151
|
Foveal Avascular Zone and Choroidal Thickness Are Decreased in Subjects with Hard Drusen and without High Genetic Risk of Developing Alzheimer's Disease. Biomedicines 2021; 9:biomedicines9060638. [PMID: 34199664 PMCID: PMC8229973 DOI: 10.3390/biomedicines9060638] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/01/2021] [Accepted: 06/01/2021] [Indexed: 01/08/2023] Open
Abstract
A family history (FH+) of Alzheimer’s disease (AD) and ɛ4 allele of the ApoE gene are the main genetic risk factors for developing AD, whereas ɛ4 allele plays a protective role in age-related macular degeneration. Ocular vascular changes have been reported in both pathologies. We analyzed the choroidal thickness using optical coherence tomography (OCT) and the foveal avascular zone (FAZ) using OCT-angiography and compared the results with ApoE gene expression, AD FH+, and the presence or absence of hard drusen (HD) in 184 cognitively healthy subjects. Choroidal thickness was statistically significantly different in the (FH−, ɛ4−, HD+) group compared with (i) both the (FH−, ɛ4−, HD−) and the (FH+, ɛ4+, HD+) groups in the superior and inferior points at 1500 μm, and (ii) the (FH+, ɛ4−, HD+) group in the superior point at 1500 μm. There were statistically significant differences in the superficial FAZ between the (FH+, ɛ4−, HD+) group and (i) the (FH+, ɛ4−, HD−) group and (ii) the (FH+, ɛ4+, HD−) group. In conclusion, ocular vascular changes are not yet evident in participants with a genetic risk of developing AD.
Collapse
|
152
|
Kim KJ, Diaz JR, Presa JL, Muller PR, Brands MW, Khan MB, Hess DC, Althammer F, Stern JE, Filosa JA. Decreased parenchymal arteriolar tone uncouples vessel-to-neuronal communication in a mouse model of vascular cognitive impairment. GeroScience 2021; 43:1405-1422. [PMID: 33410092 PMCID: PMC8190257 DOI: 10.1007/s11357-020-00305-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/22/2020] [Indexed: 01/18/2023] Open
Abstract
Chronic hypoperfusion is a key contributor to cognitive decline and neurodegenerative conditions, but the cellular mechanisms remain ill-defined. Using a multidisciplinary approach, we sought to elucidate chronic hypoperfusion-evoked functional changes at the neurovascular unit. We used bilateral common carotid artery stenosis (BCAS), a well-established model of vascular cognitive impairment, combined with an ex vivo preparation that allows pressurization of parenchymal arterioles in a brain slice. Our results demonstrate that mild (~ 30%), chronic hypoperfusion significantly altered the functional integrity of the cortical neurovascular unit. Although pial cerebral perfusion recovered over time, parenchymal arterioles progressively lost tone, exhibiting significant reductions by day 28 post-surgery. We provide supportive evidence for reduced adenosine 1 receptor-mediated vasoconstriction as a potential mechanism in the adaptive response underlying the reduced baseline tone in parenchymal arterioles. In addition, we show that in response to the neuromodulator adenosine, the action potential frequency of cortical pyramidal neurons was significantly reduced in all groups. However, a significant decrease in adenosine-induced hyperpolarization was observed in BCAS 14 days. At the microvascular level, constriction-induced inhibition of pyramidal neurons was significantly compromised in BCAS mice. Collectively, these results suggest that BCAS uncouples vessel-to-neuron communication-vasculo-neuronal coupling-a potential early event in cognitive decline.
Collapse
Affiliation(s)
- Ki Jung Kim
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - Juan Ramiro Diaz
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - Jessica L Presa
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - P Robinson Muller
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - Michael W Brands
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - Mohammad B Khan
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - David C Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | | | - Javier E Stern
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Jessica A Filosa
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
153
|
Ebersole J, Rose G, Eid T, Behar K, Patrylo P. Altered hippocampal astroglial metabolism is associated with aging and preserved spatial learning and memory. Neurobiol Aging 2021; 102:188-199. [PMID: 33774381 DOI: 10.1016/j.neurobiolaging.2021.02.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 11/23/2022]
Abstract
An age-related decrease in hippocampal metabolism correlates with cognitive decline. Hippocampus-dependent learning and memory requires glutamatergic neurotransmission supported by glutamate-glutamine (GLU-GLN) cycling between neurons and astrocytes. We examined whether GLU-GLN cycling in hippocampal subregions (dentate gyrus and CA1) in Fischer 344 rats was altered with age and cognitive status. Hippocampal slices from young adult, aged cognitively-unimpaired (AU) and aged cognitively-impaired (AI) rats were incubated in artificial cerebrospinal fluid (aCSF) containing 1-13C-glucose to assess neural metabolism. Incorporation of 13C-glucose into glutamate and glutamine, measured by mass spectroscopy/liquid chromatography tandem mass spectroscopy, did not significantly differ between groups. However, when 13C-acetate, a preferential astrocytic metabolite, was used, a significant increase in 13C-labeled glutamate was observed in slices from AU rats. Taken together, the data suggest that resting state neural metabolism and GLU-GLN cycling may be preserved during aging when sufficient extracellular glucose is available, but that enhanced astroglial metabolism can occur under resting state conditions. This may be an aging-related compensatory change to maintain hippocampus-dependent cognitive function.
Collapse
Affiliation(s)
- Jeremy Ebersole
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL, USA
| | - Gregory Rose
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL, USA; Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL, USA; Center for Integrated Research in the Cognitive and Neural Sciences, Southern Illinois University School of Medicine, Carbondale, IL, USA
| | - Tore Eid
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Kevin Behar
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; MRRC Neurometabolism Research Laboratory, Yale University School of Medicine, New Haven, CT, USA
| | - Peter Patrylo
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL, USA; Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL, USA; Center for Integrated Research in the Cognitive and Neural Sciences, Southern Illinois University School of Medicine, Carbondale, IL, USA.
| |
Collapse
|
154
|
Jiang H, Wang J, Levin BE, Baumel BS, Camargo CJ, Signorile JF, Rundek T. Retinal Microvascular Alterations as the Biomarkers for Alzheimer Disease: Are We There Yet? J Neuroophthalmol 2021; 41:251-260. [PMID: 33136677 PMCID: PMC8079547 DOI: 10.1097/wno.0000000000001140] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Alzheimer disease (AD) is a heterogeneous and multifactorial disorder with an insidious onset and slowly progressive disease course. To date, there are no effective treatments, but biomarkers for early diagnosis and monitoring of disease progression offer a promising first step in developing and testing potential interventions. Cerebral vascular imaging biomarkers to assess the contributions of vascular dysfunction to AD are strongly recommended to be integrated into the current amyloid-β (Aβ) [A], tau [T], and neurodegeneration [(N)]-the "AT(N)" biomarker system for clinical research. However, the methodology is expensive and often requires invasive procedures to document cerebral vascular dysfunction. The retina has been used as a surrogate to study cerebral vascular changes. There is growing interest in the identification of retinal microvascular changes as a safe, easily accessible, low cost, and time-efficient approach to enhancing our understanding of the vascular pathogenesis associated with AD. EVIDENCE ACQUISITION A systemic review of the literature was performed regarding retinal vascular changes in AD and its prodromal stages, focusing on functional and structural changes of large retinal vessels (vessels visible on fundus photographs) and microvasculature (precapillary arterioles, capillary, and postcapillary venules) that are invisible on fundus photographs. RESULTS Static and dynamic retinal microvascular alterations such as retinal arterial wall motion, blood flow rate, and microvascular network density were reported in AD, mild cognitive impairment, and even in the preclinical stages of the disease. The data are somewhat controversial and inconsistent among the articles reviewed and were obtained based on cross-sectional studies that used different patient cohorts, equipment, techniques, and analysis methods. CONCLUSIONS Retinal microvascular alterations exist across the AD spectrum. Further large scale, within-subject longitudinal studies using standardized imaging and analytical methods may advance our knowledge concerning vascular contributions to the pathogenesis of AD.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jianhua Wang
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Bonnie E. Levin
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Bernard S. Baumel
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Christian J. Camargo
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Tania Rundek
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
155
|
Chang SK, Kim J, Lee D, Yoo CH, Jin S, Rhee HY, Ryu CW, Lee JK, Cho H, Jahng GH. Mapping of microvascular architecture in the brain of an Alzheimer's disease mouse model using MRI. NMR IN BIOMEDICINE 2021; 34:e4481. [PMID: 33590547 DOI: 10.1002/nbm.4481] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 11/15/2020] [Accepted: 01/08/2021] [Indexed: 06/12/2023]
Abstract
Increasing evidence suggests that alterations in cerebral microvasculature play a critical role in the pathogenesis of Alzheimer's disease (AD). The objective of this study was to characterize and evaluate the cerebral microvascular architecture of AD transgenic (Tg) mice and compare it with that of non-Tg mice using brain microvascular indices obtained by MRI. Seven non-Tg mice and 10 5xFAD Tg mice were scanned using a 7-T animal MRI system to measure the transverse relaxation rates of R2 and R2* before and after the injection of the monocrystalline iron oxide nanoparticle contrast agent. After calculating ΔR2* and ΔR2, the vessel size index (VSI), mean vessel diameter (mVD), mean vessel density, mean vessel-weighted image (MvWI) and blood volume fraction (BVf) were mapped. Voxel-based analyses and region of interest (ROI)-based analyses were performed to compare the indices of the non-Tg and Tg groups. Voxel comparisons showed that BVf, mVD, VSI and MvWI were greater in the Tg group than in the non-Tg group. Additionally, the ROI-based analysis showed that ΔR2*, BVf, mVD, MvWI and VSI increased in several brain regions of the Tg group compared with those in the non-Tg group. VSI and mVD increased in Tg mice; these findings indicated microvascular disruption in the brain that could be related to damage to the neurovascular unit in AD caused by cerebral amyloid angiopathy.
Collapse
Affiliation(s)
- Suk-Ki Chang
- Hallym University Medical Center, Hwasung, Republic of Korea
| | - JeongYeong Kim
- Department of Physics, Undergraduate School, Kyung Hee University, Seoul, Republic of Korea
| | - DongKyu Lee
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Chang Hyun Yoo
- Department of Physics, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Seokha Jin
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Hak Young Rhee
- Department of Neurology, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chang-Woo Ryu
- Department of Radiology, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jong Kil Lee
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - HyungJoon Cho
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Geon-Ho Jahng
- Department of Radiology, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
156
|
Leung E, Hazrati LN. Breast cancer type 1 and neurodegeneration: consequences of deficient DNA repair. Brain Commun 2021; 3:fcab117. [PMID: 34222870 PMCID: PMC8242133 DOI: 10.1093/braincomms/fcab117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 12/20/2022] Open
Abstract
Numerous cellular processes, including toxic protein aggregation and oxidative stress, have been studied extensively as potential mechanisms underlying neurodegeneration. However, limited therapeutic efficacy targeting these processes has prompted other mechanisms to be explored. Previous research has emphasized a link between cellular senescence and neurodegeneration, where senescence induced by excess DNA damage and deficient DNA repair results in structural and functional changes that ultimately contribute to brain dysfunction and increased vulnerability for neurodegeneration. Specific DNA repair proteins, such as breast cancer type 1, have been associated with both stress-induced senescence and neurodegenerative diseases, however, specific mechanisms remain unclear. Therefore, this review explores DNA damage-induced senescence in the brain as a driver of neurodegeneration, with particular focus on breast cancer type 1, and its potential contribution to sex-specific differences associated with neurodegenerative disease.
Collapse
Affiliation(s)
- Emily Leung
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 Kings College Cir, Toronto, ON M5S 1A8, Canada
- The Hospital for Sick Children, 555 University Ave, Toronto, ON M5G 1X8, Canada
| | - Lili-Naz Hazrati
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 Kings College Cir, Toronto, ON M5S 1A8, Canada
- The Hospital for Sick Children, 555 University Ave, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
157
|
Abstract
An aging-related reduction in the brain's functional reserve may explain why delirium is more frequent in the elderly than in younger people insofar as the reserve becomes inadequate to cover the metabolic requirements that are critically increased by stressors. The aim of this paper is to review the normal aging-related changes that theoretically compromise complex mental activities, neuronal and synaptic densities, and the neurocomputational flexibility of the functional reserve. A pivotal factor is diminished connectivity, which is substantially due to the loss of synapses and should specifically affect association systems and cholinergic fibres in delirious patients. However, micro-angiopathy with impaired blood flow autoregulation, increased blood/brain barrier permeability, changes in cerebrospinal fluid dynamics, weakened mitochondrial performance, and a pro-inflammatory involution of the immune system may also jointly affect neurons and their synaptic assets, and even cause the progression of delirium to dementia regardless of the presence of co-existing plaques, tangles, or other pathological markers. On the other hand, the developmental growth in functional reserve during childhood and adolescence makes the brain increasingly resistant to delirium, and residual reserve can allow the elderly to recover. These data support the view that functional reserve is the variable that confronts stressors and governs the risk and intensity of and recovery from delirium. Although people of any age are at risk of delirium, the elderly are at greater risk because aging and age-dependent structural changes inevitably affect the brain's functional reserve.
