51
|
Guo X, Zhu Y, Li X, Lu Z, Cao Z, Yi X, Zhu X. Increased insulin resistance is associated with vascular cognitive impairment in Chinese patients with cerebral small vessel disease. Psychogeriatrics 2021; 21:342-349. [PMID: 33641231 DOI: 10.1111/psyg.12675] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/30/2021] [Accepted: 02/14/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND The aim of this study was to investigate the association between insulin resistance (IR) and vascular cognitive impairment (VCI) in patients with cerebral small vessel disease (CSVD). METHODS A total of 275 CSVD patients were enrolled in this retrospective case-control study. The homeostatic model assessment of insulin resistance (HOMA-IR) was used to measure the index of insulin resistance. Cognitive function was assessed using the Montreal Cognitive Assessment (MoCA). Spearman's correlation coefficient was used to evaluate the correlation between HOMA-IR and MoCA score. The variance inflation factor (VIF) was used to detect collinearity between variables. Multivariate logistic regression analysis was employed to confirm whether HOMA-IR is an independent risk factor for VCI in CVSD. Finally, receiver operating characteristic (ROC) curve analysis was conducted to assess the diagnostic value of HOMA-IR in VCI. RESULTS Of the 275 patients, 164 displayed VCI. VCI patients showed a significantly higher level of HOMA-IR compared to non-VCI patients (P < 0.001). HOMA-IR was negatively correlated with the MoCA score (r = -0.593, P < 0.001). After adjusting for potential confounding variables, using HOMA-IR quartile 1 (<1.11) as the reference, HOMA-IR quartile 3 (1.71-2.50) and quartile 4 (≥2.50) were independently associated with the occurrence of VCI; for each one unit increase in the HOMA-IR, the risk of VCI increased by 177.3% (odds ratio 2.773, 95% confidence interval: 1.050-7.324, P = 0.040) and 444.3% (odds ratio 5.443, 95% confidence interval: 2.109-14.050, P < 0.001), respectively. According to the ROC curve, the optimal cut-off point of HOMA-IR in predicting VCI was 1.55, and the area under the curve was 0.744, with a sensitivity of 71.3% and a specificity of 69.4%. CONCLUSION This study demonstrated that increased IR is significantly associated with VCI in CSVD patients.
Collapse
Affiliation(s)
- Xiaoming Guo
- Department of Neurology, Second Affiliated Hospital of Nantong University, Nantong, China
| | - Yuting Zhu
- Department of Neurology, Second Affiliated Hospital of Nantong University, Nantong, China
| | - Xinling Li
- Department of Neurology, Second Affiliated Hospital of Nantong University, Nantong, China
| | - Zhenhui Lu
- Department of Neurology, Second Affiliated Hospital of Nantong University, Nantong, China
| | - Zhiyong Cao
- Department of Neurology, Second Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaoyi Yi
- Department of Neurology, Second Affiliated Hospital of Nantong University, Nantong, China
| | - Xiangyang Zhu
- Department of Neurology, Second Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
52
|
Diomedi M, Rocco A, Bonomi CG, Mascolo AP, De Lucia V, Marrama F, Sallustio F, Koch G, Martorana A. Haemodynamic impairment along the Alzheimer's disease continuum. Eur J Neurol 2021; 28:2168-2173. [PMID: 33759296 DOI: 10.1111/ene.14834] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND PURPOSE Alzheimer's disease (AD) is considered a clinical and biological continuum identified via cerebrospinal fluid (CSF) or imaging biomarkers. Chronic hypoperfusion is held as one of the main features of Alzheimer's disease, as part of the processes causing neuronal degeneration. The mechanism responsible for such condition is still debated, although recently a direct connection with amyloid peptides has been shown. Here the aim was to investigate whether measures of hypoperfusion change along the AD continuum. METHODS Seventy patients with mild AD were recruited and stratified according to their CSF biomarker profile-as indicated by the National Institute on Aging and Alzheimer's Association research framework-into patients with either isolated amyloid pathology (A+T-) or full-blown AD (A+T+), and further layered according to apolipoprotein E genotype. After evaluation of vascular risk factors, a transcranial Doppler was performed on each patient, to evaluate mean flow velocity and pulsatility index in the middle cerebral artery, and to calculate the breath-holding index. Patients were compared to a cohort of 17 healthy controls. RESULTS The breath-holding index was reduced in the AD continuum and was inversely correlated to CSF amyloid β42 levels. Such correlation was stronger in the A+T+ than in the A+T- group, and unexpectedly reached statistical significance only in the E3 and not in the E4 genotype carriers. CONCLUSIONS These results suggest a tight and effective relationship between amyloid β42, vascular hypoperfusion, cerebrovascular reactivity and epsilon genotype.
Collapse
Affiliation(s)
- Marina Diomedi
- Stroke Center, Policlinico Tor Vergata, Rome University 'Tor Vergata', Rome, Italy
| | - Alessandro Rocco
- Stroke Center, Policlinico Tor Vergata, Rome University 'Tor Vergata', Rome, Italy
| | | | | | - Vincenzo De Lucia
- Memory Clinic, Policlinico Tor Vergata, Rome University 'Tor Vergata', Rome, Italy
| | - Federico Marrama
- Stroke Center, Policlinico Tor Vergata, Rome University 'Tor Vergata', Rome, Italy
| | - Fabrizio Sallustio
- Stroke Center, Policlinico Tor Vergata, Rome University 'Tor Vergata', Rome, Italy
| | - Giacomo Koch
- Non Invasive Brain Stimulation Unit, IRCCS Santa Lucia, Rome, Italy.,Dipartimento di Neuroscienze e Riabilitazione, Sezione di Fisiologia Umana, Università di Ferrara, Ferrara, Italy
| | - Alessandro Martorana
- Memory Clinic, Policlinico Tor Vergata, Rome University 'Tor Vergata', Rome, Italy
| |
Collapse
|
53
|
Kim MS, Bang J, Kim BY, Jeon WK. Impaired Cognitive Flexibility Induced by Chronic Cerebral Hypoperfusion in the 5XFAD Transgenic Mouse Model of Mixed Dementia. J Gerontol A Biol Sci Med Sci 2021; 76:1169-1178. [PMID: 33709149 PMCID: PMC8202140 DOI: 10.1093/gerona/glab075] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Indexed: 12/27/2022] Open
Abstract
Cerebrovascular lesions are widely prevalent in patients with Alzheimer’s disease (AD), but their relationship to the pathophysiology of AD remains poorly understood. An improved understanding of the interaction of cerebrovascular damage with AD is crucial for the development of therapeutic approaches. Herein, we investigated the effects of chronic cerebral hypoperfusion (CCH) in a 5XFAD transgenic (Tg) mouse model of AD. We established CCH conditions in both Tg and non-Tg mice by inducing unilateral common carotid artery occlusion (UCCAO). Cognitive performance in mice was evaluated, and their brain tissue was examined for amyloid-beta (Aβ) pathology to elucidate possible mechanisms. We found that UCCAO-operated Tg mice showed impaired cognitive flexibility in the reversal phase of the hidden-platform water maze task compared to sham-operated Tg mice. Interestingly, UCCAO-operated Tg mice used fewer spatial cognitive strategies than sham-operated Tg mice during reversal learning. These cognitive deficits were accompanied by increased Aβ plaque burden and Aβ42 levels in the hippocampus and prefrontal cortex, 2 regions that play essential roles in the regulation of cognitive flexibility. Furthermore, changes in cognitive flexibility are strongly correlated with the expression levels of enzymes related to Aβ clearance, such as neprilysin and insulin-degrading enzymes. These findings suggest that, in 5XFAD mice, impaired cognitive flexibility is related to CCH, and that Aβ clearance might be involved in this process.
Collapse
Affiliation(s)
- Min-Soo Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea.,Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul, South Korea
| | - Jihye Bang
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea.,Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul, South Korea
| | - Bu-Yeo Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Won Kyung Jeon
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea.,Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul, South Korea
| |
Collapse
|
54
|
Abstract
PURPOSE OF REVIEW The relationship between hypertension and Alzheimer's disease (AD) is complex and varies across the lifespan. Studies have suggested that midlife hypertension is a risk factor for AD, although studies of late life hypertension have suggested that it either has no effect or a weak protective effect. RECENT FINDINGS Animal models of induced and spontaneous hypertension have found that AD pathological change (β-amyloid plaques and tau tangles) occurs within weeks of a hypertensive insult. Human imaging and autopsy studies indicate that midlife and late life hypertension are associated with increased AD pathological change. Meta-analyses of longitudinal studies indicate that midlife rather than late life hypertension is a risk factor for AD. New areas of research have suggested that rather than mean blood pressure (BP), it is the negative BP trajectories or the variability of BP that contributes to AD. In a number of meta-analyses of antihypertensive medications and their effect on AD, there were weak associations between improved AD outcomes and treatment. SUMMARY The combined analysis of animal, human clinical/pathological, epidemiological and drug trial data indicates that hypertension increases the risk of AD and treatment of hypertension may be an appropriate preventive measure.
Collapse
|
55
|
Gottesman RF, Mosley TH, Knopman DS, Hao Q, Wong D, Wagenknecht LE, Hughes TM, Qiao Y, Dearborn J, Wasserman BA. Association of Intracranial Atherosclerotic Disease With Brain β-Amyloid Deposition: Secondary Analysis of the ARIC Study. JAMA Neurol 2021; 77:350-357. [PMID: 31860001 DOI: 10.1001/jamaneurol.2019.4339] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Importance Intracranial atherosclerotic disease (ICAD) is an important cause of stroke and has also been recently identified as an important risk factor for all-cause dementia, but the mechanism of its association with cognitive performance is not fully understood. Objective To test the hypothesis that ICAD is associated with cerebral β-amyloid deposition as a marker of Alzheimer disease. Design, Setting, and Participants This cross-sectional analysis of data collected from August 2011 through November 2014 was a community-based cohort study conducted in 3 US communities. Of 346 adults without dementia aged 70 to 90 years who were sequentially recruited from 3 of 4 sites of the larger Atherosclerosis Risk in Communities study into a study of brain florbetapir positron emission tomography (ARIC-PET), 300 met inclusion criteria. A total of 589 were approached about recruitment, of whom 346 (58.7%) consented (the remainder either met exclusion criteria for ARIC-PET or refused to participate). Data were analyzed from July 2017 through October 2019. Exposures Intracranial atherosclerotic disease presence, frequency, and extent of stenosis, by high-resolution vessel wall magnetic resonance imaging. Main Outcomes and Measures Global cortical standardized uptake value ratio (SUVR) of greater than 1.2 as measured by florbetapir PET. Models were conducted using logistic regression methods. In secondary analyses, we tested effect modifications by apolipoprotein E ε4 genotype with interaction terms and in stratified models and evaluated regional patterns of associations. Results In 300 participants (mean [SD] age, 76 [5] years; 132 African American individuals [44%], 167 women [56%], and 94 carriers of at least 1 apolipoprotein E ε4 allele [31%]), ICAD was found in 105 participants (35%) and mean (SD) SUVR was higher in individuals with vs without intracranial plaques (1.34 [0.29] vs 1.27 [0.23]; P = .03). In adjusted models, ICAD presence (plaque presence [adjusted odds ratio (aOR), 1.20; 95% CI, 0.69-2.07] and frequency [aOR, 1.10; 95% CI, 0.96-1.26]) was not associated significantly with elevated SUVR in the total sample. Furthermore, modest stenosis of the intracranial vessels (defined as >50% stenosis) was not associated with elevated SUVR (aOR, 2.33; 95% CI, 0.82-6.60). Conclusions and Relevance In this community-based cohort of adults without dementia, intracranial atherosclerotic plaque or stenosis was not associated with brain β-amyloid deposition.
Collapse
Affiliation(s)
- Rebecca F Gottesman
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland.,Department of Epidemiology, Johns Hopkins University, Baltimore, Maryland
| | - Thomas H Mosley
- Department of Medicine, University of Mississippi Medical Center, Jackson
| | | | - Qing Hao
- Department of Neurology, Mount Sinai Medical Center, New York, New York
| | - Dean Wong
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Lynne E Wagenknecht
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Timothy M Hughes
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ye Qiao
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Jennifer Dearborn
- Department of Neurology, Beth Israel Deaconness Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Bruce A Wasserman
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
56
|
Wang R, Qiu C, Dintica CS, Shang Y, Calderón Larrañaga A, Wang HX, Xu W. Shared risk and protective factors between Alzheimer's disease and ischemic stroke: A population-based longitudinal study. Alzheimers Dement 2021; 17:191-204. [PMID: 33527694 DOI: 10.1002/alz.12203] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 08/31/2020] [Accepted: 09/06/2020] [Indexed: 11/06/2022]
Abstract
INTRODUCTION Stroke, especially ischemic stroke's (IS) link with Alzheimer's disease (AD) remains unclear. METHODS This prospective cohort study included 2459 AD- and cerebrovascular disease-free older adults at baseline (mean age 71.9 ± 10.3 years, Stockholm, Sweden). Using Cox regressions, shared risk factors (SRFs) and shared protective factors (SPFs) between AD and IS were recognized when their hazard ratios in both AD and IS models were significant and in the same direction. RESULTS During the follow-up period of up to 15 years, 132 AD and 260 IS mutually exclusive cases were identified. SRFs were low education, sedentary lifestyle, and heart diseases. High levels of psychological well-being, actively engaging in leisure activities, and a rich social network were SPFs. Having ≥1 SPF reduced 47% of AD and 28% of IS risk among people with a low risk profile (<2 SRFs), and 38% of AD and 31% of IS risk with a high risk profile (≥2 SRFs). In total, 57.8% of AD/IS cases could be prevented if individuals have ≥1 SPF and no SRF. DISCUSSION AD and IS share risk/protective profiles, and SPFs seem to counteract the adverse effects of SRFs on both AD and IS.
