0.01). The multivariate analysis showed that age, years of education and MMSE independently predicted the FMD variation (r (2) = 0.73; p < 0.0001). In addition, MCI patients with prevalent amnestic multiple domain impairment showed the worst brachial FMD. CONCLUSIONS This finding suggests that vascular dysfunction may play a role in the pathogenesis of cognitive impairment and underlines the lack of therapeutic strategies targeted to such dysfunctions.
Collapse
|
274
|
Tamashiro-Duran JH, Squarzoni P, de Souza Duran FL, Curiati PK, Vallada HP, Buchpiguel CA, Lotufo PA, Wajngarten M, Menezes PR, Scazufca M, de Toledo Ferraz Alves TC, Busatto GF. Cardiovascular risk in cognitively preserved elderlies is associated with glucose hypometabolism in the posterior cingulate cortex and precuneus regardless of brain atrophy and apolipoprotein gene variations. AGE (DORDRECHT, NETHERLANDS) 2013; 35:777-792. [PMID: 22544617 PMCID: PMC3636408 DOI: 10.1007/s11357-012-9413-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 04/17/2012] [Indexed: 05/31/2023]
Abstract
Cardiovascular risk factors (CVRF) possibly contribute to the emergence of Alzheimer's disease (AD). Fluorodeoxyglucose-positron emission tomography (FDG-PET) has been widely used to demonstrate specific patterns of reduced cerebral metabolic rates of glucose (CMRgl) in subjects with AD and in non-demented carriers of the apolipoprotein ε4 (APOE ε4) allele, the major genetic risk factor for AD. However, functional neuroimaging studies investigating the impact of CVRF on cerebral metabolism have been scarce to date. The present FDG-PET study investigated 59 cognitively preserved elderlies divided into three groups according to their cardiovascular risk based on the Framingham 10-year risk Coronary Heart Disease Risk Profile (low-, medium-, and high-risk) to examine whether different levels of CVRF would be associated with reduced CMRgl, involving the same brain regions affected in early stages of AD. Functional imaging data were corrected for partial volume effects to avoid confounding effects due to regional brain atrophy, and all analyses included the presence of the APOE ε4 allele as a confounding covariate. Significant cerebral metabolism reductions were detected in the high-risk group when compared to the low-risk group in the left precuneus and posterior cingulate gyrus. This suggests that findings of brain hypometabolism similar to those seen in subjects with AD can be detected in association with the severity of cardiovascular risk in cognitively preserved individuals. Thus, a greater knowledge about how such factors influence brain functioning in healthy subjects over time may provide important insigths for the future development of strategies aimed at delaying or preventing the vascular-related triggering of pathologic brain changes in the AD.
Collapse
Affiliation(s)
- Jaqueline Hatsuko Tamashiro-Duran
- Laboratory of Neuroimaging in Psychiatry (LIM-21), Department of Psychiatry, University of Sao Paulo Medical School, Sao Paulo, Sao Paulo, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
275
|
Tripathy D, Sanchez A, Yin X, Luo J, Martinez J, Grammas P. Thrombin, a mediator of cerebrovascular inflammation in AD and hypoxia. Front Aging Neurosci 2013; 5:19. [PMID: 23675346 PMCID: PMC3648692 DOI: 10.3389/fnagi.2013.00019] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 04/09/2013] [Indexed: 12/16/2022] Open
Abstract
Considerable evidence implicates hypoxia and vascular inflammation in Alzheimer's disease (AD). Thrombin, a multifunctional inflammatory mediator, is demonstrable in the brains of AD patients both in the vessel walls and senile plaques. Hypoxia-inducible factor 1α (HIF-1α), a key regulator of the cellular response to hypoxia, is also upregulated in the vasculature of human AD brains. The objective of this study is to investigate inflammatory protein expression in the cerebrovasculature of transgenic AD mice and to explore the role of thrombin as a mediator of cerebrovascular inflammation and oxidative stress in AD and in hypoxia-induced changes in brain endothelial cells. Immunofluorescent analysis of the cerebrovasculature in AD mice demonstrates significant (p < 0.01–0.001) increases in thrombin, HIF-1α, interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), matrix metalloproteinases (MMPs), and reactive oxygen species (ROS) compared to controls. Administration of the thrombin inhibitor dabigatran (100 mg/kg) to AD mice for 34 weeks significantly decreases expression of inflammatory proteins and ROS. Exposure of cultured brain endothelial cells to hypoxia for 6 h causes an upregulation of thrombin, HIF-1α, MCP-1, IL-6, and MMP2 and ROS. Treatment of endothelial cells with the dabigatran (1 nM) reduces ROS generation and inflammatory protein expression (p < 0.01–0.001). The data demonstrate that inhibition of thrombin in culture blocks the increase in inflammatory protein expression and ROS generation evoked by hypoxia. Also, administration of dabigatran to transgenic AD mice diminishes ROS levels in brain and reduces cerebrovascular expression of inflammatory proteins. Taken together, these results suggest that inhibiting thrombin generation could have therapeutic value in AD and other disorders where hypoxia, inflammation, and oxidative stress are involved.