Collapse
|
158
|
Brazile BL, Yang B, Waxman S, Lam P, Voorhees AP, Hua Y, Loewen RT, Loewen NA, Rizzo JF, Jakobs T, Sigal IA. Lamina Cribrosa Capillaries Straighten as Intraocular Pressure Increases. Invest Ophthalmol Vis Sci 2021; 61:2. [PMID: 33001158 PMCID: PMC7545063 DOI: 10.1167/iovs.61.12.2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Purpose The purpose of this study was to visualize the lamina cribrosa (LC) capillaries and collagenous beams, measure capillary tortuosity (path length over straight end-to-end length), and determine if capillary tortuosity changes when intraocular pressure (IOP) increases. Methods Within 8 hours of sacrifice, 3 pig heads were cannulated via the external ophthalmic artery, perfused with PBS to remove blood, and then perfused with a fluorescent dye to label the capillaries. The posterior pole of each eye was mounted in a custom-made inflation chamber for control of IOP with simultaneous imaging. Capillaries and collagen beams were visualized with structured light illumination enhanced imaging at IOPs from 5 to 50 mm Hg at each 5 mm Hg increment. Capillary tortuosity was measured from the images and paired two-sample t-tests were used to assess for significant changes in relation to changes in IOP. Results Capillaries were highly tortuous at 15 mm Hg (up to 1.45). In all but one eye, tortuosity decreased significantly as IOP increased from 15 to 25 mm Hg (P < 0.01), and tortuosity decreased significantly in every eye as IOP increased from 15 to 40 mm Hg (P < 0.01). In only 16% of capillaries, tortuosity increased with elevated IOP. Capillaries had a surprisingly different topology from the collagen beams. Conclusions Although high capillary tortuosity is sometimes regarded as potentially problematic because it can reduce blood flow, LC capillary tortuosity may provide slack that mitigates against reduced flow and structural damage caused by excessive stretch under elevated IOP. We speculate that low capillary tortuosity could be a risk factor for damage under high IOP.
Collapse
Affiliation(s)
- Bryn L Brazile
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh Pennsylvania, United States
| | - Bin Yang
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh Pennsylvania, United States.,Department of Engineering, Duquesne University, Pittsburgh Pennsylvania, United States
| | - Susannah Waxman
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh Pennsylvania, United States
| | - Po Lam
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh Pennsylvania, United States
| | - Andrew P Voorhees
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh Pennsylvania, United States
| | - Yi Hua
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh Pennsylvania, United States
| | - Ralitsa T Loewen
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh Pennsylvania, United States
| | - Nils A Loewen
- Department of Ophthalmology, University of Würzburg, Würzburg, Germany
| | - Joseph F Rizzo
- Neuro-Ophthalmology Service, Department of Ophthalmology, Massachusetts Eye and Ear and Harvard Medical School, Boston, Massachusetts, United States
| | - Tatjana Jakobs
- Department of Ophthalmology, Harvard Medical School, Boston Massachusetts, United States
| | - Ian A Sigal
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh Pennsylvania, United States.,Department of Bioengineering, University of Pittsburgh, Pittsburgh Pennsylvania, United States
| |
Collapse
|
159
|
Taccola C, Barneoud P, Cartot-Cotton S, Valente D, Schussler N, Saubaméa B, Chasseigneaux S, Cochois V, Mignon V, Curis E, Lochus M, Nicolic S, Dodacki A, Cisternino S, Declèves X, Bourasset F. Modifications of physical and functional integrity of the blood-brain barrier in an inducible mouse model of neurodegeneration. Neuropharmacology 2021; 191:108588. [PMID: 33940010 DOI: 10.1016/j.neuropharm.2021.108588] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 04/10/2021] [Accepted: 04/20/2021] [Indexed: 12/28/2022]
Abstract
The inducible p25 overexpression mouse model recapitulate many hallmark features of Alzheimer's disase including progressive neuronal loss, elevated Aβ, tau pathology, cognitive dysfunction, and impaired synaptic plasticity. We chose p25 mice to evaluate the physical and functional integrity of the blood-brain barrier (BBB) in a context of Tau pathology (pTau) and severe neurodegeneration, at an early (3 weeks ON) and a late (6 weeks ON) stage of the pathology. Using in situ brain perfusion and confocal imaging, we found that the brain vascular surface area and the physical integrity of the BBB were unaltered in p25 mice. However, there was a significant 14% decrease in cerebrovascular volume in 6 weeks ON mice, possibly explained by a significant 27% increase of collagen IV in the basement membrane of brain capillaries. The function of the BBB transporters GLUT1 and LAT1 was evaluated by measuring brain uptake of d-glucose and phenylalanine, respectively. In 6 weeks ON p25 mice, d-glucose brain uptake was significantly reduced by about 17% compared with WT, without any change in the levels of GLUT1 protein or mRNA in brain capillaries. The brain uptake of phenylalanine was not significantly reduced in p25 mice compared with WT. Lack of BBB integrity, impaired BBB d-glucose transport have been observed in several mouse models of AD. In contrast, reduced cerebrovascular volume and an increased basement membrane thickness may be more specifically associated with pTau in mouse models of neurodegeneration.
Collapse
Affiliation(s)
- Camille Taccola
- Pharmacokinetics, Dynamics and Metabolism, Translational Medicine & Early Development, Sanofi, 3 Digue d'Alfortville, 94140, Alfortville, France; INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Pascal Barneoud
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, 1 Avenue Pierre Brossolette, 91380, Chilly-Mazarin, France
| | - Sylvaine Cartot-Cotton
- Pharmacokinetics, Dynamics and Metabolism, Translational Medicine & Early Development, Sanofi, 3 Digue d'Alfortville, 94140, Alfortville, France
| | - Delphine Valente
- Drug Metabolism & Pharmacokinetics, Research platform, Sanofi, 3 Digue d'Alfortville, 94140, Alfortville, France
| | - Nathalie Schussler
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, 1 Avenue Pierre Brossolette, 91380, Chilly-Mazarin, France
| | - Bruno Saubaméa
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Stéphanie Chasseigneaux
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Véronique Cochois
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Virginie Mignon
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Emmanuel Curis
- Laboratoire de biomathématiques, plateau iB(2), EA 7537 « BioSTM », UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France; Service de bioinformatique et statistique médicale, hôpital Saint-Louis, APHP, 1, avenue Claude Vellefaux, 75010, Paris, France
| | - Murielle Lochus
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Sophie Nicolic
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Agnès Dodacki
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Salvatore Cisternino
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Xavier Declèves
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Fanchon Bourasset
- Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, Université Bourgogne Franche-Comté, 19 rue Ambroise Paré, 25000, Besançon, France.
| |
Collapse
|
160
|
Insoluble Vascular Amyloid Deposits Trigger Disruption of the Neurovascular Unit in Alzheimer's Disease Brains. Int J Mol Sci 2021; 22:ijms22073654. [PMID: 33915754 PMCID: PMC8036769 DOI: 10.3390/ijms22073654] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease, characterized histopathologically by intra-neuronal tau-related lesions and by the accumulation of amyloid β-peptide (Aβ) in the brain parenchyma and around cerebral blood vessels. According to the vascular hypothesis of AD, an alteration in the neurovascular unit (NVU) could lead to Aβ vascular accumulation and promote neuronal dysfunction, accelerating neurodegeneration and dementia. To date, the effects of insoluble vascular Aβ deposits on the NVU and the blood-brain barrier (BBB) are unknown. In this study, we analyze different Aβ species and their association with the cells that make up the NVU. We evaluated post-mortem AD brain tissue. Multiple immunofluorescence assays were performed against different species of Aβ and the main elements that constitute the NVU. Our results showed that there are insoluble vascular deposits of both full-length and truncated Aβ species. Besides, insoluble aggregates are associated with a decrease in the phenotype of the cellular components that constitute the NVU and with BBB disruption. This approach could help identify new therapeutic targets against key molecules and receptors in the NVU that can prevent the accumulation of vascular fibrillar Aβ in AD.
Collapse
|
161
|
Wakisaka Y, Matsuo R, Nakamura K, Ago T, Kamouchi M, Kitazono T. Pre-Stroke Cholinesterase Inhibitor Treatment Is Beneficially Associated with Functional Outcome in Patients with Acute Ischemic Stroke and Pre-Stroke Dementia: The Fukuoka Stroke Registry. Cerebrovasc Dis 2021; 50:390-396. [PMID: 33744892 DOI: 10.1159/000514368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/02/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Pre-stroke dementia is significantly associated with poor stroke outcome. Cholinesterase inhibitors (ChEIs) might reduce the risk of stroke in patients with dementia. However, the association between pre-stroke ChEI treatment and stroke outcome remains unresolved. Therefore, we aimed to determine this association in patients with acute ischemic stroke and pre-stroke dementia. METHODS We enrolled 805 patients with pre-stroke dementia among 13,167 with ischemic stroke within 7 days of onset who were registered in the Fukuoka Stroke Registry between June 2007 and May 2019 and were independent in basic activities of daily living (ADLs) before admission. Primary and secondary study outcomes were poor functional outcome (modified Rankin Scale [mRS] score: 3-6) at 3 months after stroke onset and neurological deterioration (≥2-point increase in the NIH Stroke Scale [NIHSS] during hospitalization), respectively. Logistic regression analysis was used to evaluate associations between pre-stroke ChEI treatment and study outcomes. To improve covariate imbalance, we further conducted a propensity score (PS)-matched cohort study. RESULTS Among the participants, 212 (26.3%) had pre-stroke ChEI treatment. Treatment was negatively associated with poor functional outcome (odds ratio: 0.68 [95% confidence interval: 0.46-0.99]) and neurological deterioration (0.52 [0.31-0.88]) after adjusting for potential confounding factors. In the PS-matched cohort study, the same trends were observed between pre-stroke ChEI treatment and poor functional outcome (0.61 [0.40-0.92]) and between the treatment and neurological deterioration (0.47 [0.25-0.86]). CONCLUSIONS Our findings suggest that pre-stroke ChEI treatment is associated with reduced risks for poor functional outcome and neurological deterioration after acute ischemic stroke in patients with pre-stroke dementia who are independent in basic ADLs before the onset of stroke.
Collapse
Affiliation(s)
- Yoshinobu Wakisaka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan,
| | - Ryu Matsuo
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kuniyuki Nakamura
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuro Ago
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masahiro Kamouchi
- Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
162
|
PHACTR1 genetic variability is not critical in small vessel ischemic disease patients and PcomA recruitment in C57BL/6J mice. Sci Rep 2021; 11:6072. [PMID: 33727568 PMCID: PMC7966789 DOI: 10.1038/s41598-021-84919-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 02/04/2021] [Indexed: 11/29/2022] Open
Abstract
Recently, several genome-wide association studies identified PHACTR1 as key locus for five diverse vascular disorders: coronary artery disease, migraine, fibromuscular dysplasia, cervical artery dissection and hypertension. Although these represent significant risk factors or comorbidities for ischemic stroke, PHACTR1 role in brain small vessel ischemic disease and ischemic stroke most important survival mechanism, such as the recruitment of brain collateral arteries like posterior communicating arteries (PcomAs), remains unknown. Therefore, we applied exome and genome sequencing in a multi-ethnic cohort of 180 early-onset independent familial and apparently sporadic brain small vessel ischemic disease and CADASIL-like Caucasian patients from US, Portugal, Finland, Serbia and Turkey and in 2 C57BL/6J stroke mouse models (bilateral common carotid artery stenosis [BCCAS] and middle cerebral artery occlusion [MCAO]), characterized by different degrees of PcomAs patency. We report 3 very rare coding variants in the small vessel ischemic disease-CADASIL-like cohort (p.Glu198Gln, p.Arg204Gly, p.Val251Leu) and a stop-gain mutation (p.Gln273*) in one MCAO mouse. These coding variants do not cluster in PHACTR1 known pathogenic domains and are not likely to play a critical role in small vessel ischemic disease or brain collateral circulation. We also exclude the possibility that copy number variants (CNVs) or a variant enrichment in Phactr1 may be associated with PcomA recruitment in BCCAS mice or linked to diverse vascular traits (cerebral blood flow pre-surgery, PcomA size, leptomeningeal microcollateral length and junction density during brain hypoperfusion) in C57BL/6J mice, respectively. Genetic variability in PHACTR1 is not likely to be a common susceptibility factor influencing small vessel ischemic disease in patients and PcomA recruitment in C57BL/6J mice. Nonetheless, rare variants in PHACTR1 RPEL domains may influence the stroke outcome and are worth investigating in a larger cohort of small vessel ischemic disease patients, different ischemic stroke subtypes and with functional studies.