Collapse
Affiliation(s)
- Rui Wang
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden.,The Swedish School of Sport and Health Sciences, GIH, Stockholm, Sweden.,Department of Medicine and Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Chengxuan Qiu
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Christina S Dintica
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Ying Shang
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Amaia Calderón Larrañaga
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Hui-Xin Wang
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden.,Stress Research Institute, Stockholm University, Stockholm, Sweden
| | - Weili Xu
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden.,Department of Epidemiology & Biostatistics, Tianjin Medical University, Tianjin, P. R. China
| |
Collapse
|
57
|
Cortes-Canteli M, Gispert JD, Salvadó G, Toribio-Fernandez R, Tristão-Pereira C, Falcon C, Oliva B, Mendiguren J, Fernandez-Friera L, Sanz J, Garcia-Ruiz JM, Fernandez-Ortiz A, Sanchez-Gonzalez J, Ibanez B, Molinuevo JL, Fuster V. Subclinical Atherosclerosis and Brain Metabolism in Middle-Aged Individuals: The PESA Study. J Am Coll Cardiol 2021; 77:888-898. [PMID: 33602472 DOI: 10.1016/j.jacc.2020.12.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND Atherosclerosis has been linked to cognitive decline in late life; however, the impact of cardiovascular risk factors (CVRFs) and subclinical atherosclerosis on brain metabolism at earlier stages remains unexplored. OBJECTIVES This study sought to determine the association between brain metabolism, subclinical atherosclerosis, and CVRFs in middle-aged asymptomatic individuals. METHODS This study included 547 asymptomatic middle-aged participants (50 ± 4 years, 82% men) from the PESA (Progression of Early Subclinical Atherosclerosis) study with evidence of subclinical atherosclerosis. Participants underwent 18F-fluorodeoxyglucose (FDG)-positron emission tomography. Global brain FDG uptake and voxel-wise analyses were used to evaluate the associations of cerebral metabolism with CVRFs and atherosclerotic plaque burden in carotids and femorals assessed by 3-dimensional vascular ultrasound. RESULTS Global FDG uptake showed an inverse correlation with 30-year Framingham Risk Score (FRS) (β = -0.15, p < 0.001). This association was mainly driven by the presence of hypertension (d = 0.36, p < 0.001). Carotid plaque burden was inversely associated with global brain FDG uptake (β = -0.16, p < 0.001), even after adjusting for 30-year FRS. Voxel-wise approaches revealed that the brain areas most strongly affected by hypometabolism in association with 30-year FRS, hypertension, and carotid plaque burden were parietotemporal regions (angular, supramarginal, and inferior/middle temporal gyri) and the cingulate gyrus. CONCLUSIONS In asymptomatic middle-aged individuals, cardiovascular risk is associated with brain hypometabolism, with hypertension being the modifiable CVRF showing the strongest association. Subclinical carotid plaque burden is also linked to reduced brain metabolism independently of CVRFs. Cerebral areas showing hypometabolism include those known to be affected in dementia. These data reinforce the need to control CVRFs early in life in order to potentially reduce the brain's midlife vulnerability to future cognitive dysfunction.
Collapse
Affiliation(s)
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain; Universitat Pompeu Fabra, Barcelona, Spain
| | - Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | | | | | - Carles Falcon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
| | - Belen Oliva
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Leticia Fernandez-Friera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; HM Hospitales-Centro Integral de Enfermedades Cardiovasculares, Universidad San Pablo-CEU, Madrid, Spain; CIBER de enfermedades Cardiovasculares, Madrid, Spain
| | - Javier Sanz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jose M Garcia-Ruiz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de enfermedades Cardiovasculares, Madrid, Spain; Hospital Universitario Central de Oviedo, Asturias, Spain
| | - Antonio Fernandez-Ortiz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de enfermedades Cardiovasculares, Madrid, Spain; Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos, Universidad Complutense, Madrid, Spain
| | | | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de enfermedades Cardiovasculares, Madrid, Spain; Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable, Madrid, Spain.
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
58
|
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: 44] [Impact Index Per Article: 11.0] [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
|
59
|
D'Souza CE, Greenway MRF, Graff-Radford J, Meschia JF. Cognitive Impairment in Patients with Stroke. Semin Neurol 2021; 41:75-84. [PMID: 33418591 DOI: 10.1055/s-0040-1722217] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Despite substantial advances in stroke care, vascular cognitive impairment remains a prominent source of disability. Unlike sensorimotor impairments, cognition often continues to decline after stroke. An aging population will increase the prevalence of vascular cognitive impairment, with stroke playing an important role. Ten percent of patients presenting with stroke have pre-stroke dementia; an additional 10% will develop incident dementia with a first stroke, and 30% with a recurrent stroke. While stroke increases the risk of cognitive impairment, the presence of cognitive impairment also impacts acute stroke treatment and increases risk of poor outcome by nearly twofold. There is substantial overlap in the clinical and pathological aspects of vascular and degenerative dementias in many patients. How they relate to one another is controversial. The treatment of vascular cognitive impairment remains supportive, focusing on treating vascular risk factors. Cognitive rehabilitation after stroke is an area of active research, and existing pharmacologic treatments have limited benefit. Heightened awareness of cognitive impairment in the setting of stroke is imperative for prognostication and management, impetus for research and, ultimately, the discovery of efficacious treatments.
Collapse
Affiliation(s)
- Caitlin E D'Souza
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida.,Department of Neurology, Baptist Health, Jacksonville, Florida
| | | | | | - James F Meschia
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida
| |
Collapse
|
60
|
Mishra N, Mohan D, Fuad S, Basavanagowda DM, Alrashid ZA, Kaur A, Rathod B, Nosher S, Heindl SE. The Association Between Hypertension and Cognitive Impairment, and the Role of Antihypertensive Medications: A Literature Review. Cureus 2020; 12:e12035. [PMID: 33457135 PMCID: PMC7797448 DOI: 10.7759/cureus.12035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Vascular dementia (VD) is one of the leading causes of dementia, and hypertension is a known risk factor for VD. Hypertension treatment guidelines have previously discussed an optimal blood pressure goal to prevent further cardiovascular complications with long-term management. The treatment of hypertension can prevent stroke, kidney failure, and perhaps prevent cognitive decline as well. We reviewed studies that demonstrated an association between hypertension and cognitive impairment (CI). The role of antihypertensive medications (AHM) in preventing CI was also investigated. This topic is worth exploring as dementia has high healthcare costs and will become prominent as the population in the United States ages. We used the medical subject heading (MeSH) search strategy on Pubmed and reviewed 22 articles. The studies showed that there might be a link between hypertension, AHM, and CI. The studies did not suggest a superiority of any specific AHM class to prevent CI. Further research on optimal hypertension treatment goals to prevent cognitive impairment and dementia is recommended.
Collapse
Affiliation(s)
- Nupur Mishra
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Devyani Mohan
- Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sehrish Fuad
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Deepak M Basavanagowda
- Psychiatry and Behavioral Sciences, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Zaid A Alrashid
- Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Arveen Kaur
- Psychiatry and Behavioral Sciences, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Bindu Rathod
- Psychiatry and Behavioral Sciences, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sadia Nosher
- Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Stacey E Heindl
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Medicine, Avalon University School of Medicine, Willemstad, CUW
| |
Collapse
|
61
|
Alzheimer's Disease and Vascular Aging: JACC Focus Seminar. J Am Coll Cardiol 2020; 75:942-951. [PMID: 32130930 PMCID: PMC8046164 DOI: 10.1016/j.jacc.2019.10.062] [Citation(s) in RCA: 237] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/17/2019] [Accepted: 10/27/2019] [Indexed: 01/23/2023]
Abstract
Alzheimer’s disease, the leading cause of dementia in the elderly, is a neurodegenerative condition characterized by accumulation of amyloid plaques and neurofibrillary tangles in the brain. However, age-related vascular changes accompany or even precede the development of Alzheimer’s pathology, raising the possibility that they may have a pathogenic role. This review provides an appraisal of the alterations in cerebral and systemic vasculature, the heart, and hemostasis that occur in Alzheimer’s disease and their relationships to cognitive impairment. Although the molecular pathogenesis of these alterations remains to be defined, amyloid-β is a likely contributor in the brain as in the heart. Collectively, the evidence suggests that vascular pathology is a likely pathogenic contributor to age-related dementia, including Alzheimer’s disease, inextricably linked to disease onset and progression. Consequently, the contribution of vascular factors should be considered in preventive, diagnostic, and therapeutic approaches to address one of the major health challenges of our time.
Collapse
|
62
|
Yin Z, Wang X, Zheng S, Cao P, Chen Y, Yu M, Liao C, Zhang Z, Han J, Duan Y, Yang X, Zhang S. LongShengZhi Capsule Attenuates Alzheimer-Like Pathology in APP/PS1 Double Transgenic Mice by Reducing Neuronal Oxidative Stress and Inflammation. Front Aging Neurosci 2020; 12:582455. [PMID: 33328962 PMCID: PMC7719723 DOI: 10.3389/fnagi.2020.582455] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 10/16/2020] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia in the elderly. It may be caused by oxidative stress, inflammation, and cerebrovascular dysfunctions in the brain. LongShengZhi Capsule (LSZ), a traditional Chinese medicine, has been approved by the China Food and Drug Administration for treatment of patients with cardiovascular/cerebrovascular disease. LSZ contains several neuroprotective ingredients, including Hirudo, Astmgali Radix, Carthami Flos (Honghua), Persicae Semen (Taoren), Acori Tatarinowii Rhizoma (Shichangpu), and Acanthopanax Senticosus (Ciwujia). In this study, we aimed to determine the effect of LSZ on the AD process. Double transgenic mice expressing the amyloid-β precursor protein and mutant human presenilin 1 (APP/PS1) to model AD were treated with LSZ for 7 months starting at 2 months of age. LSZ significantly improved the cognition of the mice without adverse effects, indicating its high degree of safety and efficacy after a long-term treatment. LSZ reduced AD biomarker Aβ plaque accumulation by inhibiting β-secretase and γ-secretase gene expression. LSZ also reduced p-Tau expression, cell death, and inflammation in the brain. Consistently, in vitro, LSZ ethanol extract enhanced neuronal viability by reducing L-glutamic acid-induced oxidative stress and inflammation in HT-22 cells. LSZ exerted antioxidative effects by enhancing superoxide dismutase and glutathione peroxidase expression, reduced Aβ accumulation by inhibiting β-secretase and γ-secretase mRNA expression, and decreased p-Tau level by inhibiting NF-κB-mediated inflammation. It also demonstrated neuroprotective effects by regulating the Fas cell surface death receptor/B-cell lymphoma 2/p53 pathway. Taken together, our study demonstrates the antioxidative stress, anti-inflammatory, and neuroprotective effects of LSZ in the AD-like pathological process and suggests it could be a potential medicine for AD treatment.
Collapse
Affiliation(s)
- Zequn Yin
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xuerui Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Shihong Zheng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Peichang Cao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Maoyun Yu
- School of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China
| | - Chenzhong Liao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | | | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.,College of Life Sciences, Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| |
Collapse
|
63
|
Toppala S, Ekblad LL, Lötjönen J, Helin S, Hurme S, Johansson J, Jula A, Karrasch M, Koikkalainen J, Laine H, Parkkola R, Viitanen M, Rinne JO. Midlife Insulin Resistance as a Predictor for Late-Life Cognitive Function and Cerebrovascular Lesions. J Alzheimers Dis 2020; 72:215-228. [PMID: 31561373 PMCID: PMC6839606 DOI: 10.3233/jad-190691] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background: Type 2 diabetes (T2DM) increases the risk for Alzheimer’s disease (AD) but not for AD neuropathology. The association between T2DM and AD is assumed to be mediated through vascular mechanisms. However, insulin resistance (IR), the hallmark of T2DM, has been shown to associate with AD neuropathology and cognitive decline. Objective: To evaluate if midlife IR predicts late-life cognitive performance and cerebrovascular lesions (white matter hyperintensities and total vascular burden), and whether cerebrovascular lesions and brain amyloid load are associated with cognitive functioning. Methods: This exposure-to-control follow-up study examined 60 volunteers without dementia (mean age 70.9 years) with neurocognitive testing, brain 3T-MRI and amyloid-PET imaging. The volunteers were recruited from the Finnish Health 2000 survey (n = 6062) to attend follow-up examinations in 2014–2016 according to their insulin sensitivity in 2000 and their APOE genotype. The exposure group (n = 30) had IR in 2000 and the 30 controls had normal insulin sensitivity. There were 15 APOEɛ4 carriers per group. Statistical analyses were performed with multivariable linear models. Results: At follow-up the IR+group performed worse on executive functions (p = 0.02) and processing speed (p = 0.007) than the IR- group. The groups did not differ in cerebrovascular lesions. No associations were found between cerebrovascular lesions and neurocognitive test scores. Brain amyloid deposition associated with slower processing speed. Conclusion: Midlife IR predicted poorer executive functions and slower processing speed, but not cerebrovascular lesions. Brain amyloid deposition was associated with slower processing speed. The association between midlife IR and late-life cognition might not be mediated through cerebrovascular lesions measured here.