Collapse
Affiliation(s)
- Debjani Tripathy
- Garrison Institute on Aging, Department of Neurology, Texas Tech University Health Sciences Center Lubbock, TX, USA
| | | | | | | | | | | |
Collapse
|
276
|
Jonsdottir G, Ingolfsdottir IE, Thormodsson FR, Petersen PH. Endogenous aggregates of amyloidogenic cystatin C variant are removed by THP-1 cells in vitro and induce differentiation and a proinflammatory response. Neurobiol Aging 2013; 34:1389-96. [DOI: 10.1016/j.neurobiolaging.2012.11.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 11/05/2012] [Accepted: 11/22/2012] [Indexed: 12/16/2022]
|
277
|
De Marinis E, Acaz-Fonseca E, Arevalo MA, Ascenzi P, Fiocchetti M, Marino M, Garcia-Segura LM. 17β-Oestradiol anti-inflammatory effects in primary astrocytes require oestrogen receptor β-mediated neuroglobin up-regulation. J Neuroendocrinol 2013. [PMID: 23190172 DOI: 10.1111/jne.12007] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neuroglobin (Ngb), so named after its initial discovery in brain neurones, has received great attention as a result of its neuroprotective effects both in vitro and in vivo. Recently, we demonstrated that, in neurones, Ngb is a 17β-oestradiol (E(2) ) inducible protein that is pivotal for hormone-induced anti-apoptotic effects against H(2) O(2) toxicity. The involvement of Ngb in other brain cell populations, as well as in other neuroprotective effects of E(2) , is completely unknown at present. We demonstrate Ngb immunoreactivity in reactive astrocytes located in the proximity of a penetrating cortical injury in vivo and the involvement of Ngb in the E(2) -mediated anti-inflammatory effect in primary cortical astrocytes. Upon binding to oestrogen receptor (ER)β, E(2) enhances Ngb levels in a dose-dependent manner. Although with a lesser degree than E(2) , the pro-inflammatory stimulation with lipopolysaccharide (LPS) also induces the increase of Ngb protein levels via nuclear factor-(NF)κB signal(s). Moreover, a negative cross-talk between ER subtypes and NFκB signal(s) has been demonstrated. In particular, ERα-activated signals prevent the NFκB-mediated Ngb increase, whereas LPS impairs the ERβ-induced up-regulation of Ngb. Therefore, the co-expression of both ERα and ERβ is pivotal for mediating E(2) -induced Ngb expression in the presence of NFκB-activated signals. Interestingly, Ngb silencing prevents the effect of E(2) on the expression of inflammatory markers (i.e. interleukin 6 and interferon γ-inducible protein 10). Ngb can be regarded as a key mediator of the different protective effects of E(2) in the brain, including protection against oxidative stress and the control of inflammation, both of which are at the root of several neurodegenerative diseases.
Collapse
Affiliation(s)
- E De Marinis
- Department of Biology, University Roma Tre, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
278
|
Achyuta AKH, Conway AJ, Crouse RB, Bannister EC, Lee RN, Katnik CP, Behensky AA, Cuevas J, Sundaram SS. A modular approach to create a neurovascular unit-on-a-chip. LAB ON A CHIP 2013; 13:542-53. [PMID: 23108480 DOI: 10.1039/c2lc41033h] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
In this work, we describe the fabrication and working of a modular microsystem that recapitulates the functions of the "Neurovascular Unit". The microdevice comprised a vertical stack of a poly(dimethylsiloxane) (PDMS) neural parenchymal chamber separated by a vascular channel via a microporous polycarbonate (PC) membrane. The neural chamber housed a mixture of neurons (~4%), astrocytes (~95%), and microglia (~1%). The vascular channel was lined with a layer of rat brain microvascular endothelial cell line (RBE4). Cellular components in the neural chamber and vascular channel showed viability (>90%). The neural cells fired inhibitory as well as excitatory potentials following 10 days of culture. The endothelial cells showed diluted-acetylated low density lipoprotein (dil-a-LDL) uptake, expressed von Willebrand factor (vWF) and zonula occludens (ZO-1) tight junctions, and showed decreased Alexafluor™-conjugated dextran leakage across their barriers significantly compared with controls (p < 0.05). When the vascular layer was stimulated with TNF-α for 6 h, about 75% of resident microglia and astrocytes on the neural side were activated significantly (p < 0.05 compared to controls) recapitulating tissue-mimetic responses resembling neuroinflammation. The impact of this microsystem lies in the fact that this biomimetic neurovascular platform might not only be harnessed for obtaining mechanistic insights for neurodegenerative disorders, but could also serve as a potential screening tool for central nervous system (CNS) therapeutics in toxicology and neuroinfectious diseases.
Collapse
Affiliation(s)
- Anil Kumar H Achyuta
- The Charles Stark Draper Laboratory, Bioengineering Center, 3802 Spectrum Blvd. Suite 201, Tampa, FL, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
279
|
Nilsson K, Gustafson L, Hultberg B. Elevated plasma homocysteine level in vascular dementia reflects the vascular disease process. Dement Geriatr Cogn Dis Extra 2013; 3:16-24. [PMID: 23569455 PMCID: PMC3618029 DOI: 10.1159/000345981] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Patients with vascular dementia (VaD) exhibit particularly elevated levels of plasma total homocysteine (tHcy) compared to patients with other psychogeriatric diseases. Methods We investigated the main determinants (age, renal impairment, cobalamin/folate status and presence of extracerebral vascular disease) of plasma tHcy in 525 patients with VaD. Furthermore, 270 patients with depression were used as a reference group to reveal the potential specificity of elevated plasma tHcy in patients with VaD. Results Elevated plasma tHcy levels in patients with VaD could only partly be attributed to cobalamin/folate deficiency or renal impairment. Plasma tHcy might also be related to the vascular disease process since patients with depression and vascular disease exhibited similar plasma tHcy levels to patients with VaD. Conclusion Our findings suggest that elevated plasma tHcy might be a sensitive marker for the vascular disease process in patients with VaD and that the level also is a reflection of changes in the other main determinants of plasma tHcy.