Collapse
|
163
|
Sharma S, Nozohouri S, Vaidya B, Abbruscato T. Repurposing metformin to treat age-related neurodegenerative disorders and ischemic stroke. Life Sci 2021; 274:119343. [PMID: 33716063 DOI: 10.1016/j.lfs.2021.119343] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022]
Abstract
Aging is a risk factor for major central nervous system (CNS) disorders. More specifically, aging can be inked to neurodegenerative diseases (NDs) because of its deteriorating impact on neurovascular unit (NVU). Metformin, a first line FDA-approved anti-diabetic drug, has gained increasing interest among researchers for its role in improving aging-related neurodegenerative disorders. Additionally, numerous studies have illustrated metformin's role in ischemic stroke, a cerebrovascular disorder in which the NVU becomes dysfunctional which can lead to permanent life-threatening disabilities. Considering metformin's beneficial preclinical actions on various disorders, and the drug's role in alleviating severity of these conditions through involvement in commonly characterized cellular pathways, we discuss the potential of metformin as a suitable drug candidate for repurposing in CNS disorders.
Collapse
Affiliation(s)
- Sejal Sharma
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA
| | - Saeideh Nozohouri
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA
| | - Bhuvaneshwar Vaidya
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA
| | - Thomas Abbruscato
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA.
| |
Collapse
|
164
|
Rifai OM, McGrory S, Robbins CB, Grewal DS, Liu A, Fekrat S, MacGillivray TJ. The application of optical coherence tomography angiography in Alzheimer's disease: A systematic review. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12149. [PMID: 33718582 PMCID: PMC7927164 DOI: 10.1002/dad2.12149] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/12/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Discovering non-invasive and easily acquired biomarkers that are conducive to the accurate diagnosis of dementia is an urgent area of ongoing clinical research. One promising approach is retinal imaging, as there is homology between retinal and cerebral vasculature. Recently, optical coherence tomography angiography (OCT-A) has emerged as a promising new technology for imaging the microvasculature of the retina. METHODS A systematic review and meta-analysis was conducted to examine the application of OCT-A in dementia. RESULTS Fourteen studies assessing OCT-A in preclinical Alzheimer's disease (AD), mild cognitive impairment, or AD were included. Exploratory meta-analyses revealed a significant increase in the foveal avascular zone area and a significant decrease in superficial parafoveal and whole vessel density in AD, although there was significant heterogeneity between studies. DISCUSSION Although certain OCT-A metrics may have the potential to serve as biomarkers for AD, the field requires further standardization to allow conclusions to be reached regarding their clinical utility.
Collapse
Affiliation(s)
- Olivia M. Rifai
- Translational Neuroscience PhD ProgrammeUniversity of EdinburghEdinburghUK
- Centre for Clinical Brain SciencesCollege of Medicine and Veterinary MedicineUniversity of EdinburghEdinburghUK
| | - Sarah McGrory
- Centre for Clinical Brain SciencesCollege of Medicine and Veterinary MedicineUniversity of EdinburghEdinburghUK
| | - Cason B. Robbins
- Department of OphthalmologyDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Dilraj S. Grewal
- Department of OphthalmologyDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Andy Liu
- Department of NeurologyDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Sharon Fekrat
- Department of OphthalmologyDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Thomas J. MacGillivray
- Centre for Clinical Brain SciencesCollege of Medicine and Veterinary MedicineUniversity of EdinburghEdinburghUK
| |
Collapse
|
165
|
Ao DH, Zhang DD, Zhai FF, Zhang JT, Han F, Li ML, Ni J, Yao M, Zhang SY, Cui LY, Jin ZY, Zhou LX, Zhu YC. Brain deep medullary veins on 3-T MRI in a population-based cohort. J Cereb Blood Flow Metab 2021; 41:561-568. [PMID: 32312169 PMCID: PMC7922755 DOI: 10.1177/0271678x20918467] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Our aim is to investigate whether vascular risk factors are associated with cerebral deep medullary veins (DMVs) and whether DMVs are associated with MRI markers of cerebral small vessel disease (CSVD) or risk of stroke. In a community-based cohort of 1056 participants (mean age 55.7 years), DMVs were identified on susceptibility-weighted imaging (SWI) and counted in periventricular regions. Neuroimaging markers including lacunes, whiter matter hyperintensity (WMH), microbleeds, enlarged perivascular space, and brain atrophy were evaluated. The number of DMVs decreased with age (p = 0.007). After adjusting for age and sex, the number of DMVs was not associated with traditional vascular risk factors. Fewer DMVs was associated with increase of WMH and lacunes, but the association vanished after adjustment for vascular risk factors. However, fewer DMVs were independently associated with brain atrophy (p < 0.001). DMVs were not associated with three-year risk of stroke. Our results suggest that DMV is significantly different from other MRI markers of CSVD regarding risk factors, association with other CSVD markers, and risk of stroke. Nonetheless, the significant association between DMV and brain atrophy suggested the potential role of venules in age-related neurodegenerative process, which deserves further investigation.
Collapse
Affiliation(s)
- Dong-Hui Ao
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ding-Ding Zhang
- Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei-Fei Zhai
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiang-Tao Zhang
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Han
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Li Li
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Ni
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming Yao
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shu-Yang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Ying Cui
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng-Yu Jin
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Xin Zhou
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi-Cheng Zhu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
166
|
Lee W, Weisholtz DS, Strangman GE, Yoo SS. Safety Review and Perspectives of Transcranial Focused Ultrasound Brain Stimulation. BRAIN & NEUROREHABILITATION 2021; 14:e4. [PMID: 36742103 PMCID: PMC9879416 DOI: 10.12786/bn.2021.14.e4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/17/2021] [Accepted: 03/04/2021] [Indexed: 11/08/2022] Open
Abstract
Ultrasound is an important theragnostic modality in modern medicine. Technical advancement of both acoustic focusing and transcranial delivery have enabled administration of ultrasound waves to localized brain areas with few millimeters of spatial specificity and penetration depth sufficient to reach the thalamus. Transcranial focused ultrasound (tFUS) given at a low acoustic intensity has been shown to increase or suppress the excitability of region-specific brain areas. The neuromodulatory effects can outlast the sonication, suggesting the possibility of inducing neural plasticity needed for neurorehabilitation. Increasing numbers of studies have shown the efficacy and excellent safety profile of the technique, yet comparisons among the safety-related parameters have not been compiled. This review aims to provide safety information and perspectives of tFUS brain stimulation. First, the acoustic parameters most relevant to thermal/mechanical tissue damage are discussed along with regulated parameters for existing ultrasound therapies/diagnostic imaging. Subsequently, the parameters used in studies of large animals, non-human primates, and humans are surveyed and summarized in terms of the acoustic intensity and the mechanical index. The pulse-mode operation and the use of low ultrasound frequency for tFUS-mediated brain stimulation warrant the establishment of new safety guidelines/recommendations for the use of the technique among healthy volunteers, with additional cautionary requirements for its clinical translation.
Collapse
Affiliation(s)
- Wonhye Lee
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel S. Weisholtz
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gary E. Strangman
- Neural Systems Group, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
- Translational Research Institute, Houston, TX, USA
| | - Seung-Schik Yoo
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
167
|
Rand D, Ravid O, Atrakchi D, Israelov H, Bresler Y, Shemesh C, Omesi L, Liraz-Zaltsman S, Gosselet F, Maskrey TS, Schnaider Beeri M, Wipf P, Cooper I. Endothelial Iron Homeostasis Regulates Blood-Brain Barrier Integrity via the HIF2α-Ve-Cadherin Pathway. Pharmaceutics 2021; 13:pharmaceutics13030311. [PMID: 33670876 PMCID: PMC7997362 DOI: 10.3390/pharmaceutics13030311] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 12/14/2022] Open
Abstract
The objective of this study was to investigate the molecular response to damage at the blood-brain barrier (BBB) and to elucidate critical pathways that might lead to effective treatment in central nervous system (CNS) pathologies in which the BBB is compromised. We have used a human, stem-cell derived in-vitro BBB injury model to gain a better understanding of the mechanisms controlling BBB integrity. Chemical injury induced by exposure to an organophosphate resulted in rapid lipid peroxidation, initiating a ferroptosis-like process. Additionally, mitochondrial ROS formation (MRF) and increase in mitochondrial membrane permeability were induced, leading to apoptotic cell death. Yet, these processes did not directly result in damage to barrier functionality, since blocking them did not reverse the increased permeability. We found that the iron chelator, Desferal© significantly decreased MRF and apoptosis subsequent to barrier insult, while also rescuing barrier integrity by inhibiting the labile iron pool increase, inducing HIF2α expression and preventing the degradation of Ve-cadherin specifically on the endothelial cell surface. Moreover, the novel nitroxide JP4-039 significantly rescued both injury-induced endothelium cell toxicity and barrier functionality. Elucidating a regulatory pathway that maintains BBB integrity illuminates a potential therapeutic approach to protect the BBB degradation that is evident in many neurological diseases.
Collapse
Affiliation(s)
- Daniel Rand
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Orly Ravid
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
| | - Dana Atrakchi
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
| | - Hila Israelov
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
| | - Yael Bresler
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Chen Shemesh
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
| | - Liora Omesi
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
| | - Sigal Liraz-Zaltsman
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
- Department of Pharmacology, The Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem 97905, Israel
- Department of Sports Therapy, Institute for Health and Medical Professions, Ono Academic College, Kiryat Ono 55000, Israel
| | - Fabien Gosselet
- Blood-Brain Barrier Laboratory (LBHE), Artois University, UR 2465, F-62300 Lens, France;
| | - Taber S. Maskrey
- Department of Chemistry and Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (T.S.M.); (P.W.)
| | - Michal Schnaider Beeri
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
- Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- School of Psychology, Interdisciplinary Center (IDC), Herzliya 4610101, Israel
| | - Peter Wipf
- Department of Chemistry and Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (T.S.M.); (P.W.)
| | - Itzik Cooper
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
- School of Psychology, Interdisciplinary Center (IDC), Herzliya 4610101, Israel
- The Nehemia Rubin Excellence in Biomedical Research—The TELEM Program, Sheba Medical Center, Tel-Hashomer 5262000, Israel
- Correspondence:
| |
Collapse
|
168
|
Han F, Zhai FF, Li ML, Zhou LX, Ni J, Yao M, Jin ZY, Cui LY, Zhang SY, Zhu YC. Arterial Stiffness Is Associated with White Matter Disruption and Cognitive Impairment: A Community-Based Cohort Study. J Alzheimers Dis 2021; 80:567-576. [PMID: 33579854 DOI: 10.3233/jad-201424] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Mechanisms through which arterial stiffness impacts cognitive function are crucial for devising better strategies to prevent cognitive decline. OBJECTIVE To examine the associations of arterial stiffness with white matter integrity and cognition in community dwellings, and to investigate whether white matter injury was the intermediate of the associations between arterial stiffness and cognition. METHODS This study was a cross-sectional analysis on 952 subjects (aged 55.5±9.1 years) who underwent diffusion tensor imaging and measurement of brachial-ankle pulse wave velocity (baPWV). Both linear regression and tract-based spatial statistics were used to investigate the association between baPWV and white matter integrity. The association between baPWV and global cognitive function, measured as the mini-mental state examination (MMSE) was evaluated. Mediation analysis was performed to assess the influence of white matter integrity on the association of baPWV with MMSE. RESULTS Increased baPWV was significantly associated with lower mean global fractional anisotropy (β= -0.118, p < 0.001), higher mean diffusivity (β= 0.161, p < 0.001), axial diffusivity (β= 0.160, p < 0.001), and radial diffusivity (β= 0.147, p < 0.001) after adjustment of age, sex, and hypertension, which were measures having a direct effect on arterial stiffness and white matter integrity. After adjustment of age, sex, education, apolipoprotein E ɛ4, cardiovascular risk factors, and brain atrophy, we found an association of increased baPWV with worse performance on MMSE (β= -0.093, p = 0.011). White matter disruption partially mediated the effect of baPWV on MMSE. CONCLUSION Arterial stiffness is associated with white matter disruption and cognitive decline. Reduced white matter integrity partially explained the effect of arterial stiffness on cognition.
Collapse
Affiliation(s)
- Fei Han
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Fei-Fei Zhai
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Ming-Li Li
- Department of Radiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Li-Xin Zhou
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Jun Ni
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Ming Yao
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Zheng-Yu Jin
- Department of Radiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Li-Ying Cui
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Shu-Yang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Yi-Cheng Zhu
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| |
Collapse
|
169
|
Abstract
In chronic kidney disease (CKD), disturbance of several metabolic regulatory mechanisms cause premature ageing, accelerated cardiovascular disease (CVD), and mortality. Single-target interventions have repeatedly failed to improve the prognosis for CKD patients. Epigenetic interventions have the potential to modulate several pathogenetic processes simultaneously. Alkaline phosphatase (ALP) is a robust predictor of CVD and all-cause mortality and implicated in pathogenic processes associated with CVD in CKD.