Collapse
Affiliation(s)
- Sini Toppala
- Turku PET Centre, University of Turku, Finland.,Turku City Hospital, University of Turku, Finland
| | | | | | - Semi Helin
- Turku PET Centre, University of Turku, Finland
| | - Saija Hurme
- Department of Biostatistics, University of Turku, Finland
| | - Jarkko Johansson
- Turku PET Centre, University of Turku, Finland.,Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Antti Jula
- National Institute for Health and Welfare, Turku, Finland
| | - Mira Karrasch
- Department of Psychology, Åbo Akademi University, Turku, Finland
| | | | - Hanna Laine
- Turku City Hospital, University of Turku, Finland.,Department of Medicine, University of Turku, Turku University Hospital, Turku, Finland
| | - Riitta Parkkola
- Department of Radiology, Turku University and Turku University Hospital, Turku, Finland
| | - Matti Viitanen
- Turku City Hospital, University of Turku, Finland.,Clinical Geriatrics, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Juha O Rinne
- Turku PET Centre, University of Turku, Finland.,Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland
| |
Collapse
|
64
|
Shafi O. Switching of vascular cells towards atherogenesis, and other factors contributing to atherosclerosis: a systematic review. Thromb J 2020; 18:28. [PMID: 33132762 PMCID: PMC7592591 DOI: 10.1186/s12959-020-00240-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022] Open
Abstract
Background Onset, development and progression of atherosclerosis are complex multistep processes. Many aspects of atherogenesis are not yet properly known. This study investigates the changes in vasculature that contribute to switching of vascular cells towards atherogenesis, focusing mainly on ageing. Methods Databases including PubMed, MEDLINE and Google Scholar were searched for published articles without any date restrictions, involving atherogenesis, vascular homeostasis, aging, gene expression, signaling pathways, angiogenesis, vascular development, vascular cell differentiation and maintenance, vascular stem cells, endothelial and vascular smooth muscle cells. Results Atherogenesis is a complex multistep process that unfolds in a sequence. It is caused by alterations in: epigenetics and genetics, signaling pathways, cell circuitry, genome stability, heterotypic interactions between multiple cell types and pathologic alterations in vascular microenvironment. Such alterations involve pathological changes in: Shh, Wnt, NOTCH signaling pathways, TGF beta, VEGF, FGF, IGF 1, HGF, AKT/PI3K/ mTOR pathways, EGF, FOXO, CREB, PTEN, several apoptotic pathways, ET - 1, NF-κB, TNF alpha, angiopoietin, EGFR, Bcl - 2, NGF, BDNF, neurotrophins, growth factors, several signaling proteins, MAPK, IFN, TFs, NOs, serum cholesterol, LDL, ephrin, its receptor pathway, HoxA5, Klf3, Klf4, BMPs, TGFs and others.This disruption in vascular homeostasis at cellular, genetic and epigenetic level is involved in switching of the vascular cells towards atherogenesis. All these factors working in pathologic manner, contribute to the development and progression of atherosclerosis. Conclusion The development of atherosclerosis involves the switching of gene expression towards pro-atherogenic genes. This happens because of pathologic alterations in vascular homeostasis. When pathologic alterations in epigenetics, genetics, regulatory genes, microenvironment and vascular cell biology accumulate beyond a specific threshold, then the disease begins to express itself phenotypically. The process of biological ageing is one of the most significant factors in this aspect as it is also involved in the decline in homeostasis, maintenance and integrity.The process of atherogenesis unfolds sequentially (step by step) in an interconnected loop of pathologic changes in vascular biology. Such changes are involved in 'switching' of vascular cells towards atherosclerosis.
Collapse
Affiliation(s)
- Ovais Shafi
- Sindh Medical College - Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
65
|
Eglit GML, Weigand AJ, Nation DA, Bondi MW, Bangen KJ. Hypertension and Alzheimer's disease: indirect effects through circle of Willis atherosclerosis. Brain Commun 2020; 2:fcaa114. [PMID: 33543127 PMCID: PMC7846096 DOI: 10.1093/braincomms/fcaa114] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 06/24/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023] Open
Abstract
Hypertension is common among older adults and is believed to increase susceptibility to Alzheimer's disease, but mechanisms underlying this relationship are unclear. Hypertension also promotes circle of Willis atherosclerosis, which contributes to cerebral hypoperfusion and arterial wall stiffening, two potential mechanisms linking hypertension to Alzheimer's disease. To examine the role of circle of Willis atherosclerosis in the association between hypertension and Alzheimer's disease neuropathology, we analysed post-mortem neuropathological data on 2198 decedents from the National Alzheimer's Coordinating Center database [mean (standard deviation) age at last visit 80.51 (1.95) and 47.1% female] using joint simultaneous (i.e. mediation) modelling. Within the overall sample and among Alzheimer's dementia decedents, hypertension was indirectly associated with increased neuritic plaques and neurofibrillary tangles through its association with circle of Willis atherosclerosis. Similar indirect effects were observed for continuous measures of systolic and diastolic blood pressure. These results suggest that hypertension may promote Alzheimer's disease pathology indirectly through intracranial atherosclerosis by limiting cerebral blood flow and/or dampening perivascular clearance. Circle of Willis atherosclerosis may be an important point of convergence between vascular risk factors, cerebrovascular changes and Alzheimer's disease neuropathology.
Collapse
Affiliation(s)
- Graham M L Eglit
- Veteran Affairs San Diego Healthcare System, San Diego, CA 92161, USA
| | - Alexandra J Weigand
- San Diego State University/University of California, San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA 92120, USA
| | - Daniel A Nation
- Department of Psychological Science, University of California, Irvine, Irvine, CA 92697, USA
- Institute for Memory Disorders and Neurological Impairments, University of California, Irvine, Irvine, CA 92697, USA
| | - Mark W Bondi
- Veteran Affairs San Diego Healthcare System, San Diego, CA 92161, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Katherine J Bangen
- Veteran Affairs San Diego Healthcare System, San Diego, CA 92161, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
66
|
Rehiman SH, Lim SM, Neoh CF, Majeed ABA, Chin AV, Tan MP, Kamaruzzaman SB, Ramasamy K. Proteomics as a reliable approach for discovery of blood-based Alzheimer's disease biomarkers: A systematic review and meta-analysis. Ageing Res Rev 2020; 60:101066. [PMID: 32294542 DOI: 10.1016/j.arr.2020.101066] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 02/08/2023]
Abstract
In order to gauge the impact of proteomics in discovery of Alzheimer's disease (AD) blood-based biomarkers, this study had systematically reviewed articles published between 1984-2019. Articles that fulfilled the inclusion criteria were assessed for risk of bias. A meta-analysis was performed for replicable candidate biomarkers (CB). Of the 1651 articles that were identified, 17 case-control and two cohort studies, as well as three combined case-control and longitudinal designs were shortlisted. A total of 207 AD and mild cognitive impairment (MCI) CB were discovered, with 48 reported in >2 studies. This review highlights six CB, namely alpha-2-macroglobulin (α2M)ps, pancreatic polypeptide (PP)ps, apolipoprotein A-1 (ApoA-1)ps, afaminp, insulin growth factor binding protein-2 (IGFBP-2)ps and fibrinogen-γ-chainp, all of which exhibited consistent pattern of regulation in >three independent cohorts. They are involved in AD pathogenesis via amyloid-beta (Aβ), neurofibrillary tangles, diabetes and cardiovascular diseases (CVD). Meta-analysis indicated that ApoA-1ps was significantly downregulated in AD (SMD = -1.52, 95% CI: -1.89, -1.16, p < 0.00001), with low inter-study heterogeneity (I2 = 0%, p = 0.59). α2Mps was significantly upregulated in AD (SMD = 0.83, 95% CI: 0.05, 1.62, p = 0.04), with moderate inter-study heterogeneity (I2 = 41%, p = 0.19). Both CB are involved in Aβ formation. These findings provide important insights into blood-based AD biomarkers discovery via proteomics.
Collapse
|
67
|
Gao F, Zhang J, Ni T, Lin N, Lin H, Luo H, Guo H, Chi J. Herpud1 deficiency could reduce amyloid-β40 expression and thereby suppress homocysteine-induced atherosclerosis by blocking the JNK/AP1 pathway. J Physiol Biochem 2020; 76:383-391. [PMID: 32488540 DOI: 10.1007/s13105-020-00741-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 04/21/2020] [Indexed: 12/23/2022]
Abstract
Homocysteine (Hcy) is considered an independent risk factor for various cardiovascular diseases including atherosclerosis which is associated with lipid metabolism, inflammation, and oxidative stress. Results from our previous study suggested that Hcy-induced atherosclerosis could be reversed by Herpud1 knockout which inhibits vascular smooth muscle cell (VSMC) phenotype switching. Here, we aim to investigate more precise mechanisms behind the improvement in Hcy-induced atherosclerosis. Amyloid-β40 (Aβ40), a vital protein in Alzheimer disease (AD), has been regarded as an important component in the atherosclerosis program in recent years due to the biological similarity between AD and atherosclerosis. Thus, we determined to assess the value of Aβ40 in a Herpud1 knockout Hcy-induced atherosclerosis mouse model by measuring Aβ40 expression in tissue and biomarkers of lipid metabolism, inflammation, and oxidative stress in serum. Additionally, since endothelial dysfunction plays a prominent role in atherosclerosis, we tested human umbilical vein endothelial cell (HUVEC) function following Herpud1 silencing in vitro and evaluated JNK/AP1 signaling activation in our models because of its close relationship with Aβ40. As a result, our animal models showed that Herpud1 knockout reduced Aβ40 expression, inflammation, and oxidative stress levels other than lipid metabolism and alleviated atherosclerosis via JNK/AP1 signaling inhibition. Similarly, our cell experiments implied that Hcy-induced Aβ40 elevation and HUVEC dysfunction involving cell proliferation and apoptosis could be restored by Herpud1 silence through restraining JNK/AP1 pathway. Collectively, our study demonstrates that Herpud1 deficiency could reduce Aβ40 expression, thereby suppressing Hcy-induced atherosclerosis by blocking the JNK/AP1 pathway. This may provide novel potential targets for atherosclerosis prevention or treatment.
Collapse
Affiliation(s)
- Feidan Gao
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang, 312000, China
| | - Jie Zhang
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang, 312000, China
| | - Tingjuan Ni
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang, 312000, China
| | - Na Lin
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang, 312000, China
| | - Hui Lin
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang, 312000, China
| | - Hangqi Luo
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang, 312000, China
| | - Hangyuan Guo
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang, 312000, China
| | - Jufang Chi
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang, 312000, China.
| |
Collapse
|
68
|
Yu X, Ji C, Shao A. Neurovascular Unit Dysfunction and Neurodegenerative Disorders. Front Neurosci 2020; 14:334. [PMID: 32410936 PMCID: PMC7201055 DOI: 10.3389/fnins.2020.00334] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/20/2020] [Indexed: 12/11/2022] Open
Abstract
The neurovascular unit (NVU), composed of vascular cells, glial cells, and neurons, is the minimal functional unit of the brain. The NVU maintains integrity of the blood–brain barrier (BBB) and regulates supply of the cerebral blood flow (CBF), both of which are keys to maintaining normal brain function. BBB dysfunction and a decreased CBF are early pathophysiological changes in neurodegenerative disorders, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS). In this review, we primarily focus on the NVU in AD as much research has been performed on the connection between NVU dysfunction and AD. We also discuss the role of NVU dysfunction in the pathophysiological mechanisms of PD and ALS. As most neurodegenerative diseases are difficult to treat, we discuss several potential drug targets that focus on the NVU that may inform novel vascular-targeted therapies for AD, PD, and ALS.
Collapse
Affiliation(s)
- Xing Yu
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Caihong Ji
- Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
69
|
Min LJ, Iwanami J, Shudou M, Bai HY, Shan BS, Higaki A, Mogi M, Horiuchi M. Deterioration of cognitive function after transient cerebral ischemia with amyloid-β infusion-possible amelioration of cognitive function by AT 2 receptor activation. J Neuroinflammation 2020; 17:106. [PMID: 32264971 PMCID: PMC7140348 DOI: 10.1186/s12974-020-01775-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 03/17/2020] [Indexed: 12/24/2022] Open
Abstract
Background To promote understanding of the pathogenesis of cognitive impairment or dementia, we explored the potential interaction between transient cerebral ischemia and amyloid-β (Aβ) infusion in mediating cognitive decline and examined the possible ameliorative effect of angiotensin II type 2 (AT2) receptor activation in vascular smooth muscle cells (VSMC) on this cognitive deficit. Methods Adult male wild-type mice (WT) and mice with VSMC-specific AT2 receptor overexpression (smAT2) were subjected to intracerebroventricular (ICV) injection of Aβ1-40. Transient cerebral ischemia was induced by 15 min of bilateral common carotid artery occlusion (BCCAO) 24 h after Aβ injection. Results Aβ injection in WT induced a cognitive decline, whereas BCCAO did not cause a significant cognitive deficit. In contrast, WT with BCCAO following Aβ injection exhibited more marked cognitive decline compared to Aβ injection alone, in concert with increases in superoxide anion production, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity, and expression of p22phox, p40phox, monocyte chemoattractant protein (MCP)-1 and interleukin (IL)-1β in the hippocampus, and upregulation of RAGE (receptor for advanced glycation end product), an Aβ transporter. BCCAO following Aβ injection further enhanced neuronal pyknosis in the hippocampus, compared with BCCAO or Aβ injection alone. In contrast, smAT2 did not show a cognitive decline, increase in oxidative stress, inflammation, and RAGE level or neuronal pyknosis, which were induced by BCCAO with/without Aβ injection in WT. Conclusions Transient cerebral ischemia might worsen Aβ infusion-mediated cognitive decline and vice versa, with possible involvement of amplified oxidative stress and inflammation and impairment of the RAGE-mediated Aβ clearance system, contributing to exaggerated neuronal degeneration. AT2 receptor activation in VSMC could play an inhibitory role in this cognitive deficit.