Collapse
Affiliation(s)
- Karin Nilsson
- Department of Psychogeriatrics, Clinical Science, Lund University Hospital, Lund, Sweden
| | | | | |
Collapse
|
280
|
Association of COMT, MTHFR, and SLC19A1(RFC-1) polymorphisms with homocysteine blood levels and cognitive impairment in Parkinson's disease. Pharmacogenet Genomics 2013; 22:716-24. [PMID: 22890010 DOI: 10.1097/fpc.0b013e32835693f7] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Elevated plasma homocysteine (Hcy) concentration is an independent risk factor for cardiovascular disease, and its involvement in endothelial cell dysfunction is well established. However, the role of Hcy and folate in the pathogenesis of Parkinson's disease (PD) remains controversial. OBJECTIVES The study was aimed at evaluating the relationships between Hcy, vitamin B12, and folic acid levels in the blood and cognitive status in PD patients with the genetic polymorphisms of MTHFR (rs1801133: C>T-677C>T, rs1801131: A>C-1298A>C), COMT (rs4680: A>G-Val158Met, rs6269: A>G, rs4633: C>T, rs4818: C>G), or SLC19A1 (rs1051266: G>A-80G>A). METHODS A total of 502 participants (248 with PD and 254 age-matched and sex-matched controls) were included in the study. The Unified Parkinson's Disease Rating Scale score, Hoehn-Yahr staging, and the Schwab-England scale were used to assess motor abilities and activity during daily life. Complex psychological examination with a battery of tests was used to classify patients into groups with (PDD) and without (nPDD) dementia. Blood samples were examined for Hcy, vitamin B12, and folic acid levels, as well as polymorphisms in genes related to Hcy metabolism, such as COMT, MTHFR, and SLC19A1(RFC-1). RESULTS The frequency of homozygous COMT rs4680G and rs4633C allele carriers was significantly decreased in PD patients in comparison with the controls (P=0.015; odds ratio=0.60; 95% confidence interval 0.41-0.90 and P=0.020; odds ratio=0.619; 95% confidence interval 0.42-0.92, respectively). No significant differences in the distribution of MTHFR 677C>T, 1298A>C, and SLC19A1 80G>A alleles and genotypes between PD patients and the controls were found. Hcy levels were significantly increased in PD patients (18±7.8 μmol/l) as compared with the controls (14.0±9.6 μmol/l, P=10(-8)) and were significantly associated with the MTHFR 677C>T polymorphism both in PD patients and controls, in which T allele carriers were characterized by markedly elevated Hcy plasma concentrations. No association was observed between Hcy plasma level and COMT and SLC19A polymorphisms. The results of multivariate logistic regression analysis revealed age (P=0.0003) and Hcy plasma levels (P=0.07) as independent risk factors predisposing individuals to PD dementia. The studied polymorphisms were not associated with cognitive status in PD patients. CONCLUSION The genetic factors studied were not associated with cognitive status in PD patients. Only age and Hcy plasma levels were found to be independent risk factors predisposing individuals to PD dementia. However, COMT: rs4680: A>G and rs4633: C>T polymorphisms were found to significantly affect PD risk, and the MTHFR 677C>T polymorphism helped determine plasma Hcy concentrations.
Collapse
|
281
|
Ozkan S, Adapinar DO, Elmaci NT, Arslantas D. Apraxia for differentiating Alzheimer's disease from subcortical vascular dementia and mild cognitive impairment. Neuropsychiatr Dis Treat 2013; 9:947-51. [PMID: 23882142 PMCID: PMC3709829 DOI: 10.2147/ndt.s47879] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Although ideomotor limb apraxia is considered to be a typical sign of cortical pathologies such as Alzheimer's disease (AD), it has been also reported in subcortical neurodegenerative diseases and vascular lesions. We aimed to investigate the difference between AD, subcortical vascular dementia (SVaD) and mild cognitive impairment (MCI) patients by means of ideomotor limb apraxia frequency and severity. Ninety-six AD, 72 SVaD, and 84 MCI patients were assessed with the mini-mental status examination (MMSe), clinical dementia rating (CDR) and the apraxia screening test of TULIA (AST). Apraxia was significantly more frequent in the AD patients (32.3%) than in both of the SVaD (16.7%) and MCI (4.8%) patients. The frequency of apraxia was also significantly higher in SVaD patients than in MCI patients. AD patients had significantly lower apraxia scores than both SVaD and MCI patients. In addition, a significant difference was found between SVaD and MCI patients in terms of apraxia scores. These results suggest that the widespread belief of the association between apraxia and cortical dementias is not exactly correct. The significant difference between both of the dementia groups and the MCI patients suggests that the absence of apraxia can be an indicator for MCI diagnosis.
Collapse
Affiliation(s)
- Serhat Ozkan
- Department of Neurology, Eskişehir Osmangazi University Medical Faculty, Eskisehir, Turkey
| | | | | | | |
Collapse
|
282
|
Sagare AP, Bell RD, Zlokovic BV. Neurovascular defects and faulty amyloid-β vascular clearance in Alzheimer's disease. J Alzheimers Dis 2013; 33 Suppl 1:S87-100. [PMID: 22751174 PMCID: PMC4416477 DOI: 10.3233/jad-2012-129037] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The evidence that neurovascular dysfunction is an integral part of Alzheimer's disease (AD) pathogenesis has continued to emerge in the last decade. Changes in the brain vasculature have been shown to contribute to the onset and progression of the pathological processes associated with AD, such as microvascular reductions, blood brain barrier (BBB) breakdown, and faulty clearance of amyloid β-peptide (Aβ) from the brain. Herein, we review the role of the neurovascular unit and molecular mechanisms in cerebral vascular cells behind the pathogenesis of AD. In particular, we focus on molecular pathways within cerebral vascular cells and the systemic circulation that contribute to BBB dysfunction, brain hypoperfusion, and impaired clearance of Aβ from the brain. We aim to provide a summary of recent research findings implicated in neurovascular defects and faulty Aβ vascular clearance contributing to AD pathogenesis.
Collapse
Affiliation(s)
- Abhay P. Sagare
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robert D. Bell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Berislav V. Zlokovic
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
283
|
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with no available disease-modifying drugs. However, it has been postulated that neurovascular damage is a primary occurrence in this disease. Neurovascular damage is the result of the presence of cardiovascular risk factor generating hypoxia, oxidative stress, and metabolic changes that activate the endothelial cells of the brain microvasculature in order to respond to the stress by the development of angiogenesis. This endothelial activation could lead to a secretion of many proinflammatory cytokines and growth factors, such as thrombin. Heparin and related oligosaccharides have been shown to be efficient in the improvement of symptoms of AD. Their efficacy may be limited by their nonselective inhibitory effect of thrombin's activity. Direct thrombin inhibitors, such as dabigatran, might be efficient in the treatment of patients with AD because of their high selectivity for thrombin's activity inhibition while having a safer side effects profile than heparin.