Collapse
|
170
|
Steinman J, Sun HS, Feng ZP. Microvascular Alterations in Alzheimer's Disease. Front Cell Neurosci 2021; 14:618986. [PMID: 33536876 PMCID: PMC7849053 DOI: 10.3389/fncel.2020.618986] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/17/2020] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with continual decline in cognition and ability to perform routine functions such as remembering familiar places or understanding speech. For decades, amyloid beta (Aβ) was viewed as the driver of AD, triggering neurodegenerative processes such as inflammation and formation of neurofibrillary tangles (NFTs). This approach has not yielded therapeutics that cure the disease or significant improvements in long-term cognition through removal of plaques and Aβ oligomers. Some researchers propose alternate mechanisms that drive AD or act in conjunction with amyloid to promote neurodegeneration. This review summarizes the status of AD research and examines research directions including and beyond Aβ, such as tau, inflammation, and protein clearance mechanisms. The effect of aging on microvasculature is highlighted, including its contribution to reduced blood flow that impairs cognition. Microvascular alterations observed in AD are outlined, emphasizing imaging studies of capillary malfunction. The review concludes with a discussion of two therapies to protect tissue without directly targeting Aβ for removal: (1) administration of growth factors to promote vascular recovery in AD; (2) inhibiting activity of a calcium-permeable ion channels to reduce microglial activation and restore cerebral vascular function.
Collapse
Affiliation(s)
- Joe Steinman
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Hong-Shuo Sun
- Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Zhong-Ping Feng
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
171
|
Dobrynina LA, Gnedovskaya EV, Zabitova MR, Kremneva EI, Shabalina AA, Makarova AG, Tzipushtanova MM, Filatov AS, Kalashnikova LA, Krotenkova MV. [Clustering of diagnostic MRI signs of cerebral microangiopathy and its relationship with markers of inflammation and angiogenesis]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 120:22-31. [PMID: 33449529 DOI: 10.17116/jnevro202012012222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To perform cluster analysis of MRI signs of cerebral microangiopathy (small vessel disease, SVD) and to clarify the relationship between the isolated groups and circulating markers of inflammation and angiogenesis. MATERIAL AND METHODS The identification of groups of MRI signs (MRI types) using cluster hierarchical agglomerative analysis and iterative algorithm of k-means and assessment of their relationship with serum concentrations of tumor necrosis factor-α (TNF-α), transforming growth factor-β1 (TGF-β1), vascular endothelial growth factor-A (VEGF-A), hypoxia-inducible factor 1-α (HIF1-α) determined by ELISA were performed in 96 patients with SVD (STRIVE, 2013) (65 women, average age 60.91±6.57 years). RESULTS Cluster analysis of MRI signs identified two MRI types of SVD with Fazekas grade 3 of white matter hyperintensity (WMH). MRI type 1 (n=18; 6 women, mean age 59.1±6.8 years) and MRI type 2 (n=22, 15 f., mean age 63.5±6.2 years) did not differ by age, sex, severity of hypertension, presence of other risk factors. MRI type 1 had a statistically significantly more pronounced WMH in the periventricular regions, multiple lacunes and microbleeds, atrophy, severe cognitive impairment and gait disorders compared with MRI type 2. Its formation was associated with a decrease in VEGF-A level. MRI type 2 had the significantly more pronounced juxtacortical WMH, white matter lacunes, in the absence of microbleeds and atrophy, and less severe clinical manifestations compared with MRI type 1. Its formation was associated with an increase in TNF-α level. CONCLUSION Clustering of diagnostic MRI signs into MRI types of SVD with significant differences in the severity of clinical manifestations suggests the pathogenetic heterogeneity of age-related SVD. The relationship of MRI types with circulating markers of different mechanisms of vascular wall and brain damage indicates the dominant role of depletion of angiogenesis in the formation of MRI type 1 and increased inflammation in the formation of MRI type 2. Further studies are needed to clarify the criteria and diagnostic value of differentiation of MRI types of SVD, and also their mechanisms with the definition of pathogenetically justified prevention and treatment of various forms of SVD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - A S Filatov
- Research Center of Neurology, Moscow, Russia
| | | | | |
Collapse
|
172
|
Cao Y, Yan J, Zhan Z, Liang Y, Han Z. Macula Structure and Microvascular Changes in Recent Small Subcortical Infarct Patients. Front Neurol 2021; 11:615252. [PMID: 33488504 PMCID: PMC7817655 DOI: 10.3389/fneur.2020.615252] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/23/2020] [Indexed: 12/29/2022] Open
Abstract
Purpose: This study aimed to assess the macula structure and capillaries in the macula and optic nerve head in recent small subcortical infarct (RSSI) patients. Methods: This observational cross-sectional study included 40 RSSI patients and 46 healthy controls. Optical coherence tomography angiography was used to image the capillaries in the macula and optic nerve head. An inbuilt algorithm was used to measure the densities in the microvasculature of the macula [superficial retinal capillary plexus (SRCP) and deep retinal capillary plexus (DRCP)] and optic nerve head [radial peripapillary capillary (RPC)] and thickness around the optic nerve head, peripapillary retinal nerve fiber layer (pRNFL). Results: Densities in RPC (P < 0.001), SRCP (P = 0.001), and DRCP (P = 0.003) were reduced in RSSI patients when compared with healthy controls. The pRNFL thickness was thinner (P < 0.001) in RSSI patients than healthy controls. In the RSSI group, the SRCP density significantly correlated with the DRCP density (rho = 0.381, P = 0.042). The pRNFL thickness displayed a significant relationship with the RPC density (rho = 0.482, P = 0.003) in the RSSI group. Conclusions: RSSI patients showed interrupted capillary plexuses leading to its significant impairment and neurodegeneration. Our report provides insight into the macula capillary microcirculation changes in RSSI.
Collapse
Affiliation(s)
- Yungang Cao
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jueyue Yan
- School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Zhenxiang Zhan
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuanbo Liang
- Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhao Han
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
173
|
Abstract
Age-related sporadic cerebral small vessel disease (CSVD) has gained increasing attention over the past decades because of its increasing prevalence associated with an aging population. The widespread application of and advances in brain magnetic resonance imaging in recent decades have significantly increased researchers’ understanding in the in vivo evolution of CSVD, its impact upon the brain, its risk factors, and the mechanisms that explain the various clinical manifestation associated with sporadic CSVD. In this review, we aimed to provide an update on the pathophysiology, risk factors, biomarkers, and the determinants and spectrum of the clinical manifestation of sporadic CSVD.
Collapse
|
174
|
Coupling of cerebral blood flow and functional connectivity is decreased in healthy aging. Brain Imaging Behav 2021; 14:436-450. [PMID: 31250268 DOI: 10.1007/s11682-019-00157-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Aging leads to cerebral perfusion and functional connectivity changes that have been assessed using various neuroimaging techniques. In addition, a link between these two parameters has been demonstrated in healthy young adults. In this work, we employed arterial spin labeling (ASL) fMRI to measure global and voxel-wise differences in cerebral blood flow (CBF) and intrinsic connectivity contrast (ICC) in the resting state in a group of cognitively normal elderly subjects and a group of cognitively normal young subjects, in order to assess the effects of aging on CBF-ICC coupling, which had not been previously evaluated. Our results showed age-related global and regional CBF decreases in prefrontal mesial areas, lateral frontal regions, insular cortex, lateral parietal areas, precuneus and occipital regions. Subcortically, perfusion was reduced in the medial thalamus and caudate nucleus. ICC was also found reduced with age in prefrontal cortical areas and insular cortex, affecting key nodes of the default mode and salience networks. Areas of ICC and CBF decrease partially overlapped, however, the CBF reduction was more extensive and encompassed more areas. This dissociation was accompanied by a decrease in CBF-ICC coupling. These results suggest that aging leads to a disruption in the relationship between CBF and intrinsic functional connectivity that could be due to neurovascular dysregulation.
Collapse
|
175
|
Al-Bachari S, Naish JH, Parker GJM, Emsley HCA, Parkes LM. Blood-Brain Barrier Leakage Is Increased in Parkinson's Disease. Front Physiol 2020; 11:593026. [PMID: 33414722 PMCID: PMC7784911 DOI: 10.3389/fphys.2020.593026] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 11/24/2020] [Indexed: 12/21/2022] Open
Abstract
Background Blood–brain barrier (BBB) disruption has been noted in animal models of Parkinson’s disease (PD) and forms the basis of the vascular hypothesis of neurodegeneration, yet clinical studies are lacking. Objective To determine alterations in BBB integrity in PD, with comparison to cerebrovascular disease. Methods Dynamic contrast enhanced magnetic resonance images were collected from 49 PD patients, 15 control subjects with cerebrovascular disease [control positive (CP)] and 31 healthy control subjects [control negative (CN)], with all groups matched for age. Quantitative maps of the contrast agent transfer coefficient across the BBB (Ktrans) and plasma volume (vp) were produced using Patlak analysis. Differences in Ktrans and vp were assessed with voxel-based analysis as well as in regions associated with PD pathophysiology. In addition, the volume of white matter lesions (WMLs) was obtained from T2-weighted fluid attenuation inversion recovery (FLAIR) images. Results Higher Ktrans, reflecting higher BBB leakage, was found in the PD group than in the CN group using voxel-based analysis; differences were most prominent in the posterior white matter regions. Region of interest analysis confirmed Ktrans to be significantly higher in PD than in CN, predominantly driven by differences in the substantia nigra, normal-appearing white matter, WML and the posterior cortex. WML volume was significantly higher in PD compared to CN. Ktrans values and WML volume were similar in PD and CP, suggesting a similar burden of cerebrovascular disease despite lower cardiovascular risk factors. Conclusion These results show BBB disruption in PD.
Collapse
Affiliation(s)
- Sarah Al-Bachari
- Lancaster Medical School, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom.,Department of Neurology, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, United Kingdom.,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Josephine H Naish
- Division of Cardiovascular sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,Bioxydyn Limited, Manchester, United Kingdom
| | - Geoff J M Parker
- Bioxydyn Limited, Manchester, United Kingdom.,Centre for Medical Image Computing, Department of Computer Science and Department of Neuroinflammation, University College London, London, United Kingdom
| | - Hedley C A Emsley
- Lancaster Medical School, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom.,Department of Neurology, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, United Kingdom.,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Laura M Parkes
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
176
|
Rajamani K. The Cerebro-Renal System- Anatomical and Physiological Considerations. J Stroke Cerebrovasc Dis 2020; 30:105541. [PMID: 33339697 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 12/25/2022] Open
Abstract
The brain and kidney both uniquely are highly susceptible to vascular injury from shared vascular risk factors. However these are not sufficient to explain the complete extent of cerebrovascular disease especially small vessel disease in its myriad presentations that patients with chronic kidney disease manifest. They both require a large amount of blood supply to function optimally. Shared anatomical and physiological factors such as the presence of strain vessels, the local vascular autoregulation that control blood supply possible, results in the vulnerability of these organs to the vascular risk factors. Because it is a bidirectional system where each affects the other, it is best considered as a cerebro-renal unit.
Collapse
Affiliation(s)
- Kumar Rajamani
- Professor of Neurology, Wayne State University School of Medicine, Detroit, MI.
| |
Collapse
|
177
|
Parodi-Rullán R, Sone JY, Fossati S. Endothelial Mitochondrial Dysfunction in Cerebral Amyloid Angiopathy and Alzheimer's Disease. J Alzheimers Dis 2020; 72:1019-1039. [PMID: 31306129 DOI: 10.3233/jad-190357] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. Cerebrovascular dysfunction is one of the earliest events in the pathogenesis of AD, as well as in vascular and mixed dementias. Cerebral amyloid angiopathy (CAA), the deposition of amyloid around cerebral vessels, is observed in up to 90% of AD patients and in approximately 50% of elderly individuals over 80 years of age. CAA is a strong contributor to vascular dysfunction in AD. CAA-laden brain vessels are characterized by dysfunctional hemodynamics and leaky blood-brain barrier (BBB), contributing to clearance failure and further accumulation of amyloid-β (Aβ) in the cerebrovasculature and brain parenchyma. Mitochondrial dysfunction is increasingly recognized as an important early initiator of the pathogenesis of AD and CAA. The objective of this review is to discuss the effects of Aβ on cerebral microvascular cell function, focusing on its impact on endothelial mitochondria. After introducing CAA and its etiology and genetic risk factors, we describe the pathological relationship between cerebrovascular amyloidosis and brain microvascular endothelial cell dysfunction, critically analyzing its roles in disease progression, hypoperfusion, and BBB integrity. Then, we focus on discussing the effect of Aβ challenge on endothelial mitochondrial dysfunction pathways, and their contribution to the progression of neurovascular dysfunction in AD and dementia. Finally, we report potential pharmacological and non-pharmacological mitochondria-targeted therapeutic strategies which may help prevent or delay cerebrovascular failure.
Collapse
Affiliation(s)
- Rebecca Parodi-Rullán
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Je Yeong Sone
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, NY, USA
| | - Silvia Fossati
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
178
|
Schager B, Brown CE. Susceptibility to capillary plugging can predict brain region specific vessel loss with aging. J Cereb Blood Flow Metab 2020; 40:2475-2490. [PMID: 31903837 PMCID: PMC7820682 DOI: 10.1177/0271678x19895245] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/28/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022]
Abstract
Vessel loss in the aging brain is commonly reported, yet important questions remain concerning whether there are regional vulnerabilities and what mechanisms could account for these regional differences, if they exist. Here we imaged and quantified vessel length, tortuosity and width in 15 brain regions in young adult and aged mice. Our data indicate that vessel loss was most pronounced in white matter followed by cortical, then subcortical grey matter regions, while some regions (visual cortex, amygdala, thalamus) showed no decline with aging. Regions supplied by the anterior cerebral artery were more vulnerable to loss than those supplied by middle or posterior cerebral arteries. Vessel width and tortuosity generally increased with age but neither reliably predicted regional vessel loss. Since capillaries are naturally prone to plugging and prolonged obstructions often lead to vessel pruning, we hypothesized that regional susceptibilities to plugging could help predict vessel loss. By mapping the distribution of microsphere-induced capillary obstructions, we discovered that regions with a higher density of persistent obstructions were more likely to show vessel loss with aging and vice versa. These findings indicate that age-related vessel loss is region specific and can be explained, at least partially, by regional susceptibilities to capillary plugging.