Collapse
Affiliation(s)
- Li-Juan Min
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan.
| | - Jun Iwanami
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| | - Masachika Shudou
- Division of Analytical Bio-Medicine, Advanced Research Support Center (ADRES), Ehime University, Graduate School of Medicine, Tohon, Ehime, 791-0295, Japan
| | - Hui-Yu Bai
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| | - Bao-Shuai Shan
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| | - Akinori Higaki
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| | - Masaki Mogi
- Department of Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, 791-0295, Japan
| | - Masatsugu Horiuchi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| |
Collapse
|
70
|
Xie B, Shi X, Xing Y, Tang Y. Association between atherosclerosis and Alzheimer's disease: A systematic review and meta-analysis. Brain Behav 2020; 10:e01601. [PMID: 32162494 PMCID: PMC7177569 DOI: 10.1002/brb3.1601] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND To evaluate the relationship between atherosclerosis and Alzheimer's disease (AD), we conducted a systematic review and meta-analysis to study the difference of carotid intima-media thickness (CIMT) and the prevalence of atherosclerosis between AD patients and non-AD controls. METHODS The studies on the association between atherosclerosis and AD were manually searched in PubMed, Embase, Cochrane Library, and CNKI (China National Knowledge Infrastructure) spanned to September 2018 according to PRISMA (the Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. RESULTS Thirteen studies were included in the final analysis, seven studies with data on the mean CIMT (610 cases and 417 controls) and ten studies reporting on the prevalence of atherosclerosis (1,698 cases and 6,452 controls). Compared with controls, AD group showed a significantly higher CIMT (overall standard mean difference = 0.94; 95% CI, 0.48-1.40; p < .0001) and an increased prevalence of atherosclerosis (OR = 1.46; 95% CI, 1.26-1.68; p < .0001). CONCLUSIONS Atherosclerosis is significantly associated with AD. CIMT might be a useful marker to predict the risk of AD and assess the vascular burden. The finding is also important for possible prevention and treatment of AD in the future.
Collapse
Affiliation(s)
- Beijia Xie
- Department of NeurologyInnovation Center for Neurological DisordersXuanwu HospitalNational Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Xinrui Shi
- Department of NeurologyInnovation Center for Neurological DisordersXuanwu HospitalNational Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Yi Xing
- Department of NeurologyInnovation Center for Neurological DisordersXuanwu HospitalNational Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Yi Tang
- Department of NeurologyInnovation Center for Neurological DisordersXuanwu HospitalNational Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Geriatric Cognitive DisordersNeurodegenerative Laboratory of Ministry of Education of the People's Republic of ChinaBeijingChina
| |
Collapse
|
71
|
Yoo JE, Shin DW, Han K, Kim D, Lee SP, Jeong SM, Lee J, Kim S. Blood Pressure Variability and the Risk of Dementia: A Nationwide Cohort Study. Hypertension 2020; 75:982-990. [PMID: 32148122 DOI: 10.1161/hypertensionaha.119.14033] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
To investigate the association between visit-to-visit variability in blood pressure and the incidence of dementia and its subtypes in a general population, we conducted a population-based retrospective cohort study using the Korean National Health Insurance System database. We identified 7 844 814 subjects without a history of any dementia who underwent ≥3 health examinations from 2005 to 2012 in the Korean National Health Insurance System cohort. Blood pressure variability (BPV) was measured using the variability independent of the mean, coefficient of variation, and SD. During the median follow-up of 6.2 years, there were 200 574 cases of all-cause dementia (2.8%), 165 112 cases of Alzheimer's disease (2.1%), and 27 443 cases of vascular dementia (0.3%). There was a linear association between higher BPV and outcome measures. In the multivariable adjusted model, the hazard ratios and 95% CIs of all-cause dementia were 1.06 (1.04-1.07) for the highest quartile of variability independent of the mean of diastolic blood pressure only, 1.09 (1.08-1.11) for that of systolic blood pressure only, and 1.18 (1.16-1.19) for that of both systolic and diastolic blood pressure compared with subjects having no highest quartile for BPV. Consistent results were noted for Alzheimer's disease and vascular dementia using other indices of variability and in various sensitivity and subgroup analyses. BPV is an independent predictor for developing dementia and its subtypes. A dose-response relationship was noted between higher BPV and dementia incidence. Reducing BPV may be a target for preventing dementia in the general population.
Collapse
Affiliation(s)
- Jung Eun Yoo
- From the Department of Family Medicine, Healthcare System Gangnam Center Seoul National University Hospital, Korea (J.E.Y.)
| | - Dong Wook Shin
- Department of Family Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (D.W.S.).,Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Korea (D.W.S.)
| | - Kyungdo Han
- Department of Biostatistics, The Catholic University of Korea, Seoul, Korea (K.H., D.K.)
| | - Dahye Kim
- Department of Biostatistics, The Catholic University of Korea, Seoul, Korea (K.H., D.K.)
| | - Seung-Pyo Lee
- Department of Internal Medicine (S.L.), Seoul National University Bundang Hospital & Seoul National University College of Medicine, Korea
| | - Su-Min Jeong
- Department of Family Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea and Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA (S.-M.J.)
| | - Jinkook Lee
- Department of Economics, Center for Economic & Social Research, University of Southern California, Los Angeles, and RANC Corporation, Santa Monica, CA, USA (J.L.)
| | - SangYun Kim
- Department of Neurology (S.Y.K.), Seoul National University Bundang Hospital & Seoul National University College of Medicine, Korea
| |
Collapse
|
72
|
Conti I, Varano G, Simioni C, Laface I, Milani D, Rimondi E, Neri LM. miRNAs as Influencers of Cell-Cell Communication in Tumor Microenvironment. Cells 2020; 9:cells9010220. [PMID: 31952362 PMCID: PMC7016744 DOI: 10.3390/cells9010220] [Citation(s) in RCA: 55] [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: 12/16/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/14/2022] Open
Abstract
microRNAs (miRNAs) are small noncoding RNAs that regulate gene expression at the posttranscriptional level, inducing the degradation of the target mRNA or translational repression. MiRNAs are involved in the control of a multiplicity of biological processes, and their absence or altered expression has been associated with a variety of human diseases, including cancer. Recently, extracellular miRNAs (ECmiRNAs) have been described as mediators of intercellular communication in multiple contexts, including tumor microenvironment. Cancer cells cooperate with stromal cells and elements of the extracellular matrix (ECM) to establish a comfortable niche to grow, to evade the immune system, and to expand. Within the tumor microenvironment, cells release ECmiRNAs and other factors in order to influence and hijack the physiological processes of surrounding cells, fostering tumor progression. Here, we discuss the role of miRNAs in the pathogenesis of multicomplex diseases, such as Alzheimer’s disease, obesity, and cancer, focusing on the contribution of both intracellular miRNAs, and of released ECmiRNAs in the establishment and development of cancer niche. We also review growing evidence suggesting the use of miRNAs as novel targets or potential tools for therapeutic applications.
Collapse
Affiliation(s)
- Ilaria Conti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Gabriele Varano
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Carolina Simioni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Ilaria Laface
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Daniela Milani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Erika Rimondi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Luca M. Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
- LTTA—Electron Microscopy Center, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: ; Tel.: +39-0532-455940
| |
Collapse
|
73
|
Ciacciarelli A, Sette G, Giubilei F, Orzi F. Chronic cerebral hypoperfusion: An undefined, relevant entity. J Clin Neurosci 2020; 73:8-12. [PMID: 31948882 DOI: 10.1016/j.jocn.2020.01.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/06/2020] [Indexed: 01/11/2023]
Abstract
Despite the large body of data available, chronic cerebral hypoperfusion lacks an operative definition. In a tautological way, the term hypoperfusion is being referred to conditions of "inadequate blood flow", "defects of perfusion" or "dysfunction of autoregulation". The chronicity refers to sustained conditions or wavering states characterized by repeated phases of inefficient functional hyperemia. The phenomenon may affect the whole brain or defined areas. A few defined clinical disorders, including heart failure, hypotension, atherosclerosis of large or small vessels and carotid stenosis are thought to cause progressive brain disorders due to chronic hypoperfusion. The clinical relevance manifests mostly as neurocognitive disorders associated with neuroimaging changes.The available data support a conceptual framework that considerschronic cerebral hypoperfusiona likely, relevant pathogenic mechanism for the neurodegeneration-like progression of the neurocognitive disorders. The relationship between neuropathology, cerebral perfusion, and symptoms progression is, however, elusive for several aspects. Typical microangiopathy findings, such as MRI white matter hyperintensities, may appear in individuals without any cerebrovascular risk or vascular lesions. Pathology features of the MRI changes, such as demyelination and gliosis, may result from dysfunction of the neuro-vascular unit not directly associated withvascular mechanisms. In this review, we aim to overview the most common clinical conditions thought to reflect chronic hypoperfusion.
Collapse
Affiliation(s)
- Antonio Ciacciarelli
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), "SAPIENZA" University of Rome, Sant'Andrea University Hospital, Rome, Italy.
| | - Giuliano Sette
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), "SAPIENZA" University of Rome, Sant'Andrea University Hospital, Rome, Italy
| | - Franco Giubilei
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), "SAPIENZA" University of Rome, Sant'Andrea University Hospital, Rome, Italy
| | - Francesco Orzi
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), "SAPIENZA" University of Rome, Sant'Andrea University Hospital, Rome, Italy
| |
Collapse
|
74
|
Stovicek PO, Friedmann C, Marinescu D, Văduva IA, Bondari S, Trifu SC, Marinescu I. Mild TBI in the elderly - risk factor for rapid cognitive impairment in Alzheimer's disease. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2020; 61:61-72. [PMID: 32747896 PMCID: PMC7728108 DOI: 10.47162/rjme.61.1.07] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/15/2020] [Indexed: 11/23/2022]
Abstract
In recent decades, traumatic brain injury (TBI) has become one of the most important health problems worldwide and is a major cause of morbidity, mortality and economic losses. Mild traumatic brain injury (mTBI) is less considered, with clinical underestimation leading to an epidemiological underevaluation of its incidence. Many of the signs and symptoms induced by mTBI are difficult to highlight clinically, especially those related to cognitive, behavioral, or emotional impairment. The complexity of the biological mechanisms induced by mTBI in the elderly determines synchronous pathogenic actions in which the vascular, inflammatory and neurodegenerative elements are intertwined. It is difficult to highlight a major pathogenic factor, since they act simultaneously, multimodally, in a real pathogenic cascade. The identification of mTBI and cerebral vascular changes by neuroimaging techniques, transcranial Doppler (TCD) or biological markers, suggests a potential prophylactic intervention by using neuroprotective factors as early as possible. Proper prophylaxis measures with neurotrophic treatment, rebalancing the gamma-aminobutyric acid (GABA)∕glutamate balance and combating the chronic inflammatory process, can become important pharmacological therapeutic targets.
Collapse
|
75
|
Gustavsson AM, van Westen D, Stomrud E, Engström G, Nägga K, Hansson O. Midlife Atherosclerosis and Development of Alzheimer or Vascular Dementia. Ann Neurol 2019; 87:52-62. [PMID: 31721283 PMCID: PMC6973178 DOI: 10.1002/ana.25645] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 10/03/2019] [Accepted: 11/10/2019] [Indexed: 12/28/2022]
Abstract
Objective To investigate whether midlife atherosclerosis is associated with different dementia subtypes and related underlying pathologies. Methods Participants comprised the cardiovascular cohort of the Swedish prospective population‐based Malmö Diet and Cancer Study (N = 6,103). Carotid plaques and intima media thickness (IMT) were measured at baseline (1991–1994). Dementia incidence until 2014 was obtained from national registers. Diagnoses were reviewed and validated in medical records. In a cognitively unimpaired subcohort (n = 330), β‐amyloid42 and tau were quantified in cerebrospinal fluid (CSF), and white matter hyperintensity volume, lacunar infarcts, and cerebral microbleeds were estimated on magnetic resonance imaging (2009–2015). Results During 20 years of follow‐up, 462 individuals developed dementia (mean age at baseline = 57.5 ± 5.9 years, 58% women). Higher IMT in midlife was associated with an increased hazard ratio (HR) of all‐cause dementia (adjusted HR = 1.14 [95% confidence interval (CI) = 1.03–1.26]) and vascular dementia (adjusted HR = 1.32 [95% CI = 1.10–1.57]) but not Alzheimer disease (AD) dementia (adjusted HR = 0.95 [95% CI = 0.77–1.17]). Carotid plaques were associated with vascular dementia when assessed as a 3‐graded score (adjusted HR = 1.90 [95% CI = 1.07–3.38]). In the cognitively unimpaired subcohort (53.8 ± 4.6 years at baseline, 60% women), higher IMT in midlife was associated with development of small vessel disease (adjusted odds ratio [OR] = 1.47 [95% CI = 1.05–2.06]) but not significantly with abnormal CSF AD biomarkers (adjusted OR = 1.28 [95% CI = 0.87–1.90] for Aβ42 and 1.35 [95% CI = 0.86–2.13] for Aβ42/p‐tau). Carotid plaques revealed no significant association with any of the underlying brain pathologies. Interpretation Our findings support an association between midlife atherosclerosis and development of vascular dementia and cerebral small vessel disease but not between atherosclerosis and subsequent AD dementia or AD pathology. ANN NEUROL 2020;87:52–62
Collapse
Affiliation(s)
- Anna-Märta Gustavsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö.,Memory Clinic, Skåne University Hospital, Malmö
| | - Danielle van Westen
- Diagnostic Radiology, Department of Clinical Sciences Lund, Lund University, Lund
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö.,Memory Clinic, Skåne University Hospital, Malmö
| | - Gunnar Engström
- Cardiovascular Epidemiology, Department of Clinical Sciences Malmö, Lund University, Malmö
| | - Katarina Nägga
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö.,Department of Acute Internal Medicine and Geriatrics, Linköping University, Linköping, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö.,Memory Clinic, Skåne University Hospital, Malmö
| |
Collapse
|
76
|
Sharma A, Kumar Y. Nature's Derivative(s) as Alternative Anti-Alzheimer's Disease Treatments. J Alzheimers Dis Rep 2019; 3:279-297. [PMID: 31867567 PMCID: PMC6918879 DOI: 10.3233/adr-190137] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2019] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD), the 'Plague of Twenty-First Century,' is a crippling neurodegenerative disease that affects a majority of the older population globally. By 2050, the incidence of AD is expected to rise to 135 million, while no treatment(s) that can reverse or control the progression of AD are currently available. The treatment(s) in use are limited in their ability to manage the symptoms or slow the progression of the disease and can lead to some severe side effects. The overall care is economically burdensome for the affected individuals as well as the caretakers or family members. Thus, there is a pressing need to identify and develop much safer alternative therapies that can better manage AD. This review discusses a multitude of such treatments borrowed from Ayurveda, traditional Chinese practices, meditation, and exercising for AD treatment. These therapies are in practice since ancient times and reported to be beneficial as anti-AD therapies. Ayurvedic drugs like turmeric, Brahmi, Ashwagandha, etc., management of stress by meditation, regular exercising, and acupuncture have been reported to be efficient in their anti-AD usage. Besides, a combination of vitamins and natural dietary intakes is likely to play a significant role in combating AD. We conclude that the use of such alternative strategies will be a stepping-stone in preventing, treating, curing, or managing the disease.