Collapse
Affiliation(s)
- Bou Khalil Rami
- Psychiatric Hospital of the Cross, Saint Joseph University, Beirut, Lebanon.
| |
Collapse
|
284
|
Currais A, Prior M, Lo D, Jolivalt C, Schubert D, Maher P. Diabetes exacerbates amyloid and neurovascular pathology in aging-accelerated mice. Aging Cell 2012; 11:1017-26. [PMID: 22938075 DOI: 10.1111/acel.12002] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mounting evidence supports a link between diabetes, cognitive dysfunction, and aging. However, the physiological mechanisms by which diabetes impacts brain function and cognition are not fully understood. To determine how diabetes contributes to cognitive dysfunction and age-associated pathology, we used streptozotocin to induce type 1 diabetes (T1D) in senescence-accelerated prone 8 (SAMP8) and senescence-resistant 1 (SAMR1) mice. Contextual fear conditioning demonstrated that T1D resulted in the development of cognitive deficits in SAMR1 mice similar to those seen in age-matched, nondiabetic SAMP8 mice. No further cognitive deficits were observed when the SAMP8 mice were made diabetic. T1D dramatically increased Aβ and glial fibrillary acidic protein immunoreactivity in the hippocampus of SAMP8 mice and to a lesser extent in age-matched SAMR1 mice. Further analysis revealed aggregated Aβ within astrocyte processes surrounding vessels. Western blot analyses from T1D SAMP8 mice showed elevated amyloid precursor protein processing and protein glycation along with increased inflammation. T1D elevated tau phosphorylation in the SAMR1 mice but did not further increase it in the SAMP8 mice where it was already significantly higher. These data suggest that aberrant glucose metabolism potentiates the aging phenotype in old mice and contributes to early stage central nervous system pathology in younger animals.
Collapse
Affiliation(s)
- Antonio Currais
- The Salk Institute for Biological Studies, Laboratory of Cellular Neurobiology, 10010 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
285
|
Sengillo JD, Winkler EA, Walker CT, Sullivan JS, Johnson M, Zlokovic BV. Deficiency in mural vascular cells coincides with blood-brain barrier disruption in Alzheimer's disease. Brain Pathol 2012; 23:303-10. [PMID: 23126372 DOI: 10.1111/bpa.12004] [Citation(s) in RCA: 417] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Accepted: 10/30/2012] [Indexed: 12/13/2022] Open
Abstract
Neurovascular dysfunction contributes to Alzheimer's disease (AD). Cerebrovascular abnormalities and blood-brain barrier (BBB) damage have been shown in AD. The BBB dysfunction can lead to leakage of potentially neurotoxic plasma components in brain that may contribute to neuronal injury. Pericytes are integral in maintaining the BBB integrity. Pericyte-deficient mice develop a chronic BBB damage preceding neuronal injury. Moreover, loss of pericytes was associated with BBB breakdown in patients with amyotrophic lateral sclerosis. Here, we demonstrate a decrease in mural vascular cells in AD, and show that pericyte number and coverage in the cortex and hippocampus of AD subjects compared with neurologically intact controls are reduced by 59% and 60% (P < 0.01), and 32% and 33% (P < 0.01), respectively. An increase in extravascular immunoglobulin G (IgG) and fibrin deposition correlated with reductions in pericyte coverage in AD cases compared with controls; the Pearson's correlation coefficient r for the magnitude of BBB breakdown to IgG and fibrin vs. reduction in pericyte coverage was -0.96 (P < 0.01) and -0.81 (P < 0.01) in the cortex, respectively, and -0.86 (P < 0.01) and -0.98 (P < 0.01) in the hippocampus, respectively. Thus, deficiency in mural vascular cells may contribute to disrupted vascular barrier properties and resultant neuronal dysfunction during AD pathogenesis.
Collapse
Affiliation(s)
- Jesse D Sengillo
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
286
|
Burke BA, Diamond SG. Measuring cerebral hemodynamics with a modified magnetoencephalography system. Physiol Meas 2012; 33:2079-98. [PMID: 23171539 DOI: 10.1088/0967-3334/33/12/2079] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Magnetoencephalography (MEG) systems are designed to noninvasively measure magnetic fields produced by neural electrical currents. This project examines the possibility of measuring hemodynamics with an MEG system that has been modified with dc electromagnets to measure magnetic susceptibility while maintaining the capability of measuring neural dynamics. A forward model is presented that simulates the interaction of an applied magnetic field with changes in magnetic susceptibility in the brain associated with hemodynamics. Model predictions are compared with an experiment where deionized water was pumped into an inverted flask under the MEG sensor array of superconducting quantum interference device (SQUID) gradiometers (R(2) = 0.98, p < 0.001). The forward model was used to simulate the SQUID readouts from hemodynamics in the scalp and brain induced by performing the Valsalva maneuver. Experimental human subject recordings (N = 10) were made from the prefrontal region during Valsalva using concurrent measurement with the modified MEG system and near-infrared spectroscopy (NIRS). The NIRS deoxyhemoglobin signal was found to correlate significantly with the SQUID readouts (R(2) = 0.84, p < 0.01). SQUID noise was found to increase with the applied field, which will need to be mitigated in future work. These results demonstrate the potential and technical challenges of measuring cerebral hemodynamics with a modified MEG system.