Collapse
Affiliation(s)
- Ben Schager
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Craig E Brown
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
179
|
Notch1-mediated inflammation is associated with endothelial dysfunction in human brain microvascular endothelial cells upon particulate matter exposure. Arch Toxicol 2020; 95:529-540. [PMID: 33159583 DOI: 10.1007/s00204-020-02942-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 10/28/2020] [Indexed: 10/23/2022]
Abstract
Exposure to atmospheric particulate matter (PM) is an emerging risk factor for the pathogenesis of several diseases in humans, including cerebrovascular diseases. However, the mechanisms underlying PM-induced endothelial dysfunction are currently unclear. In this study, we examined how PM leads to endothelial dysfunction in human brain microvascular endothelial cells (HBMECs). We demonstrated that PM10 exposure (up to 25 μg/mL) increase Notch1 cleavage, and it regulates endothelial dysfunction through NICD-mediated inflammation and senescence. PM10-induced NICD signaling causes increased expression of interleukin-1 beta (IL-1β) and enhances characteristics of cellular senescence, which leads to increased endothelial permeability in HBMECs. Knockdown of Notch1 by siRNA blocks PM10-induced endothelial dysfunction via the suppression of inflammation and senescence. Furthermore, we found that Notch1-mediated inflammation accelerates endothelial senescence, which eventually leads to endothelial dysfunction. Altogether, our data suggest that Notch1 and NICD are potential target regulators for the prevention of cerebrovascular endothelial dysfunction induced by ambient air pollutants such as PM.
Collapse
|
180
|
Xiong L, Charidimou A, Pasi M, Boulouis G, Pongpitakmetha T, Schirmer MD, Singh S, Benson E, Gurol EM, Rosand J, Greenberg SM, Biffi A, Viswanathan A. Predictors for Late Post-Intracerebral Hemorrhage Dementia in Patients with Probable Cerebral Amyloid Angiopathy. J Alzheimers Dis 2020; 71:435-442. [PMID: 31403947 DOI: 10.3233/jad-190346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND OBJECTIVE Cerebral amyloid angiopathy (CAA) accounts for the majority of lobar intracerebral hemorrhage (ICH); however, the risk factors for dementia conversion after ICH occurrence in CAA patients are unknown, especially in the long-term period after ICH. Therefore, we aimed to unravel the predictors for late post-ICH dementia (6 months after ICH event) in probable CAA patients. METHODS From a large consecutive MRI prospective cohort of spontaneous ICH (2006-2017), we identified probable CAA patients (modified Boston criteria) without dementia 6 months post-ICH. Cognitive outcome during follow-up was determined based on the information from standardized clinical visit notes. We used Cox regression analysis to investigate the association between baseline demographic characteristics, past medical history, MRI biomarkers, and late post-ICH dementia conversion (dementia occurred after 6 months). RESULTS Among 97 non-demented lobar ICH patients with probable CAA, 25 patients (25.8%) developed dementia during a median follow-up time of 2.5 years (IQR 1.5-3.8 years). Pre-existing mild cognitive impairment, increased white matter hyperintensities (WMH) burden, the presence of disseminated cortical superficial siderosis (cSS), and higher total small vessel disease score for CAA were all independent predictors for late dementia conversion. CONCLUSION In probable CAA patients presenting with lobar ICH, high WMH burden and presence of disseminated cSS are useful neuroimaging biomarkers for dementia risk stratification. These findings have implications for clinical practice and future trial design.
Collapse
Affiliation(s)
- Li Xiong
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Andreas Charidimou
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Marco Pasi
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Gregoire Boulouis
- Centre Hospitalier Sainte-Anne, Université Paris Descartes, Paris, France
| | - Thanakit Pongpitakmetha
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA.,Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Markus D Schirmer
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA.,Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Boston, MA, USA.,Department of Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), Germany
| | - Sanjula Singh
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Emily Benson
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Edip M Gurol
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Jonathan Rosand
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Alessandro Biffi
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Anand Viswanathan
- Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
181
|
Halder SK, Milner R. Mild hypoxia triggers transient blood-brain barrier disruption: a fundamental protective role for microglia. Acta Neuropathol Commun 2020; 8:175. [PMID: 33115539 PMCID: PMC7592567 DOI: 10.1186/s40478-020-01051-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 12/11/2022] Open
Abstract
We recently demonstrated that when mice are exposed to chronic mild hypoxia (CMH, 8% O2), blood vessels in the spinal cord show transient vascular leak that is associated with clustering and activation of microglia around disrupted vessels. Importantly, microglial depletion profoundly increased hypoxia-induced vascular leak, implying that microglia play a critical role maintaining vascular integrity in the hypoxic spinal cord. The goal of the current study was to examine if microglia play a similar vasculo-protective function in the brain. Employing extravascular fibrinogen leak as an index of blood-brain barrier (BBB) disruption, we found that CMH provoked transient vascular leak in cerebral blood vessels that was associated with activation and aggregation of Mac-1-positive microglia around leaky vessels. Interestingly, CMH-induced vascular leak showed regional selectivity, being much more prevalent in the brainstem and olfactory bulb than the cerebral cortex and cerebellum. Pharmacological depletion of microglia with the colony stimulating factor-1 receptor inhibitor PLX5622, had no effect under normoxic conditions, but markedly increased hypoxia-induced cerebrovascular leak in all regions examined. As in the spinal cord, this was associated with endothelial induction of MECA-32, a marker of leaky CNS endothelium, and greater loss of endothelial tight junction proteins. Brain regions displaying the highest levels of hypoxic-induced vascular leak also showed the greatest levels of angiogenic remodeling, suggesting that transient BBB disruption may be an unwanted side-effect of hypoxic-induced angiogenic remodeling. As hypoxia is common to a multitude of human diseases including obstructive sleep apnea, lung disease, and age-related pulmonary, cardiac and cerebrovascular dysfunction, our findings have important translational implications. First, they point to a potential pathogenic role of chronic hypoxia in triggering BBB disruption and subsequent neurological dysfunction, and second, they demonstrate an important protective role for microglia in maintaining vascular integrity in the hypoxic brain.
Collapse
Affiliation(s)
- Sebok K. Halder
- San Diego Biomedical Research Institute, 10865 Road to the Cure, Suite 100, San Diego, CA 92121 USA
| | - Richard Milner
- San Diego Biomedical Research Institute, 10865 Road to the Cure, Suite 100, San Diego, CA 92121 USA
| |
Collapse
|
182
|
Pietroboni AM, Colombi A, Carandini T, Scarpini E, Galimberti D, Bozzali M. The Role of Amyloid-β in White Matter Damage: Possible Common Pathogenetic Mechanisms in Neurodegenerative and Demyelinating Diseases. J Alzheimers Dis 2020; 78:13-22. [PMID: 32925075 DOI: 10.3233/jad-200868] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Just as multiple sclerosis (MS) has long been primarily considered a white matter (WM) disease, Alzheimer's disease (AD) has for decades been regarded only as a grey matter disorder. However, convergent evidences have suggested that WM abnormalities are also important components of AD, at the same extent as axonal and neuronal loss is critically involved in MS pathophysiology since early clinical stages. These observations have motivated a more thorough investigation about the possible mechanisms that could link neuroinflammation and neurodegeneration, focusing on amyloid-β (Aβ). Neuroimaging studies have found that patients with AD have widespread WM abnormalities already at the earliest disease stages and prior to the presence of Aβ plaques. Moreover, a correlation between cerebrospinal fluid (CSF) Aβ levels and WM lesion load was found. On the other hand, recent studies suggest a predictive role for CSF Aβ levels in MS, possibly due in the first instance to the reduced capacity for remyelination, consequently to a higher risk of WM damage progression, and ultimately to neuronal loss. We undertook a review of the recent findings concerning the involvement of CSF Aβ levels in the MS disease course and of the latest evidence of AD related WM abnormalities, with the aim to discuss the potential causes that may connect WM damage and amyloid pathology.
Collapse
Affiliation(s)
- Anna M Pietroboni
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,University of Milan, Dino Ferrari Centre, Milan, Italy
| | | | - Tiziana Carandini
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,University of Milan, Dino Ferrari Centre, Milan, Italy
| | - Elio Scarpini
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,University of Milan, Dino Ferrari Centre, Milan, Italy
| | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,University of Milan, Dino Ferrari Centre, Milan, Italy
| | - Marco Bozzali
- Department of Neuroscience 'Rita Levi Montalcini', University of Torino, Turin, Italy.,Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| |
Collapse
|
183
|
Hill LK, Hoang DM, Chiriboga LA, Wisniewski T, Sadowski MJ, Wadghiri YZ. Detection of Cerebrovascular Loss in the Normal Aging C57BL/6 Mouse Brain Using in vivo Contrast-Enhanced Magnetic Resonance Angiography. Front Aging Neurosci 2020; 12:585218. [PMID: 33192479 PMCID: PMC7606987 DOI: 10.3389/fnagi.2020.585218] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/11/2020] [Indexed: 12/28/2022] Open
Abstract
Microvascular rarefaction, or the decrease in vascular density, has been described in the cerebrovasculature of aging humans, rats, and, more recently, mice in the presence and absence of age-dependent diseases. Given the wide use of mice in modeling age-dependent human diseases of the cerebrovasculature, visualization, and quantification of the global murine cerebrovasculature is necessary for establishing the baseline changes that occur with aging. To provide in vivo whole-brain imaging of the cerebrovasculature in aging C57BL/6 mice longitudinally, contrast-enhanced magnetic resonance angiography (CE-MRA) was employed using a house-made gadolinium-bearing micellar blood pool agent. Enhancement in the vascular space permitted quantification of the detectable, or apparent, cerebral blood volume (aCBV), which was analyzed over 2 years of aging and compared to histological analysis of the cerebrovascular density. A significant loss in the aCBV was detected by CE-MRA over the aging period. Histological analysis via vessel-probing immunohistochemistry confirmed a significant loss in the cerebrovascular density over the same 2-year aging period, validating the CE-MRA findings. While these techniques use widely different methods of assessment and spatial resolutions, their comparable findings in detected vascular loss corroborate the growing body of literature describing vascular rarefaction aging. These findings suggest that such age-dependent changes can contribute to cerebrovascular and neurodegenerative diseases, which are modeled using wild-type and transgenic laboratory rodents.
Collapse
Affiliation(s)
- Lindsay K. Hill
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, NY, United States
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), NYU Grossman School of Medicine, New York, NY, United States
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, NYU Grossman School of Medicine, New York, NY, United States
- Department of Biomedical Engineering, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Dung Minh Hoang
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), NYU Grossman School of Medicine, New York, NY, United States
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, NYU Grossman School of Medicine, New York, NY, United States
| | - Luis A. Chiriboga
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, United States
| | - Thomas Wisniewski
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, United States
| | - Martin J. Sadowski
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, United States
| | - Youssef Z. Wadghiri
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), NYU Grossman School of Medicine, New York, NY, United States
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, NYU Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
184
|
Menet R, Bourassa P, Calon F, ElAli A. Dickkopf-related protein-1 inhibition attenuates amyloid-beta pathology associated to Alzheimer's disease. Neurochem Int 2020; 141:104881. [PMID: 33068684 DOI: 10.1016/j.neuint.2020.104881] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) constitutes the leading cause of dementia worldwide. It is associated to amyloid-β (Aβ) aggregation and tau hyper-phosphorylation, accompanied by a progressive cognitive decline. Evidence suggests that the canonical Wnt pathway is deregulated in AD. Pathway activity is mediated by β-catenin stabilization in the cytosol, and subsequent translocation to the nucleus to regulate the expression of several genes implicated in brain homeostasis and functioning. It was recently proposed that Dickkopf-related protein-1 (DKK1), an endogenous antagonist of the pathway, might be implicated in AD pathogenesis. Here, we hypothesized that canonical Wnt pathway deactivation associated to DKK1 induction contributes to late-onset AD pathogenesis, and thus DKK1 neutralization could attenuate AD pathology. For this purpose, human post-mortem AD brain samples were used to assess pathway activity, and aged APPswe/PS1 mice were used to investigate DKK1 in late-onset AD-like pathology and therapy. Our findings indicate that β-catenin levels progressively decrease in the brain of AD patients, correlating with the duration of symptoms. Next, we found that Aβ pathology in APPswe/PS1 mediates DKK1 induction in the brain. Pharmacological neutralization of DKK1's biological activity in APPswe/PS1 mice restores pathway activity by stabilizing β-catenin, attenuates Aβ pathology, and ameliorates the memory of mice. Attenuation of AD-like pathology upon DKK1 inhibition is accompanied by a reduced protein expression of beta-site amyloid precursor protein (APP) cleaving enzyme-1 (BACE1). Moreover, DKK1 inhibition enhances vascular density, promotes blood-brain barrier (BBB) integrity by increasing claudin 5, glucose transporter-1 (GLUT1), and ATP-binding cassette sub-family B member-1 (ABCB1) protein expression, as well as ameliorates synaptic plasticity by increasing brain-derived neurotrophic factor (BDNF), and postsynaptic density protein-95 (PSD-95) protein expression. DKK1 conditional induction reduces claudin 5, abcb1, and psd-95 mRNA expression, validating its inhibition effects. Our results indicate that neutralization of DKK1's biological activity attenuates AD-like pathology by restoring canonical Wnt pathway activity.