Collapse
Affiliation(s)
- Anuja Sharma
- Department of Biological Sciences and Engineering (BSE), Netaji Subhas University of Technology (NSUT), Formerly Netaji Subhas Institute of Technology (NSIT), Azad Hind Fauz Marg, New Delhi, India
| | - Yatender Kumar
- Department of Biological Sciences and Engineering (BSE), Netaji Subhas University of Technology (NSUT), Formerly Netaji Subhas Institute of Technology (NSIT), Azad Hind Fauz Marg, New Delhi, India
| |
Collapse
|
77
|
de Heus RA, Olde Rikkert MG, Tully PJ, Lawlor BA, Claassen JA. Blood Pressure Variability and Progression of Clinical Alzheimer Disease. Hypertension 2019; 74:1172-1180. [DOI: 10.1161/hypertensionaha.119.13664] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Blood pressure variability (BPV) has been shown to have predictive value over blood pressure (BP) levels alone in stroke patients. We assessed whether BPV predicts cognitive and functional decline in Alzheimer disease, using data from a randomized trial (NILVAD [A European Multicentre Double-blind Placebo-controlled Phase III Trial of Nilvadipine in Mild to Moderate Alzheimer’s Disease]). Patients with mild-to-moderate Alzheimer disease were included if they had ≥3 office BP measurements available to determine visit-to-visit BPV. Day-to-day BPV was assessed using home BP measurements in a subsample. The variation independent of mean was used to calculate BPV. Outcomes were change in Alzheimer’s Disease Assessment Scale–cognitive subscale-12 and Disability Assessment for Dementia after 1 and 1.5 years. A total of 460 patients aged 72.1 (SD=8.1) years, with mean BP of 134.0/75.1 (10.9/6.3) mm Hg were included. After 1 year, patients in the highest quartile of BPV had deteriorated more on Alzheimer’s Disease Assessment Scale–cognitive subscale compared with patients in the lowest quartile (systolic: β, 2.24 [95% CI, 0.11–4.38],
P
=0.040; diastolic: β, 2.54 [95% CI, 0.33–4.75]
P
=0.024). This association was still present after 1.5 years (systolic: β, 2.86 [95% CI, 0.35–5.36],
P
=0.026; diastolic: β, 3.30 [95% CI, 0.67–5.93],
P
=0.014). There was no effect of visit-to-visit BPV on Disability Assessment for Dementia. Day-to-day BPV was available for 46 patients. Significant associations were observed between day-to-day BPV and deterioration on Alzheimer’s Disease Assessment Scale–cognitive subscale (systolic:
P
=0.036) and Disability Assessment for Dementia (systolic:
P
=0.020; diastolic:
P
=0.007) after 1 year, but not after 1.5 years. All associations were adjusted for potential confounders, including intervention group. In conclusion, this post hoc analysis indicates that higher visit-to-visit and day-to-day BPV might be associated with progression of Alzheimer disease. Targeting BPV may be a future target to slow decline in patients with Alzheimer disease.
Clinical Trial Registration
URL:
https://www.clinicaltrials.gov
. Unique identifier: NCT02017340.
Collapse
Affiliation(s)
- Rianne A.A. de Heus
- From the Department of Geriatric Medicine, Radboud umc University Medical Center, Radboudumc Alzheimer Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands (R.A.A.d.H., M.G.M.O.R., J.A.H.R.C.)
| | - Marcel G.M. Olde Rikkert
- From the Department of Geriatric Medicine, Radboud umc University Medical Center, Radboudumc Alzheimer Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands (R.A.A.d.H., M.G.M.O.R., J.A.H.R.C.)
| | - Phillip J. Tully
- School of Medicine, The University of Adelaide, Australia (P.J.T.)
| | - Brian A. Lawlor
- Mercer’s Institute for Research on Ageing, St. James’s Hospital, Dublin, Ireland (B.A.L.)
- Department of Medical Gerontology, Trinity College Institute of Neuroscience, Dublin, Ireland (B.A.L.)
| | - Jurgen A.H.R. Claassen
- From the Department of Geriatric Medicine, Radboud umc University Medical Center, Radboudumc Alzheimer Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, the Netherlands (R.A.A.d.H., M.G.M.O.R., J.A.H.R.C.)
| | | |
Collapse
|
78
|
Ajikumar A, Long MB, Heath PR, Wharton SB, Ince PG, Ridger VC, Simpson JE. Neutrophil-Derived Microvesicle Induced Dysfunction of Brain Microvascular Endothelial Cells In Vitro. Int J Mol Sci 2019; 20:E5227. [PMID: 31652502 PMCID: PMC6834153 DOI: 10.3390/ijms20205227] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/16/2019] [Accepted: 10/18/2019] [Indexed: 12/19/2022] Open
Abstract
The blood-brain barrier (BBB), composed of brain microvascular endothelial cells (BMEC) that are tightly linked by tight junction (TJ) proteins, restricts the movement of molecules between the periphery and the central nervous system. Elevated systemic levels of neutrophils have been detected in patients with altered BBB function, but the role of neutrophils in BMEC dysfunction is unknown. Neutrophils are key players of the immune response and, when activated, produce neutrophil-derived microvesicles (NMV). NMV have been shown to impact the integrity of endothelial cells throughout the body and we hypothesize that NMV released from circulating neutrophils interact with BMEC and induce endothelial cell dysfunction. Therefore, the current study investigated the interaction of NMV with human BMEC and determined whether they altered gene expression and function in vitro. Using flow cytometry and confocal imaging, NMV were shown to be internalized by the human cerebral microvascular endothelial cell line hCMEC/D3 via a variety of energy-dependent mechanisms, including endocytosis and macropinocytosis. The internalization of NMV significantly altered the transcriptomic profile of hCMEC/D3, specifically inducing the dysregulation of genes associated with TJ, ubiquitin-mediated proteolysis and vesicular transport. Functional studies confirmed NMV significantly increased permeability and decreased the transendothelial electrical resistance (TEER) of a confluent monolayer of hCMEC/D3. These findings indicate that NMV interact with and affect gene expression of BMEC as well as impacting their integrity. We conclude that NMV may play an important role in modulating the permeability of BBB during an infection.
Collapse
Affiliation(s)
- Anjana Ajikumar
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK.
| | - Merete B Long
- Department of Infection Immunity and Cardiovascular Diseases, University of Sheffield, Medical School, Sheffield S10 2RX, UK.
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK.
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK.
| | - Paul G Ince
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK.
| | - Victoria C Ridger
- Department of Infection Immunity and Cardiovascular Diseases, University of Sheffield, Medical School, Sheffield S10 2RX, UK.
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK.
| |
Collapse
|
79
|
Rabin JS, Schultz AP, Hedden T, Viswanathan A, Marshall GA, Kilpatrick E, Klein H, Buckley RF, Yang HS, Properzi M, Rao V, Kirn DR, Papp KV, Rentz DM, Johnson KA, Sperling RA, Chhatwal JP. Interactive Associations of Vascular Risk and β-Amyloid Burden With Cognitive Decline in Clinically Normal Elderly Individuals: Findings From the Harvard Aging Brain Study. JAMA Neurol 2019; 75:1124-1131. [PMID: 29799986 DOI: 10.1001/jamaneurol.2018.1123] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Importance Identifying asymptomatic individuals at high risk of impending cognitive decline because of Alzheimer disease is crucial for successful prevention of dementia. Vascular risk and β-amyloid (Aβ) pathology commonly co-occur in older adults and are significant causes of cognitive impairment. Objective To determine whether vascular risk and Aβ burden act additively or synergistically to promote cognitive decline in clinically normal older adults; and, secondarily, to evaluate the unique influence of vascular risk on prospective cognitive decline beyond that of commonly used imaging biomarkers, including Aβ burden, hippocampal volume, fludeoxyglucose F18-labeled (FDG) positron emission tomography (PET), and white matter hyperintensities, a marker of cerebrovascular disease. Design, Setting, and Participants In this longitudinal observational study, we examined clinically normal older adults from the Harvard Aging Brain Study. Participants were required to have baseline imaging data (FDG-PET, Aβ-PET, and magnetic resonance imaging), baseline medical data to quantify vascular risk, and at least 1 follow-up neuropsychological visit. Data collection began in 2010 and is ongoing. Data analysis was performed on data collected between 2010 and 2017. Main Outcomes and Measures Vascular risk was quantified using the Framingham Heart Study general cardiovascular disease (FHS-CVD) risk score. We measured Aβ burden with Pittsburgh Compound-B PET. Cognition was measured annually with the Preclinical Alzheimer Cognitive Composite. Models were corrected for baseline age, sex, years of education, and apolipoprotein E ε4 status. Results Of the 223 participants, 130 (58.3%) were women. The mean (SD) age was 73.7 (6.0) years, and the mean (SD) follow-up time was 3.7 (1.2) years. Faster cognitive decline was associated with both a higher FHS-CVD risk score (β = -0.064; 95% CI, -0.094 to -0.033; P < .001) and higher Aβ burden (β = -0.058; 95% CI, -0.079 to -0.037; P < .001). The interaction of the FHS-CVD risk score and Aβ burden with time was significant (β = -0.040, 95% CI, -0.062 to -0.018; P < .001), suggesting a synergistic effect. The FHS-CVD risk score remained robustly associated with prospective cognitive decline (β = -0.055; 95% CI, -0.086 to -0.024; P < .001), even after adjustment for Aβ burden, hippocampal volume, FDG-PET uptake, and white matter hyperintensities. Conclusions and Relevance In this study, vascular risk was associated with prospective cognitive decline in clinically normal older adults, both alone and synergistically with Aβ burden. Vascular risk may complement imaging biomarkers in assessing risk of prospective cognitive decline in preclinical Alzheimer disease.
Collapse
Affiliation(s)
- Jennifer S Rabin
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Trey Hedden
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Anand Viswanathan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston.,J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston
| | - Gad A Marshall
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Emily Kilpatrick
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Hannah Klein
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston.,Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia.,Melbourne School of Psychological Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Hyun-Sik Yang
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael Properzi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Vaishnavi Rao
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Dylan R Kirn
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Kathryn V Papp
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dorene M Rentz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
80
|
Saito S, Yamamoto Y, Ihara M. Development of a Multicomponent Intervention to Prevent Alzheimer's Disease. Front Neurol 2019; 10:490. [PMID: 31139139 PMCID: PMC6518668 DOI: 10.3389/fneur.2019.00490] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
Recent advances in vascular risk management have successfully reduced the prevalence of Alzheimer's Disease (AD) in several epidemiologic investigations. It is now widely accepted that cerebrovascular disease is both directly and indirectly involved in AD pathogenesis. Herein, we review the non-pharmacological and pharmacological therapeutic approaches for AD treatment. MIND [Mediterranean and DASH (Dietary Approaches to Stop Hypertension) Intervention for Neurodegenerative Delay] diet is an important dietary treatment for prevention of AD. Multi domain intervention including diet, exercise, cognitive training, and intensive risk managements also prevented cognitive decline in the Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) study. To confirm these favorable effects of life-style intervention, replica studies are being planned worldwide. Promotion of β-amyloid (Aβ) clearance has emerged as a promising pharmacological approach because insufficient removal of Aβ is more important than excessive Aβ production in the pathogenesis of the majority of AD patients. Most AD brains exhibit accompanying cerebral amyloid angiopathy, and Aβ distribution in cerebral amyloid angiopathy closely corresponds with the intramural periarterial drainage (IPAD) route, emphasizing the importance of Aβ clearance. In view of these facts, promotion of the major vascular-mediated Aβ elimination systems, including capillary transcytosis, the glymphatic system, and IPAD, have emerged as new treatment strategies in AD. In particular, the beneficial effects of cilostazol were shown in several clinical observation studies, and cilostazol facilitated IPAD in a rodent AD model. The COMCID (Cilostazol for prevention of Conversion from MCI to Dementia) trial, evaluating the efficacy of cilostazol for patients with mild cognitive impairment is currently ongoing in Japan. Such therapeutic approaches involving maintenance of cerebrovascular integrity and promotion of vascular-mediated Aβ clearance have the potential to be mainstream treatments for sporadic AD.