Collapse
Affiliation(s)
- Broc A Burke
- Thayer School of Engineering at Dartmouth, Hanover, NH, USA
| | | |
Collapse
|
287
|
Tripathy D, Sanchez A, Yin X, Martinez J, Grammas P. Age-related decrease in cerebrovascular-derived neuroprotective proteins: effect of acetaminophen. Microvasc Res 2012; 84:278-85. [PMID: 22944728 PMCID: PMC3483357 DOI: 10.1016/j.mvr.2012.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 07/17/2012] [Accepted: 08/17/2012] [Indexed: 12/19/2022]
Abstract
As the population ages, the need for effective methods to maintain brain function in older adults is increasingly pressing. Vascular disease and neurodegenerative disorders commonly co-occur in older persons. Cerebrovascular products contribute to the neuronal milieu and have important consequences for neuronal viability. In this regard vascular derived neuroprotective proteins, Such as vascular endothelial growth factor (VEGF), pigment epithelium-derived factor (PEDF), and pituitary adenylate cyclase activating peptide (PACAP) are important for maintaining neuronal viability, especially in the face of injury and disease. The objective of this study is to measure and compare levels of VEGF, PEDF and PACAP released from isolated brain microvessels of Fischer 344 rats at 6, 12, 18, and 24 months of age. Addition of acetaminophen to isolated brain microvessels is employed to determine whether this drug affects vascular expression of these neuroprotective proteins. Experiments on cultured brain endothelial cells are performed to explore the mechanisms/mediators that regulate the effect of acetaminophen on endothelial cells. The data indicate cerebrovascular expression of VEGF, PEDF and PACAP significantly decreases with age. The age-associated decrease in VEGF and PEDF is ameliorated by addition of acetaminophen to isolated brain microvessels. Also, release of VEGF, PEDF, and PACAP from cultured brain endothelial cells decreases with exposure to the oxidant stressor menadione. Acetaminophen treatment upregulates VEGF, PEDF and PACAP in brain endothelial cells exposed to oxidative stress. The effect of acetaminophen on cultured endothelial cells is in part inhibited by the selective thrombin inhibitor hirudin. The results of this study suggest that acetaminophen may be a useful agent for preserving cerebrovascular function. If a low dose of acetaminophen can counteract the decrease in vascular-derived neurotrophic factors evoked by age and oxidative stress, this drug might be useful for improving brain function in the elderly.
Collapse
Affiliation(s)
- Debjani Tripathy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Alma Sanchez
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Xiangling Yin
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Joseph Martinez
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Paula Grammas
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, Texas
| |
Collapse
|
288
|
Østergaard L, Aamand R, Gutiérrez-Jiménez E, Ho YCL, Blicher JU, Madsen SM, Nagenthiraja K, Dalby RB, Drasbek KR, Møller A, Brændgaard H, Mouridsen K, Jespersen SN, Jensen MS, West MJ. The capillary dysfunction hypothesis of Alzheimer's disease. Neurobiol Aging 2012; 34:1018-31. [PMID: 23084084 DOI: 10.1016/j.neurobiolaging.2012.09.011] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Revised: 08/17/2012] [Accepted: 09/07/2012] [Indexed: 12/21/2022]
Abstract
It is widely accepted that hypoperfusion and changes in capillary morphology are involved in the etiopathogenesis of Alzheimer's disease (AD). This is difficult to reconcile with the hyperperfusion observed in young high-risk subjects. Differences in the way cerebral blood flow (CBF) is coupled with the local metabolic needs during different phases of the disease can explain this apparent paradox. This review describes this coupling in terms of a model of cerebral oxygen availability that takes into consideration the heterogeneity of capillary blood flow patterns. The model predicts that moderate increases in heterogeneity requires elevated CBF in order to maintain adequate oxygenation. However, with progressive increases in heterogeneity, the resulting low tissue oxygen tension will require a suppression of CBF in order to maintain tissue metabolism. The observed biphasic nature of CBF responses in preclinical AD and AD is therefore consistent with progressive disturbances of capillary flow patterns. Salient features of the model are discussed in the context of AD pathology along with potential sources of increased capillary flow heterogeneity.
Collapse
Affiliation(s)
- Leif Østergaard
- Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University, Aarhus, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
289
|
Elevated angiopoietin-1 serum levels in patients with Alzheimer's disease. Int J Alzheimers Dis 2012; 2012:324016. [PMID: 23094194 PMCID: PMC3474986 DOI: 10.1155/2012/324016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 09/13/2012] [Indexed: 12/17/2022] Open
Abstract
Background. Alzheimer's disease (AD) is the most common cause of dementia in the elderly. AD is characterized by the accumulation of amyloid plaques and neurofibrillary tangles and by massive neuronal loss in the brain. There is epidemiologic and pathologic evidence that AD is associated with vascular risk factors and vascular diseases, contributing to cerebral hypoperfusion with consecutive stimulation of angiogenesis and upregulation of proangiogenic factors such as Angiopoietin-1 (Ang-1). Methods. In the present study, we measured Ang-1 serum levels in 42 patients with AD, 20 patients with mild cognitive impairment (MCI), and in 40 healthy elderly controls by ELISA. Results. We found significantly increased Ang-1 serum levels in patients with AD compared to control subjects (P = 0.003). There was no significant difference between MCI patients and healthy controls (P = 0.553) or between AD and MCI patients (P = 0.054). The degree of cognitive impairment as measured by the mini-mental status examination (MMSE) score was significantly correlated with the Ang-1 serum levels in all patients and healthy controls. Conclusions. We found significantly increased Ang-1 serum levels in AD patients. We could also show an association between Ang-1 serum levels and the cognitive status in all patients and healthy controls. Thus, serum Ang-1 could be a potential candidate for a biomarker panel for AD diagnosis.