Collapse
Affiliation(s)
- Romain Menet
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Philippe Bourassa
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
| | - Frédéric Calon
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
185
|
Sadeghmousavi S, Eskian M, Rahmani F, Rezaei N. The effect of insomnia on development of Alzheimer's disease. J Neuroinflammation 2020; 17:289. [PMID: 33023629 PMCID: PMC7542374 DOI: 10.1186/s12974-020-01960-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia and a neurodegenerative disorder characterized by memory deficits especially forgetting recent information, recall ability impairment, and loss of time tracking, problem-solving, language, and recognition difficulties. AD is also a globally important health issue but despite all scientific efforts, the treatment of AD is still a challenge. Sleep has important roles in learning and memory consolidation. Studies have shown that sleep deprivation (SD) and insomnia are associated with the pathogenesis of Alzheimer's disease and may have an impact on the symptoms and development. Thus, sleep disorders have decisive effects on AD; this association deserves more attention in research, diagnostics, and treatment, and knowing this relation also can help to prevent AD through screening and proper management of sleep disorders. This study aimed to show the potential role of SD and insomnia in the pathogenesis and progression of AD.
Collapse
Affiliation(s)
- Shaghayegh Sadeghmousavi
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Eskian
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farzaneh Rahmani
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Nima Rezaei
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
186
|
Bisht I, Ambasta RK, Kumar P. An integrated approach to unravel a putative crosstalk network in Alzheimer's disease and Parkinson's disease. Neuropeptides 2020; 83:102078. [PMID: 32807513 DOI: 10.1016/j.npep.2020.102078] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/07/2020] [Accepted: 08/09/2020] [Indexed: 12/31/2022]
Abstract
Integration of multiple profiling data and construction of functional regulatory networks provide a powerful approach to uncover functional relationships and significant molecular entities from transcriptomic data, highlighting the molecular mechanisms of complex diseases. Despite having an overlap in the neuropathologies of AD and PD, the molecular entities overlapped and mechanisms behind them are less known. Here we used an integrated strategy to analyze miRNA and gene transcriptomic data to understand the role of miRNAs and genes in regulatory activities taking place in cells, and find transcriptomic signatures linking AD and PD. We preprocessed and analyzed publicly available microarray datasets and identified 97 DEGs and 21 DEmiRs that may be involved in the overlapped mechanisms between these two disorders. Among the DEGs, we found HSPA9, PGK1, SDHC, FH, DLD, YWHAZ and ACLY as the major protein-coding genes involved in the crosstalk for AD-PD pathogenesis. Further we integrated these DEGs and DEmiRs with regulatory TFs to construct an overlapped dysregulated network of AD and PD. In the network, miR-27a-3p, miR-148a-3p and miR-15a-5p were found to be the most relevant with maximum interactions, describing their significance in the potential crosstalk. We also looked into the dysregulated biological processes and pathways overlapped in AD and PD. In conclusion, we highlighted the DEGs, DEmiRs, their interactions and related pathways overlapped in AD and PD pathogenesis, also describing a potential crosstalk at molecular level. Besides, our findings can further be used for molecular studies to reveal an assured AD-PD crosstalk.
Collapse
Affiliation(s)
- Indu Bisht
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly DCE), Delhi, 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly DCE), Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly DCE), Delhi, 110042, India.
| |
Collapse
|
187
|
Shah A, Apte RS. Optical Coherence Tomography Angiography: A Window into Central Nervous System Neurodegeneration. Trends Mol Med 2020; 26:892-895. [DOI: 10.1016/j.molmed.2020.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/26/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022]
|
188
|
Watanabe C, Imaizumi T, Kawai H, Suda K, Honma Y, Ichihashi M, Ema M, Mizutani KI. Aging of the Vascular System and Neural Diseases. Front Aging Neurosci 2020; 12:557384. [PMID: 33132896 PMCID: PMC7550630 DOI: 10.3389/fnagi.2020.557384] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/01/2020] [Indexed: 12/14/2022] Open
Abstract
Vertebrates have acquired complex high-order functions facilitated by the dispersion of vascular and neural networks to every corner of the body. Blood vessels deliver oxygen and nutrients to all cells and provide essential transport systems for removing waste products. For these functions, tissue vascularization must be spatiotemporally appropriate. Recent studies revealed that blood vessels create a tissue-specific niche, thus attracting attention as biologically active sites for tissue development. Each capillary network is critical for maintaining proper brain function because age-related and disease-related impairment of cognitive function is associated with the loss or diminishment of brain capillaries. This review article highlights how structural and functional alterations in the brain vessels may change with age and neurogenerative diseases. Capillaries are also responsible for filtering toxic byproducts, providing an appropriate vascular environment for neuronal function. Accumulation of amyloid β is a key event in Alzheimer’s disease pathogenesis. Recent studies have focused on associations reported between Alzheimer’s disease and vascular aging. Furthermore, the glymphatic system and meningeal lymphatic systems contribute to a functional unit for clearance of amyloid β from the brain from the central nervous system into the cervical lymph nodes. This review article will also focus on recent advances in stem cell therapies that aim at repopulation or regeneration of a degenerating vascular system for neural diseases.
Collapse
Affiliation(s)
- Chisato Watanabe
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan.,Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, Japan
| | - Tsutomu Imaizumi
- Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Osaka, Japan
| | - Hiromi Kawai
- Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Osaka, Japan
| | - Kazuma Suda
- Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Osaka, Japan
| | - Yoichi Honma
- Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Osaka, Japan
| | - Masamitsu Ichihashi
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, Japan.,Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University Institute for Advanced Study, Kyoto, Japan
| | - Ken-Ichi Mizutani
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| |
Collapse
|
189
|
Damodarasamy M, Vernon RB, Pathan JL, Keene CD, Day AJ, Banks WA, Reed MJ. The microvascular extracellular matrix in brains with Alzheimer's disease neuropathologic change (ADNC) and cerebral amyloid angiopathy (CAA). Fluids Barriers CNS 2020; 17:60. [PMID: 32993718 PMCID: PMC7525948 DOI: 10.1186/s12987-020-00219-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/09/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The microvasculature (MV) of brains with Alzheimer's disease neuropathologic change (ADNC) and cerebral amyloid angiopathy (CAA), in the absence of concurrent pathologies (e.g., infarctions, Lewy bodies), is incompletely understood. OBJECTIVE To analyze microvascular density, diameter and extracellular matrix (ECM) content in association with ADNC and CAA. METHODS We examined samples of cerebral cortex and isolated brain microvasculature (MV) from subjects with the National Institute on Aging-Alzheimer's Association (NIA-AA) designations of not-, intermediate-, or high ADNC and from subjects with no CAA and moderate-severe CAA. Cases for all groups were selected with no major (territorial) strokes, ≤ 1 microinfarct in screening sections, and no Lewy body pathology. MV density and diameter were measured from cortical brain sections. Levels of basement membrane (BM) ECM components, the protein product of TNF-stimulated gene-6 (TSG-6), and the ubiquitous glycosaminoglycan hyaluronan (HA) were assayed by western blots or HA ELISA of MV lysates. RESULTS We found no significant changes in MV density or diameter among any of the groups. Levels of BM laminin and collagen IV (col IV) were lower in MV isolated from the high ADNC vs. not-ADNC groups. In contrast, BM laminin was significantly higher in MV from the moderate-severe CAA vs. the no CAA groups. TSG-6 and HA content were higher in the presence of both high ADNC and CAA, whereas levels of BM fibronectin and perlecan were similar among all groups. CONCLUSIONS Cortical MV density and diameter are not appreciably altered by ADNC or CAA. TSG-6 and HA are increased in both ADNC and CAA, with laminin and col IV decreased in the BM of high ADNC, but laminin increased in moderate-severe CAA. These results show that changes in the ECM occur in AD and CAA, but independently of one another, and likely reflect on the regional functioning of the brain microvasculature.
Collapse
Affiliation(s)
- Mamatha Damodarasamy
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Robert B Vernon
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Jasmine L Pathan
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - C Dirk Keene
- Division of Neuropathology, Department of Pathology, University of Washington, Seattle, WA, USA
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research and Lydia Becker Institute of Immunology and Inflammation, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - William A Banks
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - May J Reed
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA.
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA.
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington Harborview Medical Center, Seattle, WA, 98104, USA.
| |
Collapse
|
190
|
Do CT, Manjaly ZM, Heinzle J, Schöbi D, Kasper L, Pruessmann KP, Stephan KE, Frässle S. Hemodynamic modeling of long-term aspirin effects on blood oxygenated level dependent responses at 7 Tesla in patients at cardiovascular risk. Eur J Neurosci 2020; 53:1262-1278. [PMID: 32936980 DOI: 10.1111/ejn.14970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 08/06/2020] [Accepted: 09/02/2020] [Indexed: 11/29/2022]
Abstract
Aspirin is considered a potential confound for functional magnetic resonance imaging (fMRI) studies. This is because aspirin affects the synthesis of prostaglandin, a vasoactive mediator centrally involved in neurovascular coupling, a process underlying blood oxygenated level dependent (BOLD) responses. Aspirin-induced changes in BOLD signal are a potential confound for fMRI studies of at-risk individuals or patients (e.g. with cardiovascular conditions or stroke) who receive low-dose aspirin prophylactically and are compared to healthy controls without aspirin. To examine the severity of this potential confound, we combined high field (7 Tesla) MRI during a simple hand movement task with a biophysically informed hemodynamic model. We compared elderly individuals receiving aspirin for primary or secondary prophylactic purposes versus age-matched volunteers without aspirin medication, testing for putative differences in BOLD responses. Specifically, we fitted hemodynamic models to BOLD responses from 14 regions activated by the task and examined whether model parameter estimates were significantly altered by aspirin. While our analyses indicate that hemodynamics differed across regions, consistent with the known regional variability of BOLD responses, we neither found a significant main effect of aspirin (i.e., an average effect across brain regions) nor an expected drug × region interaction. While our sample size is not sufficiently large to rule out small-to-medium global effects of aspirin, we had adequate statistical power for detecting the expected interaction. Altogether, our analysis suggests that patients with cardiovascular risk receiving low-dose aspirin for primary or secondary prophylactic purposes do not show strongly altered BOLD signals when compared to healthy controls without aspirin.
Collapse
Affiliation(s)
- Cao-Tri Do
- Translational Neuromodeling Unit, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Zina-Mary Manjaly
- Department of Neurology, Schulthess Clinic, Zurich, Switzerland.,Department of Health Science and Technology (D-HEST), ETH Zurich, Zurich, Switzerland
| | - Jakob Heinzle
- Translational Neuromodeling Unit, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Dario Schöbi
- Translational Neuromodeling Unit, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Lars Kasper
- Translational Neuromodeling Unit, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.,MR Technology Group, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Klaas P Pruessmann
- MR Technology Group, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Klaas Enno Stephan
- Translational Neuromodeling Unit, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.,Wellcome Centre for Human Neuroimaging, University College London, London, UK.,Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Stefan Frässle
- Translational Neuromodeling Unit, Institute of Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| |
Collapse
|
191
|
Asanad S, Mohammed I, Sadun AA, Saeedi OJ. OCTA in neurodegenerative optic neuropathies: emerging biomarkers at the eye-brain interface. Ther Adv Ophthalmol 2020; 12:2515841420950508. [PMID: 32923939 PMCID: PMC7457690 DOI: 10.1177/2515841420950508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 07/13/2020] [Indexed: 12/22/2022] Open
Abstract
OCTA imaging in optic neuropathies.
Collapse
Affiliation(s)
- Samuel Asanad
- Department of Ophthalmology and Visual Sciences, University of Maryland Eye Associates, University of Maryland Medical Center and University of Maryland School of Medicine, 419 W. Redwood St., Baltimore, MD 21201, USA
| | - Isa Mohammed
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alfredo A Sadun
- Doheny Eye Center, Los Angeles, CA, USA; Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Osamah J Saeedi
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
192
|
Xu X, Meng T, Wen Q, Tao M, Wang P, Zhong K, Shen Y. Dynamic changes in vascular size and density in transgenic mice with Alzheimer's disease. Aging (Albany NY) 2020; 12:17224-17234. [PMID: 32908022 PMCID: PMC7521516 DOI: 10.18632/aging.103672] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 06/29/2020] [Indexed: 01/24/2023]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases. Here, we used vessel size imaging to investigate the specific microvascular changes and most susceptible brain regions during AD progression in an amyloid precursor protein 23 (APP23) transgenic AD mouse model. Using 9.4 Tesla magnetic resonance imaging (MRI), the values of microvascular density (Density), mean vessel diameter (mVD), and vessel size index (VSI) were compared between APP23 and wild-type (WT) mice at 3, 6, 9, 14, and 20 months of age. Our results demonstrate that in 20-month old APP23 and WT mice, the Density values were significantly decreased, while the vascular dilatation and diameter had increased. However, a transient increase in the cortex Density at 14-months was observed in APP23 mice. Additionally, our results suggest that the hippocampus is the susceptible brain region affected by the abnormal microvascular angiogenesis during the early stages of AD. Together, our findings indicate that vessel size imaging using MRI can provide novel biomarkers for the early detection of AD, and for monitoring the effects of vascular-targeted therapeutics in AD.