Collapse
Affiliation(s)
- Satoshi Saito
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan.,Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Yumi Yamamoto
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| |
Collapse
|
81
|
Marston KJ, Brown BM, Rainey-Smith SR, Peiffer JJ. Resistance Exercise-Induced Responses in Physiological Factors Linked with Cognitive Health. J Alzheimers Dis 2019; 68:39-64. [DOI: 10.3233/jad-181079] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Kieran J. Marston
- Department of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia
- Ageing, Cognition and Exercise (ACE) Research Group, Murdoch University, Perth, Western Australia, Australia
| | - Belinda M. Brown
- Department of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia
- Ageing, Cognition and Exercise (ACE) Research Group, Murdoch University, Perth, Western Australia, Australia
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Australian Alzheimer’s Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| | - Stephanie R. Rainey-Smith
- Ageing, Cognition and Exercise (ACE) Research Group, Murdoch University, Perth, Western Australia, Australia
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Australian Alzheimer’s Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| | - Jeremiah J. Peiffer
- Department of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia
- Ageing, Cognition and Exercise (ACE) Research Group, Murdoch University, Perth, Western Australia, Australia
| |
Collapse
|
82
|
Yamauchi H, Kagawa S, Takahashi M, Oishi N, Ono M, Higashi T. Misery perfusion and amyloid deposition in atherosclerotic major cerebral artery disease. NEUROIMAGE-CLINICAL 2019; 22:101762. [PMID: 30884364 PMCID: PMC6424140 DOI: 10.1016/j.nicl.2019.101762] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 01/08/2019] [Accepted: 03/10/2019] [Indexed: 11/18/2022]
Abstract
Although experimental studies have shown that global cerebral hypoperfusion leads to amyloid deposition in the hemisphere with carotid artery occlusion in rodents, the results of such occurrence are controversial in humans. Hence, we aim to determine whether global cerebral hypoperfusion leading to decreased blood flow relative to metabolic demand [increased oxygen extraction fraction (OEF), misery perfusion] is associated with increases in amyloid deposition in the hemisphere with atherosclerotic major cerebral artery disease in patients. We evaluated the distribution of β-amyloid plaques using positron emission tomography and a [18F]-pyridylbenzofuran derivative (18F-FPYBF-2) in 13 patients with unilateral atherosclerotic disease of the internal carotid artery (ICA) or middle cerebral artery (MCA) disease and no cortical infarction. The distribution volume ratio (DVR) of 18F- FPYBF-2 was calculated using dynamic data and Logan graphical analysis with reference tissue and was correlated with the cerebral blood flow (CBF), cerebral metabolic rate of oxygen (CMRO2), and OEF, obtained from 15O-gas PET. The mean cortical value was calculated as the mean value within the frontal, posterior cingulate, precuneus, parietal, and lateral temporal cortical regions. Significant reductions in CBF and CMRO2 and increases in OEF were found in the hemisphere ipsilateral to the arterial lesion compared with the contralateral hemisphere. There was no significant difference for 18F-FPYBF-2 DVR between hemispheres. The ipsilateral to contralateral ratio of the 18F- FPYBF-2 DVR was increased in 3 patients, while the ipsilateral to contralateral OEF ratio was increased in 4 patients. The incidence of an increased hemispheric DVR ratio was significantly higher in patients with an increased hemispheric OEF ratio (3/4) than in patients without (0/9) (p < 0.02). Although the 18F- FPYBF-2 DVR in the ipsilateral hemisphere was positively correlated with OEF after adjustment for the 18F- FPYBF-2 DVR in the contralateral hemisphere using multiple regression analysis (p < 0.05), the contribution rate of OEF was small (R2 = 5.5%). Only one of the 4 patients with an increased hemispheric OEF ratio showed amyloid positivity based on the DVR value. In atherosclerotic major cerebral artery disease, misery perfusion accompanied only small increases of amyloid deposition at best. Misery perfusion was not associated with amyloid positivity. Misery perfusion accompanied only small increases of amyloid deposition at best. Relative oxygen extraction fraction correlated with relative amyloid deposition. Misery perfusion was not associated with amyloid positivity.
Collapse
Affiliation(s)
- Hiroshi Yamauchi
- Division of PET Imaging, Shiga Medical Centre Research Institute, Moriyama, Japan.
| | - Shinya Kagawa
- Division of PET Imaging, Shiga Medical Centre Research Institute, Moriyama, Japan
| | - Masaaki Takahashi
- Division of PET Imaging, Shiga Medical Centre Research Institute, Moriyama, Japan
| | - Naoya Oishi
- Human Brain Research Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Tatsuya Higashi
- Division of PET Imaging, Shiga Medical Centre Research Institute, Moriyama, Japan; National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
83
|
Rabin JS, Yang HS, Schultz AP, Hanseeuw BJ, Hedden T, Viswanathan A, Gatchel JR, Marshall GA, Kilpatrick E, Klein H, Rao V, Buckley RF, Yau WYW, Kirn DR, Rentz DM, Johnson KA, Sperling RA, Chhatwal JP. Vascular Risk and β-Amyloid Are Synergistically Associated with Cortical Tau. Ann Neurol 2019; 85:272-279. [PMID: 30565287 DOI: 10.1002/ana.25399] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 12/14/2018] [Accepted: 12/14/2018] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Neuropathological studies have demonstrated that cerebrovascular disease and Alzheimer disease (AD) pathology frequently co-occur in older adults. The extent to which cerebrovascular disease influences the progression of AD pathology remains unclear. Leveraging newly available positron emission tomography (PET) imaging, we examined whether a well-validated measure of systemic vascular risk and β-amyloid (Aβ) burden have an interactive association with regional tau burden. METHODS Vascular risk was quantified at baseline in 152 clinically normal older adults (mean age = 73.5 ± 6.1 years) with the office-based Framingham Heart Study cardiovascular disease risk algorithm (FHS-CVD). We acquired Aβ (11 C-Pittsburgh compound B) and tau (18 F-flortaucipir) PET imaging on the same participants. Aβ PET was performed at baseline; tau PET was acquired on average 2.98 ± 1.1 years later. Tau was measured in the entorhinal cortex (EC), an early site of tau deposition, and in the inferior temporal cortex (ITC), an early site of neocortical tau accumulation associated with AD. Linear regression models examined FHS-CVD and Aβ as interactive predictors of tau deposition, adjusting for age, sex, APOE ε4 status, and the time interval between baseline and the tau PET scan. RESULTS We observed a significant interaction between FHS-CVD and Aβ burden on subsequently measured ITC tau (p < 0.001), whereby combined higher FHS-CVD and elevated Aβ burden was associated with increased tau. The interaction was not significant for EC tau (p = 0.16). INTERPRETATION Elevated vascular risk may influence tau burden when coupled with high Aβ burden. These results suggest a potential link between vascular risk and tau pathology in preclinical AD. Ann Neurol 2019; 1-8 ANN NEUROL 2019;85:272-279.
Collapse
Affiliation(s)
- Jennifer S Rabin
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Hyun-Sik Yang
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Bernard J Hanseeuw
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Trey Hedden
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Anand Viswanathan
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA
| | - Jennifer R Gatchel
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Gad A Marshall
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Emily Kilpatrick
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Hannah Klein
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Vaishnavi Rao
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia.,Melbourne School of Psychological Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Wai-Ying Wendy Yau
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Dylan R Kirn
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Dorene M Rentz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
84
|
Spallazzi M, Dobisch L, Becke A, Berron D, Stucht D, Oeltze-Jafra S, Caffarra P, Speck O, Düzel E. Hippocampal vascularization patterns: A high-resolution 7 Tesla time-of-flight magnetic resonance angiography study. NEUROIMAGE-CLINICAL 2018; 21:101609. [PMID: 30581106 PMCID: PMC6413539 DOI: 10.1016/j.nicl.2018.11.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/03/2018] [Accepted: 11/18/2018] [Indexed: 01/11/2023]
Abstract
Considerable evidence suggests a close relationship between vascular and degenerative pathology in the human hippocampus. Due to the intrinsic fragility of its vascular network, the hippocampus appears less able to cope with hypoperfusion and anoxia than other cortical areas. Although hippocampal blood supply is generally provided by the collateral branches of the posterior cerebral artery (PCA) and the anterior choroidal artery (AChA), different vascularization patterns have been detected postmortem. To date, a methodology that enables the classification of individual hippocampal vascularization patterns in vivo has not been established. In this study, using high-resolution 7 Tesla time-of-flight angiography data (0.3 mm isotropic resolution) in young adults, we classified individual variability in hippocampal vascularization patterns involved in medial temporal lobe blood supply in vivo. A strong concordance between our classification and previous autopsy findings was found, along with interesting anatomical observations, such as the variable contribution of the AChA to hippocampal supply, the relationships between hippocampal and PCA patterns, and the different distribution patterns of the right and left hemispheres. The approach presented here for determining hippocampal vascularization patterns in vivo may provide new insights into not only the vulnerability of the hippocampus to vascular and neurodegenerative diseases but also hippocampal vascular plasticity after exercise training. First attempt to classify human hippocampal vascularization in vivo using 7Tesla Angiography Good concordance between in vivo findings and autopsy studies A new avenue to investigate interindividual variability in hippocampal vascular plasticity A new avenue for linking individual vascular anatomical phenotypes to neurodegenerative and vascular pathology
Collapse
Affiliation(s)
| | - Laura Dobisch
- Institute for Cognitive Neurology and Dementia Research, University of Magdeburg, Germany; German Center for Neurodegenerative Diseases, Magdeburg, Germany
| | - Andreas Becke
- Institute for Cognitive Neurology and Dementia Research, University of Magdeburg, Germany; German Center for Neurodegenerative Diseases, Magdeburg, Germany
| | - David Berron
- Institute for Cognitive Neurology and Dementia Research, University of Magdeburg, Germany; German Center for Neurodegenerative Diseases, Magdeburg, Germany
| | - Daniel Stucht
- Department of Biomedical Magnetic Resonance, Otto-von-Guericke University Magdeburg, Germany
| | | | | | - Oliver Speck
- German Center for Neurodegenerative Diseases, Magdeburg, Germany; Department of Biomedical Magnetic Resonance, Otto-von-Guericke University Magdeburg, Germany; Leibniz Institute for Neurobiology, Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Emrah Düzel
- Institute for Cognitive Neurology and Dementia Research, University of Magdeburg, Germany; German Center for Neurodegenerative Diseases, Magdeburg, Germany; Institute of Cognitive Neuroscience, Univ. College London, London, United Kingdom.
| |
Collapse
|
85
|
Boublay N, Fédérico D, Pesce A, Verny M, Blanc F, Paccalin M, Desmidt T, Grosmaître P, Moreaud O, Relland S, Bravant E, Bouet R, Krolak-Salmon P. Study protocol on Alzheimer's disease and related disorders: focus on clinical and imaging predictive markers in co-existing lesions. BMC Geriatr 2018; 18:280. [PMID: 30428832 PMCID: PMC6236893 DOI: 10.1186/s12877-018-0949-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 10/16/2018] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND One of the crucial challenges for the future of therapeutic approaches to Alzheimer's disease (AD) is to target the main pathological processes responsible for disability and dependency. However, a progressive cognitive impairment occurring after the age of 70, the main population affected by dementia, is often related to mixed lesions of neurodegenerative and vascular origins. Whereas young patients are mostly affected by pure lesions, ageing favours the occurrence of co-lesions of AD, cerebrovascular disease (CVD) and Lewy body dementia (LBD). Most of clinical studies report on functional and clinical disabilities in patients with presumed pure pathologies. But, the weight of co-morbid processes involved in the transition from an independent functional status to disability in the elderly with co-lesions still remains to be elucidated. Neuropathological examination often performed at late stages cannot answer this question at mild or moderate stages of cognitive disorders. Brain MRI, Single Photon Emission Computed Tomography (SPECT) with DaTscan®, amyloid Positron Emission Tomography (PET) and CerebroSpinal Fluid (CSF) AD biomarkers routinely help in performing the diagnosis of underlying lesions. The combination of these measures seems to be of incremental value for the diagnosis of mixed profiles of AD, CVD and LBD. The aim is to determine the clinical, neuropsychological, neuroradiological and biological features the most predictive of cognitive, behavioral and functional impairment at 2 years in patients with co-existing lesions. METHODS A multicentre and prospective cohort study with clinical, neuro-imaging and biological markers assessment will recruit 214 patients over 70 years old with a cognitive disorder of AD, cerebrovascular and Lewy body type or with coexisting lesions of two or three of these pathologies and fulfilling the diagnostic criteria for dementia at a mild to moderate stage. Patients will be followed every 6 months (clinical, neuropsychological and imaging examination and collection of cognitive, behavioural and functional impairment) for 24 months. DISCUSSION This study aims at identifying the best combination of markers (clinical, neuropsychological, MRI, SPECT-DaTscan®, PET and CSF) to predict disability progression in elderly patients presenting coexisting patterns. TRIAL REGISTRATION NCT02052947 .