Collapse
|
290
|
Carrero I, Gonzalo M, Martin B, Sanz-Anquela J, Arévalo-Serrano J, Gonzalo-Ruiz A. Oligomers of beta-amyloid protein (Aβ1-42) induce the activation of cyclooxygenase-2 in astrocytes via an interaction with interleukin-1beta, tumour necrosis factor-alpha, and a nuclear factor kappa-B mechanism in the rat brain. Exp Neurol 2012; 236:215-27. [DOI: 10.1016/j.expneurol.2012.05.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Revised: 04/22/2012] [Accepted: 05/05/2012] [Indexed: 11/25/2022]
|
291
|
Pimentel-Coelho PM, Michaud JP, Rivest S. Effects of mild chronic cerebral hypoperfusion and early amyloid pathology on spatial learning and the cellular innate immune response in mice. Neurobiol Aging 2012; 34:679-93. [PMID: 22819135 DOI: 10.1016/j.neurobiolaging.2012.06.025] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 06/05/2012] [Accepted: 06/28/2012] [Indexed: 12/19/2022]
Abstract
Understanding the contribution of cerebrovascular factors in the progression of cognitive decline in Alzheimer's disease (AD) is a key step for the development of preventive therapies. Among these factors, chronic cerebral hypoperfusion is an early component of AD pathogenesis that can predict the progression from mild cognitive impairment to AD. Here, we investigated the effects of a protocol of mild chronic cerebral hypoperfusion in the APPswe/PS1 transgenic mouse model of AD. We observed that the permanent occlusion of the right common carotid artery induced spatial learning impairments in young APPswe/PS1 mice, but not in their wild type littermates. Furthermore, the extent of learning deficits strongly correlated with the number of cortical β-amyloid plaques, with the mobilization of monocytes into the blood and with the number of bone marrow-derived microglia in the brain. These results indicate that a mild reduction of cerebral blood flow can selectively induce cognitive deficits at an early stage of amyloid pathology, eliciting a cellular innate immune response, even without causing neuronal death.
Collapse
Affiliation(s)
- Pedro M Pimentel-Coelho
- Laboratory of Endocrinology and Genomics, CHUQ (CHUL) Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, Canada
| | | | | |
Collapse
|
292
|
Lapchak PA. Transcranial near-infrared laser therapy applied to promote clinical recovery in acute and chronic neurodegenerative diseases. Expert Rev Med Devices 2012; 9:71-83. [PMID: 22145842 DOI: 10.1586/erd.11.64] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
One of the most promising methods to treat neurodegeneration is noninvasive transcranial near-infrared laser therapy (NILT), which appears to promote acute neuroprotection by stimulating mitochondrial function, thereby increasing cellular energy production. NILT may also promote chronic neuronal function restoration via trophic factor-mediated plasticity changes or possibly neurogenesis. Clearly, NILT is a treatment that confers neuroprotection or neurorestoration using pleiotropic mechanisms. The most advanced application of NILT is for acute ischemic stroke based upon extensive preclinical and clinical studies. In laboratory settings, NILT is also being developed to treat traumatic brain injury, Alzheimer's disease and Parkinson's disease. There is some intriguing data in the literature that suggests that NILT may be a method to promote clinical improvement in neurodegenerative diseases where there is a common mechanistic component, mitochondrial dysfunction and energy impairment. This article will analyze and review data supporting the continued development of NILT to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Paul A Lapchak
- Cedars-Sinai Medical Center, Department of Neurology, Los Angeles, CA 90048, USA.
| |
Collapse
|
293
|
Mattsson MO, Simkó M. Is there a relation between extremely low frequency magnetic field exposure, inflammation and neurodegenerative diseases? A review of in vivo and in vitro experimental evidence. Toxicology 2012; 301:1-12. [PMID: 22750629 DOI: 10.1016/j.tox.2012.06.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 05/25/2012] [Accepted: 06/17/2012] [Indexed: 12/21/2022]
Abstract
Possible health consequences of exposure to extremely low frequency magnetic fields (ELF-MF) have received considerable interest during the last decades. One area of concern is neurodegenerative diseases (NDD), where epidemiological evidence suggests a correlation between MF exposure and Alzheimer's disease (AD). This review is focussing on animal and in vitro studies employing ELF-MF exposures to see if there is mechanistic support for any causal connection between NDD and MF-exposure. The hypothesis is that ELF-MF exposure can promote inflammation processes and thus influence the progression of NDD. A firm conclusion regarding this hypothesis is difficult to draw based on available studies, since there is a lack of experimental studies that have addressed the question of ELF-MF exposure and NDD. Furthermore, the heterogeneity of the performed studies regarding, e.g., the exposure duration, the flux density, the biological endpoint and the cell type and the time point of investigation is substantial and makes conclusions difficult to draw. Nevertheless, the investigated evidence from in vivo and in vitro studies suggest that short-term MF-exposure causes mild oxidative stress (modest ROS increases and changes in antioxidant levels) and possibly activates anti-inflammatory processes (decrease in pro-inflammatory and increase in anti-inflammatory cytokines). The few studies that specifically have investigated NDDs or NDD relevant end-points show that effects of exposure are either lacking or indicating positive effects on neuronal viability and differentiation. In both immune and NDD relevant studies, experiments with realistic long-term exposures are lacking. Importantly, consequences of a possible long-lasting mild oxidative stress are thus not investigated. In summary, the existing experimental studies are not adequate in answering if there is a causal relationship between MF-exposure and AD, as suggested in epidemiological studies.