Collapse
Affiliation(s)
- Xiaowen Xu
- Institute on Aging and Brain Disorders, First University Affiliated Hospital, Neurodegenerative Disorder Research Center, Division of Life and Medical Sciences, University of Science and Technology of China, Hefei Material Science National Laboratory at Microscale, CAS-Key Laboratory of Brain Functions and Brain Disorders, Center for Excellent in Brain Science and Intelligence Technology, Hefei, China,Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China,School of Medicine, Tongji University, Shanghai, China
| | - Tong Meng
- School of Medicine, Tongji University, Shanghai, China
| | - Qingqing Wen
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mengling Tao
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China,School of Medicine, Tongji University, Shanghai, China
| | - Peijun Wang
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China,School of Medicine, Tongji University, Shanghai, China
| | - Kai Zhong
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, China,Key Laboratory of Anhui Province for High Field Magnetic Resonance Imaging, Hefei, China,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yong Shen
- Institute on Aging and Brain Disorders, First University Affiliated Hospital, Neurodegenerative Disorder Research Center, Division of Life and Medical Sciences, University of Science and Technology of China, Hefei Material Science National Laboratory at Microscale, CAS-Key Laboratory of Brain Functions and Brain Disorders, Center for Excellent in Brain Science and Intelligence Technology, Hefei, China
| |
Collapse
|
193
|
Mizutani KI. [Neuro-vascular Interactions during Neocortical Development-Systematic and Accurate Regulatory Mechanisms of VEGF Signaling]. YAKUGAKU ZASSHI 2020; 140:521-527. [PMID: 32238635 DOI: 10.1248/yakushi.19-00221-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Blood vessels supply oxygen and nutrients to all the cells in a living body, and provide essential transport routes for collecting waste products. For these functions, blood vessel networks should be appropriately formed in each tissue. Therefore, blood vessels are one of the earliest organs formed during the developmental process. Development of the blood vessel system promotes tissue differentiation and organ morphogenesis, allowing each organ to maintain its unique functions under changing metabolic conditions. Blood vessels have a relatively simple structure, consisting of endothelial cells covering the inner layer, and pericytes or smooth muscle cells surrounding the outside. The structure of the vascular network is extremely diverse, with blood vessels uniquely organized depending on the tissues they serve, to create tissue-specific microenvironments. How are such tissue-specific vascular environments generated? Over the years, anatomical findings have accumulated to confirm this vascular diversity. However, the molecular basis for this diversity has remained unclear. In the present article, we review the mechanisms of coordinated developmental control of the vascular and neural systems in the cerebral cortex from the viewpoint of the accurate expression control of vascular endothelial growth factor (VEGF) signaling, and describe future perspectives.
Collapse
Affiliation(s)
- Ken-Ichi Mizutani
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University
| |
Collapse
|
194
|
Anderson VC, Tagge IJ, Li X, Quinn JF, Kaye JA, Bourdette DN, Spain RI, Riccelli LP, Sammi MK, Springer CS, Rooney WD. Observation of Reduced Homeostatic Metabolic Activity and/or Coupling in White Matter Aging. J Neuroimaging 2020; 30:658-665. [PMID: 32558031 PMCID: PMC7529981 DOI: 10.1111/jon.12744] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Transvascular water exchange plays a key role in the functional integrity of the blood-brain barrier (BBB). In white matter (WM), a variety of imaging modalities have demonstrated age-related changes in structure and metabolism, but the extent to which water exchange is altered remains unclear. Here, we investigated the cumulative effects of healthy aging on WM capillary water exchange. METHODS A total of 38 healthy adults (aged 36-80 years) were studied using 7T dynamic contrast enhanced MRI. Blood volume fraction (vb ) and capillary water efflux rate constant (kpo ) were determined by fitting changes in the 1 H2 O longitudinal relaxation rate constant (R1 ) during contrast agent bolus passage to a two-compartment exchange model. WM volume was determined by morphometric analysis of structural images. RESULTS R1 values and WM volume showed similar trajectories of age-related decline. Among all subjects, vb and kpo averaged 1.7 (±0.5) mL/100 g of tissue and 2.1 (±1.1) s-1 , respectively. While vb showed minimal changes over the 40-year-age span of participants, kpo declined 0.06 s-1 (ca. 3%) per year (r = -.66; P < .0005), from near 4 s-1 at age 30 to ca. 2 s-1 at age 70. The association remained significant after controlling for WM volume. CONCLUSIONS Previous studies have shown that kpo tracks Na+ , K+ -ATPase activity-dependent water exchange at the BBB and likely reflects neurogliovascular unit (NGVU) coupled metabolic activity. The age-related decline in kpo observed here is consistent with compromised NGVU metabolism in older individuals and the dysregulated cellular bioenergetics that accompany normal brain aging.
Collapse
Affiliation(s)
- Valerie C Anderson
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR
| | - Ian J Tagge
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR
| | - Xin Li
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR
| | - Joseph F Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Jeffrey A Kaye
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Dennis N Bourdette
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Rebecca I Spain
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Louis P Riccelli
- Diagnostic Radiology, Oregon Health & Science University, Portland, OR
| | - Manoj K Sammi
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR
| | - Charles S Springer
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR
| | - William D Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR
| |
Collapse
|
195
|
Viscusi ER, Viscusi AR. Blood-brain barrier: mechanisms governing permeability and interaction with peripherally acting μ-opioid receptor antagonists. Reg Anesth Pain Med 2020; 45:688-695. [PMID: 32723840 PMCID: PMC7476292 DOI: 10.1136/rapm-2020-101403] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/13/2022]
Abstract
The blood-brain barrier (BBB) describes the unique properties of endothelial cells (ECs) that line the central nervous system (CNS) microvasculature. The BBB supports CNS homeostasis via EC-associated transport of ions, nutrients, proteins and waste products between the brain and blood. These transport mechanisms also serve as physiological barriers to pathogens, toxins and xenobiotics to prevent them from contacting neural tissue. The mechanisms that govern BBB permeability pose a challenge to drug design for CNS disorders, including pain, but can be exploited to limit the effects of a drug to the periphery, as in the design of the peripherally acting μ-opioid receptor antagonists (PAMORAs) used to treat opioid-induced constipation. Here, we describe BBB physiology, drug properties that affect BBB penetrance and how data from randomized clinical trials of PAMORAs improve our understanding of BBB permeability.
Collapse
Affiliation(s)
- Eugene R Viscusi
- Department of Anesthesiology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Andrew R Viscusi
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
196
|
Falangola MF, Nie X, Ward R, McKinnon ET, Dhiman S, Nietert PJ, Helpern JA, Jensen JH. Diffusion MRI detects early brain microstructure abnormalities in 2-month-old 3×Tg-AD mice. NMR IN BIOMEDICINE 2020; 33:e4346. [PMID: 32557874 PMCID: PMC7683375 DOI: 10.1002/nbm.4346] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/08/2020] [Accepted: 05/20/2020] [Indexed: 06/11/2023]
Abstract
The 3×Tg-AD mouse is one of the most studied animal models of Alzheimer's disease (AD), and develops both amyloid beta deposits and neurofibrillary tangles in a temporal and spatial pattern that is similar to human AD pathology. Additionally, abnormal myelination patterns with changes in oligodendrocyte and myelin marker expression are reported to be an early pathological feature in this model. Only few diffusion MRI (dMRI) studies have investigated white matter abnormalities in 3×Tg-AD mice, with inconsistent results. Thus, the goal of this study was to investigate the sensitivity of dMRI to capture brain microstructural alterations in 2-month-old 3×Tg-AD mice. In the fimbria, the fractional anisotropy (FA), kurtosis fractional anisotropy (KFA), and radial kurtosis (K┴ ) were found to be significantly lower in 3×Tg-AD mice than in controls, while the mean diffusivity (MD) and radial diffusivity (D┴ ) were found to be elevated. In the fornix, K┴ was lower for 3×Tg-AD mice; in the dorsal hippocampus MD and D┴ were elevated, as were FA, MD, and D┴ in the ventral hippocampus. These results indicate, for the first time, dMRI changes associated with myelin abnormalities in young 3×Tg-AD mice, before they develop AD pathology. Morphological quantification of myelin basic protein immunoreactivity in the fimbria was significantly lower in the 3×Tg-AD mice compared with the age-matched controls. Our results demonstrate that dMRI is able to detect widespread, significant early brain morphological abnormalities in 2-month-old 3×Tg-AD mice.
Collapse
Affiliation(s)
- Maria Fatima Falangola
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, US
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, South Carolina, US
| | - Xingju Nie
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, US
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, South Carolina, US
| | - Ralph Ward
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, US
| | - Emilie T McKinnon
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, US
- Department of Neurology, Medical University of South Carolina, Charleston, South Carolina, US
| | - Siddhartha Dhiman
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, US
| | - Paul J Nietert
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, US
| | - Joseph A Helpern
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, US
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, South Carolina, US
| | - Jens H Jensen
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, US
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, South Carolina, US
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, South Carolina, US
| |
Collapse
|
197
|
Wang T, Wilkes DM, Li M, Wu X, Gore JC, Ding Z. Hemodynamic Response Function in Brain White Matter in a Resting State. Cereb Cortex Commun 2020; 1:tgaa056. [PMID: 33073237 PMCID: PMC7552822 DOI: 10.1093/texcom/tgaa056] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/24/2020] [Accepted: 08/24/2020] [Indexed: 11/14/2022] Open
Abstract
The hemodynamic response function (HRF) characterizes temporal variations of blood oxygenation level-dependent (BOLD) signals. Although a variety of HRF models have been proposed for gray matter responses to functional demands, few studies have investigated HRF profiles in white matter particularly under resting conditions. In the present work we quantified the nature of the HRFs that are embedded in resting state BOLD signals in white matter, and which modulate the temporal fluctuations of baseline signals. We demonstrate that resting state HRFs in white matter could be derived by referencing to intrinsic avalanches in gray matter activities, and the derived white matter HRFs had reduced peak amplitudes and delayed peak times as compared with those in gray matter. Distributions of the time delays and correlation profiles in white matter depend on gray matter activities as well as white matter tract distributions, indicating that resting state BOLD signals in white matter encode neural activities associated with those of gray matter. This is the first investigation of derivations and characterizations of resting state HRFs in white matter and their relations to gray matter activities. Findings from this work have important implications for analysis of BOLD signals in the brain.
Collapse
Affiliation(s)
- Ting Wang
- Department of Computer Science, Chengdu University of Information Technology, Chengdu, Sichuan 610225, China
- Institute of Imaging Science, Vanderbilt University, Nashville, TN 37232, USA
| | - D Mitchell Wilkes
- Department of Electrical Engineering & Computer Science, Vanderbilt University, Nashville, TN 37232, USA
| | - Muwei Li
- Institute of Imaging Science, Vanderbilt University, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Xi Wu
- Department of Computer Science, Chengdu University of Information Technology, Chengdu, Sichuan 610225, China
| | - John C Gore
- Institute of Imaging Science, Vanderbilt University, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA
| | - Zhaohua Ding
- Institute of Imaging Science, Vanderbilt University, Nashville, TN 37232, USA
- Department of Electrical Engineering & Computer Science, Vanderbilt University, Nashville, TN 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA
| |
Collapse
|
198
|
Maliszewska-Cyna E, Vecchio LM, Thomason LAM, Oore JJ, Steinman J, Joo IL, Dorr A, McLaurin J, Sled JG, Stefanovic B, Aubert I. The effects of voluntary running on cerebrovascular morphology and spatial short-term memory in a mouse model of amyloidosis. Neuroimage 2020; 222:117269. [PMID: 32818618 DOI: 10.1016/j.neuroimage.2020.117269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/27/2022] Open
Abstract
Physical activity has been correlated with a reduced risk of cognitive decline, including that associated with vascular dementia, mild cognitive impairment (MCI) and Alzheimer's disease (AD); recent literature suggests this may in part result from benefits to the cerebrovascular network. Using a transgenic (Tg) mouse model of AD, we evaluated the effect of running on cortical and hippocampal vascular morphology, cerebral amyloid angiopathy, amyloid plaque load, and spatial memory. TgCRND8 mice present with progressive amyloid pathology, advancing from the cortex to the hippocampus in a time-dependent manner. We postulated that the characteristic progression of pathology could lead to differential, time-dependent effects of physical activity on vascular morphology in these brain regions at 6 months of age. We used two-photon fluorescent microscopy and 3D vessel tracking to characterize vascular and amyloid pathology in sedentary TgCRND8 mice compared those who have a history of physical activity (unlimited access to a running wheel, from 3 to 6 months of age). In sedentary TgCRND8 mice, capillary density was found to be lower in the cortex and higher in the hippocampus compared to non-transgenic (nonTg) littermates. Capillary length, vessel branching, and non-capillary vessel tortuosity were also higher in the hippocampus of sedentary TgCRND8 compared to nonTg mice. Three months of voluntary running resulted in normalizing cortical and hippocampal microvascular morphology, with no significant difference between TgCRND8 and nonTg mice. The benefits of physical activity on cortical and hippocampal vasculature in 6-month old TgCRND8 mice were not paralleled by significant changes on parenchymal and cerebral amyloid pathology. Short-term spatial memory- as evaluated by performance in the Y-maze- was significantly improved in running compared to sedentary TgCRND8 mice. These results suggest that long-term voluntary running contributes to the maintenance of vascular morphology and spatial memory in TgCRND8 mice, even in the absence of an effect on amyloid pathology.