Collapse
Affiliation(s)
- Nawele Boublay
- Clinical and Research Memory Centre of Lyon, Hospital of Charpennes, Hospices Civils de Lyon, Lyon, France
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Brain Dynamics and Cognition Team, F-69000 Lyon, France
- University Lyon, F-69000 Lyon, France
- Hospices Civils de Lyon, Pôle Information Médicale Evaluation Recherche, F-69424 Lyon, France
- Hospices civils de Lyon, hôpital des Charpennes, 27 rue Gabriel Péri, 69100 Villeurbanne, France
| | - Denis Fédérico
- Clinical and Research Memory Centre of Lyon, Hospital of Charpennes, Hospices Civils de Lyon, Lyon, France
| | - Alain Pesce
- Département de gérontologie clinique et centre mémoire, Centre Rainier III, Monaco, France
| | - Marc Verny
- Clinical and Research Memory Centre and Geriatrics department of Ile de France Sud, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
- University Pierre et Marie Curie et DHU FAST, UMR 8256 (CNRS), Paris, France
| | - Frédéric Blanc
- Geriatrics day hospital. Geriatrics department, Memory Resources and Research Centre (CMRR), University Hospital of Strasbourg, Strasbourg, France
- Team IMIS/Neurocrypto, French National Center for Scientific Research (CNRS), ICube Laboratory and Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, Strasbourg, France
| | - Marc Paccalin
- Clinical and Research Memory Centre of Poitiers, CHU Poitiers, Poitiers, France
- Pôle de Gériatrie CHU Poitiers 86000 Poitiers, 3INSERM, CHU de Poitiers, Université de Poitiers, centre d’investigation clinique CIC1402, Poitiers, France
| | - Thomas Desmidt
- Clinical and Research Memory Centre of Tours, CHRU Tours, Tours, France
| | - Pierre Grosmaître
- Clinical and Research Memory Centre of Lyon, Hospital of Dugoujon, Hospices Civils de Lyon, Lyon, France
| | - Olivier Moreaud
- Clinical and Research Memory Centre of Grenoble Arc Alpin, Pôle de psychiatrie et neurologie, CHU de Grenoble, Laboratoire de psychologie et neurocognition, CNRS UMR 5105, Grenoble, France
| | - Solveig Relland
- Clinical and Research Memory Centre of Lyon, Hospital of Charpennes, Hospices Civils de Lyon, Lyon, France
| | - Estelle Bravant
- Hospices Civils de Lyon, Pôle Information Médicale Evaluation Recherche, F-69424 Lyon, France
| | - Romain Bouet
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Brain Dynamics and Cognition Team, F-69000 Lyon, France
| | - Pierre Krolak-Salmon
- Clinical and Research Memory Centre of Lyon, Hospital of Charpennes, Hospices Civils de Lyon, Lyon, France
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Brain Dynamics and Cognition Team, F-69000 Lyon, France
- Clinical Research Centre (Vieillissement – Cerveau - Fragilité), Hospital of Charpennes, Hospices Civils de Lyon, Lyon, France
- University Lyon, F-69000 Lyon, France
| |
Collapse
|
86
|
Jung NY, Han JC, Ong YT, Cheung CYL, Chen CP, Wong TY, Kim HJ, Kim YJ, Lee J, Lee JS, Jang YK, Kee C, Lee KH, Kim EJ, Seo SW, Na DL. Retinal microvasculature changes in amyloid-negative subcortical vascular cognitive impairment compared to amyloid-positive Alzheimer's disease. J Neurol Sci 2018; 396:94-101. [PMID: 30447606 DOI: 10.1016/j.jns.2018.10.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 10/25/2018] [Accepted: 10/29/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND PURPOSE To investigate small vessel abnormalities in patients with cognitive impairment, we compared retinal microvascular alterations between patients with cognitive impairment related to Alzheimer's disease (ADCI) and those with subcortical vascular cognitive impairment (SVCI). METHODS We prospectively recruited 29 amyloid-positive ADCI patients, 28 amyloid-negative SVCI patients that were confirmed by 11C-PiB-PET scan and 34 individuals with normal cognition (NC). The three groups were compared in terms of retinal vascular variables (retinal fractal dimension, vascular caliber, tortuosity and branching angle) by using a semi-automated, computer-assisted analysis of digital fundus photographs. We also investigated the relationship between retinal variables and white matter hyperintensities (WMH) on MRI. RESULTS Compared to NC individuals, the SVCI patients had smaller total and arteriolar fractal dimensions, whereas there was no significant difference of fractal dimension between ADCI and NC. Other retinal variables did not differ among the three groups. A significant correlation existed between fractal dimension and WMH volume. CONCLUSIONS Retinal microvascular alterations, especially retinal fractal dimension, may be useful markers that reflect cerebral microvascular changes in patients with SVCI as opposed to ADCI, who had no definite difference in retinal variables compared to the NC group.
Collapse
Affiliation(s)
- Na-Yeon Jung
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine and Research Institute for Convergence of Biomedical Science and Technology, Yangsan, Republic of Korea; Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Jong Chul Han
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yi Ting Ong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Carol Yim-Lui Cheung
- Department of Ophthalmology & Visual Sciences, Chinese University of Hong Kong, Hong Kong
| | | | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Yeo Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea; Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Juyoun Lee
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea; Department of Neurology, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Jin San Lee
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea; Department of Neurology, Kyung Hee University Hospital, Seoul, Republic of Korea
| | - Young Kyoung Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Changwon Kee
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyung Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun-Joo Kim
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan, Republic of Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
87
|
Recent progress in metal–organic frameworks for precaution and diagnosis of Alzheimer’s disease. Polyhedron 2018. [DOI: 10.1016/j.poly.2018.06.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
88
|
Ameen-Ali KE, Wharton SB, Simpson JE, Heath PR, Sharp P, Berwick J. Review: Neuropathology and behavioural features of transgenic murine models of Alzheimer's disease. Neuropathol Appl Neurobiol 2018; 43:553-570. [PMID: 28880417 DOI: 10.1111/nan.12440] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/04/2017] [Indexed: 12/11/2022]
Abstract
Our understanding of the underlying biology of Alzheimer's disease (AD) has been steadily progressing; however, this is yet to translate into a successful treatment in humans. The use of transgenic mouse models has helped to develop our understanding of AD, not only in terms of disease pathology, but also with the associated cognitive impairments typical of AD. Plaques and neurofibrillary tangles are often among the last pathological changes in AD mouse models, after neuronal loss and gliosis. There is a general consensus that successful treatments need to be applied before the onset of these pathologies and associated cognitive symptoms. This review discusses the different types of AD mouse models in terms of the temporal progression of the disease, how well they replicate the pathological changes seen in human AD and their cognitive defects. We provide a critical assessment of the behavioural tests used with AD mice to assess cognitive changes and decline, and discuss how successfully they correlate with cognitive impairments in humans with AD. This information is an important tool for AD researchers when deciding on appropriate mouse models, and when selecting measures to assess behavioural and cognitive change.
Collapse
Affiliation(s)
- K E Ameen-Ali
- Department of Psychology, University of Sheffield, Sheffield, UK
| | - S B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - J E Simpson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - P R Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - P Sharp
- Department of Psychology, University of Sheffield, Sheffield, UK
| | - J Berwick
- Department of Psychology, University of Sheffield, Sheffield, UK
| |
Collapse
|
89
|
Kaisar MA, Sivandzade F, Bhalerao A, Cucullo L. Conventional and electronic cigarettes dysregulate the expression of iron transporters and detoxifying enzymes at the brain vascular endothelium: In vivo evidence of a gender-specific cellular response to chronic cigarette smoke exposure. Neurosci Lett 2018; 682:1-9. [PMID: 29879439 DOI: 10.1016/j.neulet.2018.05.045] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/17/2018] [Accepted: 05/29/2018] [Indexed: 02/06/2023]
Abstract
It is well established that tobacco smoking is associated with vascular endothelial dysfunction in a causative and dose dependent manner primarily related to the tobacco smoke (TS) content of reactive oxygen species (ROS), nicotine, and oxidative stress (OS) -driven inflammation. Preclinical studies have also shown that nicotine (the principal e-liquid's ingredient used in e-cigarettes (e-Cigs) can also cause OS, exacerbation of cerebral ischemia and secondary brain injury. Likewise, chronic e-Cig vaping could be prodromal to vascular endothelial dysfunctions. Herein, we provide direct evidence that similarly to TS, e-Cig promotes mitochondrial depolarization in primary brain vascular endothelial cells as well as the vascular endothelial cell line bEnd3. In addition, both TS and e-Cig exposure upregulated the transmembrane iron exporter Slc40a1 (crucial to maintain cellular iron and redox homeostasis) and that of porphyrin importer Abcb6 (linked to accelerated atherosclerosis). We then investigated in vivo whether gender plays a role in how chronic TS affect vascular endothelial functions. Our results clearly show chronic TS exposure differentially impacts the expression levels of Phase-II enzymes as well as the iron transporters previously investigated in vitro. Although the physiological implications of the gender-specific differential responses to TS are not fully clear, they do demonstrate that gender is a risk factor that needs to be investigated when assessing the potential impact of chronic smoking and perhaps e-Cig vaping.
Collapse
Affiliation(s)
- Mohammad A Kaisar
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| | - Farzane Sivandzade
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| | - Aditya Bhalerao
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| | - Luca Cucullo
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Center for Blood Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| |
Collapse
|
90
|
Goshgarian C, Gorelick PB. Perspectives on the relation of blood pressure and cognition in the elderly. Trends Cardiovasc Med 2018; 29:12-18. [PMID: 29891330 DOI: 10.1016/j.tcm.2018.05.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/26/2018] [Accepted: 05/27/2018] [Indexed: 11/19/2022]
Abstract
The relationship of blood pressure to cognition in the elderly is a poorly understood topic. Many questions exist such as does treatment of hypertension prevent cognitive decline, the optimal timing of intervention and selecting the appropriate agent. In this review we will explore recent epidemiologic data and clinical trials addressing hypertension and cognition, review pathophysiology of chronic hypertension and effects of brain function, discuss the timing of intervention and finally review opportunities for future research.
Collapse
Affiliation(s)
- Christopher Goshgarian
- Vascular Neurology Program, Mercy Health Hauenstein Neurosciences, 220 Cherry Street SE Room H 3037, Grand Rapids, MI 49503, United States.
| | - Philip B Gorelick
- Vascular Neurology Program, Mercy Health Hauenstein Neurosciences, 220 Cherry Street SE Room H 3037, Grand Rapids, MI 49503, United States; Translational Science and Molecular Medicine, Michigan State University College of Human Medicine, MI United States.
| |
Collapse
|
91
|
Lattanzi S, Brigo F, Vernieri F, Silvestrini M. Visit-to-visit variability in blood pressure and Alzheimer's disease. J Clin Hypertens (Greenwich) 2018; 20:918-924. [PMID: 29693801 DOI: 10.1111/jch.13290] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/07/2018] [Accepted: 03/11/2018] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease is the most common type of dementia and one of the leading sources of disability and dependency in the elderly. Given the limited treatment options, understanding the role of modifiable risk factors implied in the disease pathogenesis is a worthwhile endeavor to limit its global burden. Recently, the variability of blood pressure has been suggested to be a significant determinant of brain alterations and a potential therapeutic target. The aim of this article is to review the clinical evidence on the association between visit-to-visit blood pressure variability and Alzheimer's disease, highlight the underlying mechanisms, and suggest future implications.
Collapse
Affiliation(s)
- Simona Lattanzi
- Neurological Clinic, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Francesco Brigo
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Verona, Italy.,Division of Neurology, "Franz Tappeiner" Hospital, Merano, BZ, Italy
| | | | - Mauro Silvestrini
- Neurological Clinic, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| |
Collapse
|
92
|
Nagai M, Dote K, Kato M, Sasaki S, Oda N, Kagawa E, Nakano Y, Yamane A, Higashihara T, Miyauchi S, Tsuchiya A. Visit-to-Visit Blood Pressure Variability and Alzheimer's Disease: Links and Risks. J Alzheimers Dis 2018; 59:515-526. [PMID: 28598842 DOI: 10.3233/jad-161172] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
While hypertension has been shown to be a risk factor for vascular dementia, several studies have also demonstrated that hypertension also increases the risk of Alzheimer's disease (AD). Although the relationship between visit-to-visit blood pressure variability (VVV) and cognitive impairment, including AD, have been provided, the mechanisms remain poorly understood. This review paper focuses on the relationship of VVV with AD and summarizes the pathophysiology underlying that relationship, which appears to be mediated by arterial stiffness.
Collapse
Affiliation(s)
- Michiaki Nagai
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| | - Keigo Dote
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| | - Masaya Kato
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| | - Shota Sasaki
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| | - Noboru Oda
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| | - Eisuke Kagawa
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| | - Yoshinori Nakano
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| | - Aya Yamane
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| | | | - Shunsuke Miyauchi
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| | - Akane Tsuchiya
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| |
Collapse
|
93
|
Sun J, Zhou H, Bai F, Zhang Z, Ren Q. Remyelination: A Potential Therapeutic Strategy for Alzheimer's Disease? J Alzheimers Dis 2018; 58:597-612. [PMID: 28453483 DOI: 10.3233/jad-170036] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myelin is a lipid-rich multilamellar membrane that wraps around long segments of neuronal axons and it increases the conduction of action potentials, transports the necessary trophic support to the neuronal axons, and reduces the energy consumed by the neuronal axons. Together with axons, myelin is a prerequisite for the higher functions of the central nervous system and complex forms of network integration. Myelin impairments have been suggested to lead to neuronal dysfunction and cognitive decline. Accumulating evidence, including brain imaging and postmortem and genetic association studies, has implicated myelin impairments in Alzheimer's disease (AD). Increasing data link myelin impairments with amyloid-β (Aβ) plaques and tau hyperphosphorylation, which are both present in patients with AD. Moreover, aging and apolipoprotein E (ApoE) may be involved in the myelin impairments observed in patients with AD. Decreased neuronal activity, increased Aβ levels, and inflammation further damage myelin in patients with AD. Furthermore, treatments that promote myelination contribute to the recovery of neuronal function and improve cognition. Therefore, strategies targeting myelin impairment may provide therapeutic opportunities for patients with AD.
Collapse
|
94
|
Tasci I, Safer U, Naharci MI, Gezer M, Demir O, Bozoglu E, Doruk H. Undetected Peripheral Arterial Disease Among Older Adults With Alzheimer's Disease and Other Dementias. Am J Alzheimers Dis Other Demen 2018; 33:5-11. [PMID: 28786294 PMCID: PMC10852521 DOI: 10.1177/1533317517724000] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2024]
Abstract
BACKGROUND Peripheral arterial disease (PAD) was reported to increase the risk of dementia(s) even more than stroke. We assessed the prevalence of PAD in a group with definite diagnosis of dementia. METHODS Patients aged 65 years or older with Alzheimer's disease (AD), vascular dementia (VaD), or AD-VaD were enrolled (n = 162, mean age: 78.87 [6.05] years). An age- and gender-matched control group was also included (n = 190). Peripheral arterial disease was diagnosed by the ankle-brachial index. RESULTS Frequency of PAD among patients with and without dementia was 35.2% and 16.3%, respectively ( P < .001), being similar among different types of dementia. After adjustment for covariates, dementia (odds ratio: 2.41, 95% confidence interval: 1.34-4.32; P = .003) was among the predictors of PAD diagnosis along with older age, female gender, and diabetes. CONCLUSIONS The prevalence of PAD was more than double in patients with dementia, with no difference among AD, VaD, and AD-VaD types.