Collapse
Affiliation(s)
- Mats-Olof Mattsson
- AIT Austrian Institute of Technology, Health and Environment Department, Environmental Resources and Technologies, Konrad-Lorenz-Strasse 24, AT-3430 Tulln, Austria.
| | | |
Collapse
|
294
|
Tang Z, Chen F, Huang J, Shi L, Gong HT, Fu H, Qu Z, Pi X. Low-dose cerebral CT perfusion imaging (CTPI) of senile dementia: diagnostic performance. Arch Gerontol Geriatr 2012; 56:61-7. [PMID: 22704835 DOI: 10.1016/j.archger.2012.05.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 05/04/2012] [Accepted: 05/19/2012] [Indexed: 10/28/2022]
Abstract
To evaluate the clinical value of low-dose cerebral CTPI in the diagnosis of senile dementia, as an attempt to develop a new imaging method to diagnose this disease and measure its severity. 95 patients of senile dementia (52 with Alzheimer's disease (AD), 43 with vascular dementia (VD)) and 30 healthy subjects (control group) were underwent low-dose cerebral CTPI examinations with multi-slices spiral CT. The CTPI images were analyzed using perfusion software. Derived perfusion parameters including cerebral blood volume (CBV), cerebral blood flow (CBF), mean transit time (MTT) and time to peak (TTP) were measured. Diagnostic value of perfusion parameters was evaluated by receiver-operating characteristic (ROC) curve. The CBV and CBF of both of frontal lobe, temporal lobe, hippocampus and basal ganglial area in the patients with senile dementia were much lower than those in the healthy group. And MTT and TTP of the areas above-mentioned in the senile dementia group were higher than those in the control group. There was statistically significant difference between them (p<0.05). The areas under ROC curves of perfusion parameters left temporal lobe MTT, left the basal ganglia MTT and left the hippocampus MTT to diagnose senile dementia were 0.959, 0.920, 0.916, and diagnostic accuracy rate is higher. The areas under ROC curve of the left frontal MTT, the left basal ganglia CBV were 0.867 and 0.819, diagnosis accuracy medium. The results showed that cerebral CTPI is valuable for the diagnosis of senile dementia.
Collapse
Affiliation(s)
- Zhen Tang
- Department of Radiology, Branch Hospital in Fengxian of Shanghai No. 6 People's Hospital, Shanghai 201400, China
| | | | | | | | | | | | | | | |
Collapse
|
295
|
Darvesh AS, Carroll RT, Bishayee A, Novotny NA, Geldenhuys WJ, Van der Schyf CJ. Curcumin and neurodegenerative diseases: a perspective. Expert Opin Investig Drugs 2012; 21:1123-40. [DOI: 10.1517/13543784.2012.693479] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
296
|
Eikelenboom P, Hoozemans JJ, Veerhuis R, van Exel E, Rozemuller AJ, van Gool WA. Whether, when and how chronic inflammation increases the risk of developing late-onset Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2012; 4:15. [PMID: 22647384 PMCID: PMC3506930 DOI: 10.1186/alzrt118] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neuropathological studies have revealed the presence of a broad variety of inflammation-related proteins (complement factors, acute-phase proteins, pro-inflammatory cytokines) in Alzheimer's disease (AD) brains. These constituents of innate immunity are involved in several crucial pathogenic events of the underlying pathological cascade in AD, and recent studies have shown that innate immunity is involved in the etiology of late-onset AD. Genome-wide association studies have demonstrated gene loci that are linked to the complement system. Neuropathological and experimental studies indicate that fibrillar amyloid-β (Aβ) can activate the innate immunity-related CD14 and Toll-like receptor signaling pathways of glial cells for pro-inflammatory cytokine production. The production capacity of this pathway is under genetic control and offspring with a parental history of late-onset AD have a higher production capacity for pro-inflammatory cytokines. The activation of microglia by fibrillar Aβ deposits in the early preclinical stages of AD can make the brain susceptible later on for a second immune challenge leading to enhanced production of pro-inflammatory cytokines. An example of a second immune challenge could be systemic inflammation in patients with preclinical AD. Prospective epidemiological studies show that elevated serum levels of acute phase reactants can be considered as a risk factor for AD. Clinical studies suggest that peripheral inflammation increases the risk of dementia, especially in patients with preexistent cognitive impairment, and accelerates further deterioration in demented patients. The view that peripheral inflammation can increase the risk of dementia in older people provides scope for prevention.
Collapse
Affiliation(s)
- Piet Eikelenboom
- Valeriuskliniek, Valeriusplein 9, 1075 BG Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
297
|
Tang Z, Pi X, Chen F, Shi L, Gong H, Fu H, Qu Z. Fifty percent reduced-dose cerebral CT perfusion imaging of Alzheimer's disease: regional blood flow abnormalities. Am J Alzheimers Dis Other Demen 2012; 27:267-74. [PMID: 22615483 PMCID: PMC10697383 DOI: 10.1177/1533317512447885] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
To evaluate the value of 50% reduced-dose cerebral computed tomography (CT)perfusion imaging (CTPI) to show the perfusion abnormalities in Alzheimer's disease (AD), as an attempt to develop a new imaging protocol with lower radiation dose to track the correlation of AD with regional blood flow abnormalities. A total of 52 patients with AD were assigned to the AD group and 28 healthy volunteers served as the control group. All participants were given a 50% reduced-dose cerebral CTPI (current was reduced from 160 to 80 mA) test by a multislice spiral CT scanner. Perfusion parameters of the bilateral frontal cortex, temporal cortex, hippocampus, and basal ganglia were measured, including the cerebral blood volume (CBV), cerebral blood flow (CBF), mean transit time (MTT), and time to peak (TTP). Both the CBV and CBF values of the measured regions were significantly higher in the healthy control group than in the AD group (P < .05), while the MTT and TTP values of these cerebral areas were significantly lower in the healthy control group than in the AD group (P < .05). Four perfusion parameters, namely the MTT of the left frontal cortex, right temporal cortex, right basal ganglia, and right hippocampus, had the greatest sensitivity and a striking correlation with the incidence of AD. The blood flow per unit of time in the regions of interest was significantly lower in the AD group, which provides new evidence for the existence of microcirculation disturbance and ischemia in AD. The 50% reduced-dose cerebral CTPI scan is valuable to show the regional perfusion abnormalities in the patients with AD.