Collapse
Affiliation(s)
- Ewelina Maliszewska-Cyna
- Hurvitz Brain Sciences, Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, S112, Toronto, Ontario M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Laura M Vecchio
- Hurvitz Brain Sciences, Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, S112, Toronto, Ontario M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada.
| | - Lynsie A M Thomason
- Hurvitz Brain Sciences, Physical Sciences, Sunnybrook Research Institute, Canada
| | - Jonathan J Oore
- Hurvitz Brain Sciences, Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, S112, Toronto, Ontario M4N 3M5, Canada
| | - Joe Steinman
- Mouse Imaging Centre, Hospital for Sick Children, Canada; Department of Medical Biophysics, University of Toronto, Canada
| | - Illsung Lewis Joo
- Hurvitz Brain Sciences, Physical Sciences, Sunnybrook Research Institute, Canada
| | - Adrienne Dorr
- Hurvitz Brain Sciences, Physical Sciences, Sunnybrook Research Institute, Canada
| | - JoAnne McLaurin
- Hurvitz Brain Sciences, Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, S112, Toronto, Ontario M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - John G Sled
- Mouse Imaging Centre, Hospital for Sick Children, Canada; Department of Medical Biophysics, University of Toronto, Canada
| | - Bojana Stefanovic
- Hurvitz Brain Sciences, Physical Sciences, Sunnybrook Research Institute, Canada; Department of Medical Biophysics, University of Toronto, Canada
| | - Isabelle Aubert
- Hurvitz Brain Sciences, Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, S112, Toronto, Ontario M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
199
|
Muhire G, Iulita MF, Vallerand D, Youwakim J, Gratuze M, Petry FR, Planel E, Ferland G, Girouard H. Arterial Stiffness Due to Carotid Calcification Disrupts Cerebral Blood Flow Regulation and Leads to Cognitive Deficits. J Am Heart Assoc 2020; 8:e011630. [PMID: 31057061 PMCID: PMC6512142 DOI: 10.1161/jaha.118.011630] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Arterial stiffness is associated with cognitive decline and dementia; however, the precise mechanisms by which it affects the brain remain unclear. Methods and Results Using a mouse model based on carotid calcification this study characterized mechanisms that could contribute to brain degeneration due to arterial stiffness. At 2 weeks postcalcification, carotid stiffness attenuated resting cerebral blood flow in several brain regions including the perirhinal/entorhinal cortex, hippocampus, and thalamus, determined by autoradiography (P<0.05). Carotid calcification impaired cerebral autoregulation and diminished cerebral blood flow responses to neuronal activity and to acetylcholine, examined by laser Doppler flowmetry (P<0.05, P<0.01). Carotid stiffness significantly affected spatial memory at 3 weeks (P<0.05), but not at 2 weeks, suggesting that cerebrovascular impairments precede cognitive dysfunction. In line with the endothelial deficits, carotid stiffness led to increased blood‐brain barrier permeability in the hippocampus (P<0.01). This region also exhibited reductions in vessel number containing collagen IV (P<0.01), as did the somatosensory cortex (P<0.05). No evidence of cerebral microhemorrhages was present. Carotid stiffness did not affect the production of mouse amyloid‐β (Aβ) or tau phosphorylation, although it led to a modest increase in the Aβ40/Aβ42 ratio in frontal cortex (P<0.01). Conclusions These findings suggest that carotid stiffness alters brain microcirculation and increases blood‐brain barrier permeability associated with cognitive impairments. Therefore, arterial stiffness should be considered a relevant target to protect the brain and prevent cognitive dysfunctions.
Collapse
Affiliation(s)
- Gervais Muhire
- 1 Département de Pharmacologie et Physiologie Université de Montréal Québec Canada
| | - M Florencia Iulita
- 2 Groupe de Recherche sur le Système Nerveux Central Université de Montréal Québec Canada.,3 Département de Neurosciences Université de Montréal Québec Canada
| | - Diane Vallerand
- 1 Département de Pharmacologie et Physiologie Université de Montréal Québec Canada
| | - Jessica Youwakim
- 1 Département de Pharmacologie et Physiologie Université de Montréal Québec Canada
| | - Maud Gratuze
- 4 Département de Psychiatrie et Neurosciences Université Laval Québec Québec Canada
| | - Franck R Petry
- 4 Département de Psychiatrie et Neurosciences Université Laval Québec Québec Canada
| | - Emmanuel Planel
- 4 Département de Psychiatrie et Neurosciences Université Laval Québec Québec Canada.,5 Centre de Recherche du CHU de Québec Québec Canada
| | - Guylaine Ferland
- 6 Département de Nutrition Université de Montréal Québec Canada.,7 Centre de Recherche de l'Institut de Cardiologie de Montréal Montréal Québec Canada
| | - Hélène Girouard
- 1 Département de Pharmacologie et Physiologie Université de Montréal Québec Canada.,2 Groupe de Recherche sur le Système Nerveux Central Université de Montréal Québec Canada.,8 Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal Montréal Québec Canada
| |
Collapse
|
200
|
Geraets AF, van Agtmaal MJ, Stehouwer CD, Sörensen BM, Berendschot TT, Webers CA, Schaper NC, Henry RM, van der Kallen CJ, Eussen SJ, Koster A, van Sloten TT, Köhler S, Schram MT, Houben AJ. Association of Markers of Microvascular Dysfunction With Prevalent and Incident Depressive Symptoms. Hypertension 2020; 76:342-349. [DOI: 10.1161/hypertensionaha.120.15260] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The etiology of late-life depression (LLD) is still poorly understood. Microvascular dysfunction (MVD) has been suggested to play a role in the etiology of LLD, but direct evidence of this association is scarce. The aim of this study was to investigate whether direct and indirect markers of early microvascular dysfunction are associated with prevalent and incident LLD in the population-based Maastricht Study cohort. We measured microvascular dysfunction at baseline by use of flicker light-induced retinal vessel dilation response (Dynamic Vessel Analyzer), heat-induced skin hyperemic response (laser- Doppler flowmetry), and plasma markers of endothelial dysfunction (endothelial dysfunction; sICAM-1 [soluble intercellular adhesion molecule-1], sVCAM-1 [soluble vascular adhesion molecule-1], sE-selectin [soluble E-selectin], and vWF [Von Willebrand Factor]). Depressive symptoms were assessed with the 9-item Patient Health Questionnaire (PHQ-9) at baseline and annually over 4 years of follow-up (n=3029; mean age 59.6±8.2 years, 49.5% were women, n=132 and n=251 with prevalent and incident depressive symptoms [PHQ-9≥10]). We used logistic, negative binominal and Cox regression analyses, and adjusted for demographic, cardiovascular, and lifestyle factors. Retinal venular dilatation and plasma markers of endothelial dysfunction were associated with the more prevalent depressive symptoms after full adjustment (PHQ-9 score, RR, 1.05 [1.00–1.11] and RR 1.06 [1.01–1.11], respectively). Retinal venular dilatation was also associated with prevalent depressive symptoms (PHQ-9≥10; odds ratio, 1.42 [1.09–1.84]), after full adjustment. Retinal arteriolar dilatation and plasma markers of endothelial dysfunction were associated with incident depressive symptoms (PHQ-9≥10; HR, 1.23 [1.04–1.46] and HR, 1.19 [1.05–1.35]), after full adjustment. These findings support the concept that microvascular dysfunction in the retina, and plasma markers of endothelial dysfunction is involved in the etiology of LLD and might help in finding additional targets for the prevention and treatment of LLD.
Collapse
Affiliation(s)
- Anouk F.J. Geraets
- Department of Psychiatry and Neuropsychology (A.F.J.G., S.K., M.T.S.), Maastricht University Medical Center (MUMC+), the Netherlands
- Department of Internal Medicine (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University Medical Center (MUMC+), the Netherlands
- School of Mental Health and Neuroscience (MHeNs) (A.F.J.G., T.T.J.M.B., S.K., M.T.S.), Maastricht University, the Netherlands
- School for Cardiovascular Diseases (CARIM) (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University, the Netherlands
| | - Marnix J.M. van Agtmaal
- Department of Internal Medicine (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University Medical Center (MUMC+), the Netherlands
- School for Cardiovascular Diseases (CARIM) (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University, the Netherlands
| | - Coen D.A. Stehouwer
- Department of Internal Medicine (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University Medical Center (MUMC+), the Netherlands
- School for Cardiovascular Diseases (CARIM) (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University, the Netherlands
| | - Ben M. Sörensen
- Department of Internal Medicine (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University Medical Center (MUMC+), the Netherlands
- School for Cardiovascular Diseases (CARIM) (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University, the Netherlands
| | - Tos T.J.M. Berendschot
- Department of Ophthalmology (T.T.J.M.B., C.A.B.W.), Maastricht University Medical Center (MUMC+), the Netherlands
- School of Mental Health and Neuroscience (MHeNs) (A.F.J.G., T.T.J.M.B., S.K., M.T.S.), Maastricht University, the Netherlands
- School for Cardiovascular Diseases (CARIM) (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University, the Netherlands
| | - Carroll A.B. Webers
- Department of Ophthalmology (T.T.J.M.B., C.A.B.W.), Maastricht University Medical Center (MUMC+), the Netherlands
| | - Nicolaas C. Schaper
- Department of Internal Medicine (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University Medical Center (MUMC+), the Netherlands
- School for Cardiovascular Diseases (CARIM) (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University, the Netherlands
| | - Ronald M.A. Henry
- Department of Internal Medicine (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University Medical Center (MUMC+), the Netherlands
- Heart and Vascular Center (R.M.A.H., M.T.S.), Maastricht University Medical Center (MUMC+), the Netherlands
- School for Cardiovascular Diseases (CARIM) (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University, the Netherlands
| | - Carla J.H. van der Kallen
- Department of Internal Medicine (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University Medical Center (MUMC+), the Netherlands
- School for Cardiovascular Diseases (CARIM) (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University, the Netherlands
| | - Simone J.P.M. Eussen
- Department of Epidemiology (S.J.P.M.E.), Maastricht University Medical Center (MUMC+), the Netherlands
- Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine & Life Sciences (S.J.P.M.E., A.K.), Maastricht University, the Netherlands
| | - Annemarie Koster
- Department of Social Medicine (A.K.), Maastricht University Medical Center (MUMC+), the Netherlands
- Care and Public Health Research Institute (CAPHRI), Faculty of Health, Medicine & Life Sciences (S.J.P.M.E., A.K.), Maastricht University, the Netherlands
| | - Thomas T. van Sloten
- Department of Internal Medicine (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University Medical Center (MUMC+), the Netherlands
- School for Cardiovascular Diseases (CARIM) (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University, the Netherlands
| | - Sebastian Köhler
- Department of Psychiatry and Neuropsychology (A.F.J.G., S.K., M.T.S.), Maastricht University Medical Center (MUMC+), the Netherlands
- School of Mental Health and Neuroscience (MHeNs) (A.F.J.G., T.T.J.M.B., S.K., M.T.S.), Maastricht University, the Netherlands
| | - Miranda T. Schram
- Department of Psychiatry and Neuropsychology (A.F.J.G., S.K., M.T.S.), Maastricht University Medical Center (MUMC+), the Netherlands
- Department of Internal Medicine (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University Medical Center (MUMC+), the Netherlands
- Heart and Vascular Center (R.M.A.H., M.T.S.), Maastricht University Medical Center (MUMC+), the Netherlands
- School of Mental Health and Neuroscience (MHeNs) (A.F.J.G., T.T.J.M.B., S.K., M.T.S.), Maastricht University, the Netherlands
- School for Cardiovascular Diseases (CARIM) (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University, the Netherlands
| | - Alfons J.H.M. Houben
- Department of Internal Medicine (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University Medical Center (MUMC+), the Netherlands
- School for Cardiovascular Diseases (CARIM) (A.F.J.G., M.J.M.v.A., C.D.A.S., B.M.S., N.C.S., R.M.A.H., C.J.H.v.d.K., T.T.v.S., M.T.S., A.J.H.M.H.), Maastricht University, the Netherlands
| |
Collapse
|