Collapse
Affiliation(s)
- Ilker Tasci
- Department of Internal Medicine, Gulhane Medical School, Health Sciences University, Ankara, Turkey
- Internal Medicine Clinic, Gulhane Teaching and Research Hospital, Turkish Ministry of Health, Ankara, Turkey
| | - Umut Safer
- Internal Medicine Clinic, Sultan Abulhamid Teaching and Research Hospital, Turkish Ministry of Health, Istanbul, Turkey
| | - Mehmet Ilkin Naharci
- Department of Internal Medicine, Gulhane Medical School, Health Sciences University, Ankara, Turkey
- Internal Medicine Clinic, Gulhane Teaching and Research Hospital, Turkish Ministry of Health, Ankara, Turkey
| | - Mustafa Gezer
- Department of Internal Medicine, Gulhane Medical School, Health Sciences University, Ankara, Turkey
- Internal Medicine Clinic, Gulhane Teaching and Research Hospital, Turkish Ministry of Health, Ankara, Turkey
| | - Orhan Demir
- Department of Internal Medicine, Gulhane Medical School, Health Sciences University, Ankara, Turkey
- Internal Medicine Clinic, Gulhane Teaching and Research Hospital, Turkish Ministry of Health, Ankara, Turkey
| | - Ergun Bozoglu
- Department of Internal Medicine, Gulhane Medical School, Health Sciences University, Ankara, Turkey
- Internal Medicine Clinic, Gulhane Teaching and Research Hospital, Turkish Ministry of Health, Ankara, Turkey
| | - Huseyin Doruk
- Department of Internal Medicine, Gulhane Medical School, Health Sciences University, Ankara, Turkey
- Internal Medicine Clinic, Gulhane Teaching and Research Hospital, Turkish Ministry of Health, Ankara, Turkey
| |
Collapse
|
95
|
Traumatic Brain Injury and Alzheimer's Disease: The Cerebrovascular Link. EBioMedicine 2018; 28:21-30. [PMID: 29396300 PMCID: PMC5835563 DOI: 10.1016/j.ebiom.2018.01.021] [Citation(s) in RCA: 264] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/19/2018] [Accepted: 01/19/2018] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) and Alzheimer's disease (AD) are devastating neurological disorders, whose complex relationship is not completely understood. Cerebrovascular pathology, a key element in both conditions, could represent a mechanistic link between Aβ/tau deposition after TBI and the development of post concussive syndrome, dementia and chronic traumatic encephalopathy (CTE). In addition to debilitating acute effects, TBI-induced neurovascular injuries accelerate amyloid β (Aβ) production and perivascular accumulation, arterial stiffness, tau hyperphosphorylation and tau/Aβ-induced blood brain barrier damage, giving rise to a deleterious feed-forward loop. We postulate that TBI can initiate cerebrovascular pathology, which is causally involved in the development of multiple forms of neurodegeneration including AD-like dementias. In this review, we will explore how novel biomarkers, animal and human studies with a focus on cerebrovascular dysfunction are contributing to the understanding of the consequences of TBI on the development of AD-like pathology. Cerebrovascular dysfunction (CVD) is emerging as a key element in the development of neurodegeneration after TBI. We propose that TBI initiates CVD, accelerating Aβ/tau deposition and leading to neurodegeneration and dementias. Clarifying this connection will support the development of novel biomarkers and therapeutic approaches for both TBI and AD.
Collapse
|
96
|
Jung YY, Kim KC, Park MH, Seo Y, Park H, Park MH, Chang J, Hwang DY, Han SB, Kim S, Son DJ, Hong JT. Atherosclerosis is exacerbated by chitinase-3-like-1 in amyloid precursor protein transgenic mice. Am J Cancer Res 2018; 8:749-766. [PMID: 29344304 PMCID: PMC5771091 DOI: 10.7150/thno.20183] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 11/09/2017] [Indexed: 02/06/2023] Open
Abstract
Although the important role of amyloid precursor protein (APP) in vascular diseases associated with Alzheimer's disease (AD) has been demonstrated, the underlying molecular mechanisms and physiological consequences are unclear. We aimed to evaluate vascular inflammation and atherosclerosis in Swedish mutant of human APP transgenic (APPsw-Tg) and ApoE-/-/APPsw-Tg mice. We also aimed to explore the mechanisms underlying any changes observed in these mice compared with non-Tg controls. Methods: The transgenic and non-Tg mouse strains were subjected to partial ligation of the left carotid artery to induce atherosclerotic changes, which were measured using histological approaches, immunohistochemistry, quantitative polymerase chain reaction, and gene expression microarrays. Results: Our results showed increased vascular inflammation, arterial wall thickness, and atherosclerosis in APPsw-Tg and ApoE-/-/APPsw-Tg mice. We further found that the expression of chitinase-3-like-1 (Chi3l1) is increased in the APPsw-Tg mouse artery and Chi3l1 mediates endothelial cell (EC) inflammation and vascular smooth muscle cell (VSMC) activation, which in turn exacerbates atherosclerosis. In addition, using two publicly available microarray datasets from the dorsolateral prefrontal cortex of people with AD and unaffected controls as well as inflamed human umbilical vein endothelial cells, we found that Chi3l1 and associated inflammatory gene were significantly associated with AD, evaluated by co-expression network analysis and functional annotation. Knockdown of Chi3l1 in the arterial endothelium in vivo suppressed the development of atherosclerosis. We also show that microRNA 342-3p (miR-342-3p) inhibits EC inflammation and VSMC activation through directly targeting Chi3l1, and that APPsw increased Chi3l1 expression by reducing miR-342-3p expression in the arterial endothelium, promoting atherosclerosis. Conclusion: Our findings suggest that targeting Chi3l1 might provide new diagnostic and therapeutic strategies for vascular diseases in patients with AD.
Collapse
|
97
|
Wen YR, Yang JH, Wang X, Yao ZB. Induced dural lymphangiogenesis facilities soluble amyloid-beta clearance from brain in a transgenic mouse model of Alzheimer's disease. Neural Regen Res 2018; 13:709-716. [PMID: 29722325 PMCID: PMC5950683 DOI: 10.4103/1673-5374.230299] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Impaired amyloid-β clearance from the brain is a core pathological event in Alzheimer's disease. The therapeutic effect of current pharmacotherapies is unsatisfactory, and some treatments cause severe side effects. The meningeal lymphatic vessels might be a new route for amyloid-β clearance. This study investigated whether promoting dural lymphangiogenesis facilitated the clearance of amyloid-β from the brain.First, human lymphatic endothelial cells were treated with 100 ng/mL recombinant human vascular endothelial growth factor-C (rhVEGF-C) protein. Light microscopy verified that rhVEGF-C, a specific ligand for vascular endothelial growth factor receptor-3 (VEGFR-3), significantly promoted tube formation of human lymphatic endothelial cells in vitro. In an in vivo study, 200 μg/mL rhVEGF-C was injected into the cisterna magna of APP/PS1 transgenic mice, once every 2 days, four times in total. Immunofluorescence staining demonstrated high levels of dural lymphangiogenesis in Alzheimer's disease mice. One week after rhVEGF-C administration, enzyme-linked immunosorbent assay results showed that levels of soluble amyloid-β were decreased in cerebrospinal fluid and brain. The Morris water maze test demonstrated that spatial cognition was restored. These results indicate that the upregulation of dural lymphangiogenesis facilities amyloid-β clearance from the brain of APP/PS1 mice, suggesting the potential of the VEGF-C/VEGFR-3 signaling pathway as a therapeutic target for Alzheimer's disease.
Collapse
Affiliation(s)
- Ya-Ru Wen
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jun-Hua Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiao Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhi-Bin Yao
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
98
|
Polis B, Samson AO. Arginase as a Potential Target in the Treatment of Alzheimer’s Disease. ACTA ACUST UNITED AC 2018. [DOI: 10.4236/aad.2018.74009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
99
|
Sun BL, Wang LH, Yang T, Sun JY, Mao LL, Yang MF, Yuan H, Colvin RA, Yang XY. Lymphatic drainage system of the brain: A novel target for intervention of neurological diseases. Prog Neurobiol 2017; 163-164:118-143. [PMID: 28903061 DOI: 10.1016/j.pneurobio.2017.08.007] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 08/11/2017] [Accepted: 08/31/2017] [Indexed: 12/20/2022]
Abstract
The belief that the vertebrate brain functions normally without classical lymphatic drainage vessels has been held for many decades. On the contrary, new findings show that functional lymphatic drainage does exist in the brain. The brain lymphatic drainage system is composed of basement membrane-based perivascular pathway, a brain-wide glymphatic pathway, and cerebrospinal fluid (CSF) drainage routes including sinus-associated meningeal lymphatic vessels and olfactory/cervical lymphatic routes. The brain lymphatic systems function physiological as a route of drainage for interstitial fluid (ISF) from brain parenchyma to nearby lymph nodes. Brain lymphatic drainage helps maintain water and ion balance of the ISF, waste clearance, and reabsorption of macromolecular solutes. A second physiological function includes communication with the immune system modulating immune surveillance and responses of the brain. These physiological functions are influenced by aging, genetic phenotypes, sleep-wake cycle, and body posture. The impairment and dysfunction of the brain lymphatic system has crucial roles in age-related changes of brain function and the pathogenesis of neurovascular, neurodegenerative, and neuroinflammatory diseases, as well as brain injury and tumors. In this review, we summarize the key component elements (regions, cells, and water transporters) of the brain lymphatic system and their regulators as potential therapeutic targets in the treatment of neurologic diseases and their resulting complications. Finally, we highlight the clinical importance of ependymal route-based targeted gene therapy and intranasal drug administration in the brain by taking advantage of the unique role played by brain lymphatic pathways in the regulation of CSF flow and ISF/CSF exchange.
Collapse
Affiliation(s)
- Bao-Liang Sun
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China.
| | - Li-Hua Wang
- Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, China
| | - Tuo Yang
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jing-Yi Sun
- Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Gangwon 220-701, Republic of Korea
| | - Lei-Lei Mao
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China
| | - Ming-Feng Yang
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China
| | - Hui Yuan
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China
| | - Robert A Colvin
- Department of Biological Sciences, Interdisciplinary Graduate Program in Molecular and Cellular Biology, Neuroscience Program, Ohio University, Athens, OH 45701, USA
| | - Xiao-Yi Yang
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China.
| |
Collapse
|
100
|
Simino J, Wang Z, Bressler J, Chouraki V, Yang Q, Younkin SG, Seshadri S, Fornage M, Boerwinkle E, Mosley TH. Whole exome sequence-based association analyses of plasma amyloid-β in African and European Americans; the Atherosclerosis Risk in Communities-Neurocognitive Study. PLoS One 2017; 12:e0180046. [PMID: 28704393 PMCID: PMC5509141 DOI: 10.1371/journal.pone.0180046] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/08/2017] [Indexed: 12/30/2022] Open
Abstract
Objective We performed single-variant and gene-based association analyses of plasma amyloid-β (aβ) concentrations using whole exome sequence from 1,414 African and European Americans. Our goal was to identify genes that influence plasma aβ42 concentrations and aβ42:aβ40 ratios in late middle age (mean = 59 years), old age (mean = 77 years), or change over time (mean = 18 years). Methods Plasma aβ measures were linearly regressed onto age, gender, APOE ε4 carrier status, and time elapsed between visits (fold-changes only) separately by race. Following inverse normal transformation of the residuals, seqMeta was used to conduct race-specific single-variant and gene-based association tests while adjusting for population structure. Linear regression models were fit on autosomal variants with minor allele frequencies (MAF)≥1%. T5 burden and Sequence Kernel Association (SKAT) gene-based tests assessed functional variants with MAF≤5%. Cross-race fixed effects meta-analyses were Bonferroni-corrected for the number of variants or genes tested. Results Seven genes were associated with aβ in late middle age or change over time; no associations were identified in old age. Single variants in KLKB1 (rs3733402; p = 4.33x10-10) and F12 (rs1801020; p = 3.89x10-8) were significantly associated with midlife aβ42 levels through cross-race meta-analysis; the KLKB1 variant replicated internally using 1,014 additional participants with exome chip. ITPRIP, PLIN2, and TSPAN18 were associated with the midlife aβ42:aβ40 ratio via the T5 test; TSPAN18 was significant via the cross-race meta-analysis, whereas ITPRIP and PLIN2 were European American-specific. NCOA1 and NT5C3B were associated with the midlife aβ42:aβ40 ratio and the fold-change in aβ42, respectively, via SKAT in African Americans. No associations replicated externally (N = 725). Conclusion We discovered age-dependent genetic effects, established associations between vascular-related genes (KLKB1, F12, PLIN2) and midlife plasma aβ levels, and identified a plausible Alzheimer’s Disease candidate gene (ITPRIP) influencing cell death. Plasma aβ concentrations may have dynamic biological determinants across the lifespan; plasma aβ study designs or analyses must consider age.
Collapse
Affiliation(s)
- Jeannette Simino
- Gertrude C. Ford MIND Center, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- Department of Data Science, John D. Bower School of Population Health, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- * E-mail:
| | - Zhiying Wang
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Jan Bressler
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Vincent Chouraki
- Lille University, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk factors and molecular determinants of aging-related diseases; Lille, France
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- The National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Steven G. Younkin
- Department of Neuroscience, Mayo Clinic College of Medicine, Mayo Clinic Jacksonville, Jacksonville, Florida, United States of America
| | - Sudha Seshadri
- The National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center, Houston, Texas, United States of America
- The Brown Foundation Institute of Molecular Medicine, Research Center for Human Genetics, The University of Texas Health Science Center, Houston, Texas, United States of America
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center, Houston, Texas, United States of America
- The Brown Foundation Institute of Molecular Medicine, Research Center for Human Genetics, The University of Texas Health Science Center, Houston, Texas, United States of America
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Thomas H. Mosley
- Gertrude C. Ford MIND Center, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- Department of Medicine, University of Mississippi Medical Center, Jackson, Massachusetts, United States of America
| |
Collapse
|