1
|
Hayat M, Syed RA, Qaiser H, Uzair M, Al-Regaiey K, Khallaf R, Albassam LAM, Kaleem I, Wang X, Wang R, Bhatti MS, Bashir S. Decoding molecular mechanisms: brain aging and Alzheimer's disease. Neural Regen Res 2025; 20:2279-2299. [PMID: 39104174 PMCID: PMC11759015 DOI: 10.4103/nrr.nrr-d-23-01403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 01/23/2024] [Accepted: 07/04/2024] [Indexed: 08/07/2024] Open
Abstract
The complex morphological, anatomical, physiological, and chemical mechanisms within the aging brain have been the hot topic of research for centuries. The aging process alters the brain structure that affects functions and cognitions, but the worsening of such processes contributes to the pathogenesis of neurodegenerative disorders, such as Alzheimer's disease. Beyond these observable, mild morphological shifts, significant functional modifications in neurotransmission and neuronal activity critically influence the aging brain. Understanding these changes is important for maintaining cognitive health, especially given the increasing prevalence of age-related conditions that affect cognition. This review aims to explore the age-induced changes in brain plasticity and molecular processes, differentiating normal aging from the pathogenesis of Alzheimer's disease, thereby providing insights into predicting the risk of dementia, particularly Alzheimer's disease.
Collapse
Affiliation(s)
- Mahnoor Hayat
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Rafay Ali Syed
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Hammad Qaiser
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad (IIUI), Islamabad, Pakistan
| | - Mohammad Uzair
- Department of Bioengineering, King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
| | - Khalid Al-Regaiey
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Roaa Khallaf
- Department of Neurology, Neuroscience Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | | | - Imdad Kaleem
- Department of Biosciences, Commission on Science and Technology for Sustainable Development in the South (COMSATS University), Islamabad, Pakistan
| | - Xueyi Wang
- Department of Psychiatry, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
- Mental Health Institute of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Ran Wang
- Department of Psychiatry, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
- Mental Health Institute of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Mehwish S. Bhatti
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Saudi Arabia
| |
Collapse
|
2
|
Luo Q, Li F, Liu X, Yan T, Yang L, Zhu W, Zheng H, Li Y, Tu J, Zhu X. Puerarin mitigates cognitive decline and white matter injury via CD36-Mediated microglial phagocytosis in chronic cerebral hypoperfusion. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156396. [PMID: 39827816 DOI: 10.1016/j.phymed.2025.156396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/24/2024] [Accepted: 01/12/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) contributes significantly to white matter injury (WMI) and cognitive impairment, often leading to vascular dementia (VaD). Inefficient clearance of myelin debris by microglia impedes white matter repair, making microglia-mediated myelin clearance a promising therapeutic strategy for WMI. Puerarin (Pu), an isoflavonoid monomer from Pueraria lobata, is known for its neuroprotective, anti-inflammatory, and immunoregulatory properties. However, its effects and underlying mechanisms in counteracting CCH-induced damage remain unclear. In this study, we aimed to investigate the therapeutic effects and underlying mechanisms of puerarin in a CCH mouse model. METHODS Right unilateral common carotid artery occlusion (rUCCAO) was used to model CCH in C57BL/6J mice. Puerarin (400 mg/kg/day) was administered intraperitoneally for 10 consecutive days starting immediately post-surgery. Cognitive function was assessed by the Morris Water Maze (MWM) test. WMI, remyelination, neuroinflammation, and microglial phagocytosis were evaluated by western blotting, immunofluorescence staining, RT-PCR, or flow cytometry both in vivo and in vitro. RESULTS Puerarin treatment significantly improved cognitive performance and mitigated WMI in rUCCAO mice. These effects were associated with enhanced microglial phagocytosis and remyelination, reduced neuroinflammation, and increased CD36 expression. Additionally, puerarin also increased the levels of IL-10 and phosphorylated STAT3 (p-STAT3) in brain tissues. Notably, IL-10 neutralization reversed these benefits effects by reducing microglial myelin debris uptake, downregulating STAT3 phosphorylation and CD36 expression. CONCLUSIONS Our findings demonstrate that puerarin has significant therapeutic potential in treating CCH-related cognitive impairments and WMI by modulating CD36-mediated microglial myelin clearance through the IL-10/STAT3 pathway. However, our study was reliant on preclinical animal models, further studies are needed to explore applicability in human subjects.