Collapse
Affiliation(s)
- Zhen Tang
- Department of Radiology, Branch Hospital in Fengxian of Shanghai No 6 People’s Hospital, Shanghai, China
| | - Xiaoling Pi
- Department of Internal Medicine, Pudong New District Gongli Hospital, Shanghai, China
| | - Feng Chen
- Department of Radiology, Branch Hospital in Fengxian of Shanghai No 6 People’s Hospital, Shanghai, China
| | - Linghua Shi
- Department of Radiology, Pudong New District Gongli Hospital, Shanghai, China
| | - Haiting Gong
- Department of Radiology, Huashan Hospital Fudan University Jing’ An Branch, Shanghai, China
| | - Hongmei Fu
- Pudong New District Mental Health Center, Shanghai, China
| | - Zhengwan Qu
- Pudong New District Mental Health Center, Shanghai, China
| |
Collapse
|
298
|
Morphological and pathological evolution of the brain microcirculation in aging and Alzheimer's disease. PLoS One 2012; 7:e36893. [PMID: 22615835 PMCID: PMC3353981 DOI: 10.1371/journal.pone.0036893] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 04/12/2012] [Indexed: 01/01/2023] Open
Abstract
Key pathological hallmarks of Alzheimer's disease (AD), including amyloid plaques, cerebral amyloid angiopathy (CAA) and neurofibrillary tangles do not completely account for cognitive impairment, therefore other factors such as cardiovascular and cerebrovascular pathologies, may contribute to AD. In order to elucidate the microvascular changes that contribute to aging and disease, direct neuropathological staining and immunohistochemistry, were used to quantify the structural integrity of the microvasculature and its innervation in three oldest-old cohorts: 1) nonagenarians with AD and a high amyloid plaque load; 2) nonagenarians with no dementia and a high amyloid plaque load; 3) nonagenarians without dementia or amyloid plaques. In addition, a non-demented (ND) group (average age 71 years) with no amyloid plaques was included for comparison. While gray matter thickness and overall brain mass were reduced in AD compared to ND control groups, overall capillary density was not different. However, degenerated string capillaries were elevated in AD, potentially suggesting greater microvascular "dysfunction" compared to ND groups. Intriguingly, apolipoprotein ε4 carriers had significantly higher string vessel counts relative to non-ε4 carriers. Taken together, these data suggest a concomitant loss of functional capillaries and brain volume in AD subjects. We also demonstrated a trend of decreasing vesicular acetylcholine transporter staining, a marker of cortical cholinergic afferents that contribute to arteriolar vasoregulation, in AD compared to ND control groups, suggesting impaired control of vasodilation in AD subjects. In addition, tyrosine hydroxylase, a marker of noradrenergic vascular innervation, was reduced which may also contribute to a loss of control of vasoconstriction. The data highlight the importance of the brain microcirculation in the pathogenesis and evolution of AD.
Collapse
|
299
|
Bell RD, Winkler EA, Singh I, Sagare AP, Deane R, Wu Z, Holtzman DM, Betsholtz C, Armulik A, Sallstrom J, Berk BC, Zlokovic BV. Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature 2012; 485:512-6. [PMID: 22622580 PMCID: PMC4047116 DOI: 10.1038/nature11087] [Citation(s) in RCA: 958] [Impact Index Per Article: 73.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 03/26/2012] [Indexed: 12/15/2022]
Abstract
Human apolipoprotein E has three isoforms: APOE2, APOE3 and APOE4. APOE4 is a major genetic risk factor for Alzheimer's disease and is associated with Down's syndrome dementia and poor neurological outcome after traumatic brain injury and haemorrhage. Neurovascular dysfunction is present in normal APOE4 carriers and individuals with APOE4-associated disorders. In mice, lack of Apoe leads to blood-brain barrier (BBB) breakdown, whereas APOE4 increases BBB susceptibility to injury. How APOE genotype affects brain microcirculation remains elusive. Using different APOE transgenic mice, including mice with ablation and/or inhibition of cyclophilin A (CypA), here we show that expression of APOE4 and lack of murine Apoe, but not APOE2 and APOE3, leads to BBB breakdown by activating a proinflammatory CypA-nuclear factor-κB-matrix-metalloproteinase-9 pathway in pericytes. This, in turn, leads to neuronal uptake of multiple blood-derived neurotoxic proteins, and microvascular and cerebral blood flow reductions. We show that the vascular defects in Apoe-deficient and APOE4-expressing mice precede neuronal dysfunction and can initiate neurodegenerative changes. Astrocyte-secreted APOE3, but not APOE4, suppressed the CypA-nuclear factor-κB-matrix-metalloproteinase-9 pathway in pericytes through a lipoprotein receptor. Our data suggest that CypA is a key target for treating APOE4-mediated neurovascular injury and the resulting neuronal dysfunction and degeneration.
Collapse
Affiliation(s)
- Robert D Bell
- Center for Neurodegenerative and Vascular Brain Disorders, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
300
|
Banks WA. Drug delivery to the brain in Alzheimer's disease: consideration of the blood-brain barrier. Adv Drug Deliv Rev 2012; 64:629-39. [PMID: 22202501 DOI: 10.1016/j.addr.2011.12.005] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 12/09/2011] [Accepted: 12/09/2011] [Indexed: 12/21/2022]
Abstract
The successful treatment of Alzheimer's disease (AD) will require drugs that can negotiate the blood-brain barrier (BBB). However, the BBB is not simply a physical barrier, but a complex interface that is in intimate communication with the rest of the central nervous system (CNS) and influenced by peripheral tissues. This review examines three aspects of the BBB in AD. First, it considers how the BBB may be contributing to the onset and progression of AD. In this regard, the BBB itself is a therapeutic target in the treatment of AD. Second, it examines how the BBB restricts drugs that might otherwise be useful in the treatment of AD and examines strategies being developed to deliver drugs to the CNS for the treatment of AD. Third, it considers how drug penetration across the AD BBB may differ from the BBB of normal aging. In this case, those differences can complicate the treatment of CNS diseases such as depression, delirium, psychoses, and pain control in the AD population.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA , USA.
| |
Collapse
|