Collapse
Affiliation(s)
- Qinghua Luo
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, China; Jiangxi Provincial Key Laboratory of Nervous System Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, China; JXHC Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, Jiangxi, China; Jiangxi Provincial Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, China; Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330000, China
| | - Fang Li
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330000, China; Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330000, China
| | - Xu Liu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, China; JXHC Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, Jiangxi, China; Jiangxi Provincial Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, China
| | - Tengfeng Yan
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, China; Jiangxi Provincial Key Laboratory of Nervous System Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, China; JXHC Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, Jiangxi, China; Jiangxi Provincial Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, China; Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330000, China
| | - Li Yang
- Jiangxi Provincial Key Laboratory of Nervous System Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, China; JXHC Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, Jiangxi, China; Jiangxi Provincial Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Wenping Zhu
- Jiangxi Provincial Key Laboratory of Nervous System Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, China; JXHC Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, Jiangxi, China; Jiangxi Provincial Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Heqing Zheng
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330000, China; Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330000, China
| | - Yan Li
- Jiangxi Provincial Key Laboratory of Nervous System Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, China; JXHC Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, Jiangxi, China; Jiangxi Provincial Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Jianglong Tu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, China; JXHC Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, Jiangxi, China; Jiangxi Provincial Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, China.
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, China; Jiangxi Provincial Key Laboratory of Nervous System Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, China; JXHC Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, Jiangxi, China; Jiangxi Provincial Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330000, China.
| |
Collapse
|
3
|
Ohnishi H, Matsuoka K, Takahashi M, Yoshikawa H, Minami A, Ueda K, Fujimoto Y, Kiuchi K, Ochi T, Miyasaka T, Tanaka T, Matsumoto R, Makinodan M, Okada T. Associations of demyelination in the right middle temporal gyrus and right praecuneus with visuospatial cognitive dysfunction in Alzheimer's disease. Psychogeriatrics 2025; 25:e13223. [PMID: 39581748 DOI: 10.1111/psyg.13223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is associated with impairments in not only memory but also visuospatial cognitive function. Despite its adverse effects on the quality of life, patients with early-stage AD are often neglected. Emerging evidence suggests that patients with AD exhibit increased vulnerability of myelin, a crucial component for neuronal conduction and survival. To test our hypothesis that myelin damage was associated with cognitive deficits in AD, we examined correlations of myelin integrity, quantified by T1-weighted/T2-weighted (T1w/T2w) ratios, with visuospatial cognitive abilities and compared them between patients with AD and cognitively normal (CN) individuals. METHODS Fifty-seven patients with AD and 22 CN subjects were enrolled in this study. To assess subjects' visuo-constructive abilities, we employed the Rey-Osterrieth Complex Figure Copy Test (ROCFT-c) paired with analysis of T1- and T2-weighted magnetic resonance imaging brain images. Voxel-based associations between T1w/T2w ratios and ROCFT-c scores in the AD group were assessed, controlling for age and handedness (voxel threshold uncorrected P < 0.001, cluster threshold uncorrected P < 0.05). Additionally, we compared the T1w/T2w ratios of these identified brain regions between the AD and CN groups. RESULTS The voxel-based analysis demonstrated positive correlations between T1w/T2w ratios and ROCFT-c scores in the right middle temporal gyrus and right praecuneus in patients with AD who exhibited significantly lower T1w/T2w ratios in the right middle temporal gyrus (P = 0.038) and a trend toward lower T1w/T2w ratios in the right praecuneus (P = 0.055). CONCLUSIONS Our results demonstrated a strong association between reduced myelin integrity in the right middle temporal gyrus and right praecuneus and visuospatial cognitive dysfunction in patients with AD. These findings are believed to shed light on the neural basis of visuospatial processing in patients with AD, underlining the necessity for developing objective biomarkers for assessing patients' visuospatial cognitive function.
Collapse
Affiliation(s)
- Hiroki Ohnishi
- Department of Psychiatry, Nara Medical University, Nara, Japan
| | - Kiwamu Matsuoka
- Department of Psychiatry, Nara Medical University, Nara, Japan
| | | | | | - Akihiro Minami
- Department of Psychiatry, Nara Medical University, Nara, Japan
| | - Kazuya Ueda
- Department of Psychiatry, Nara Medical University, Nara, Japan
| | - Yuka Fujimoto
- Department of Psychiatry, Nara Medical University, Nara, Japan
| | - Kuniaki Kiuchi
- Department of Psychiatry, Higashiosaka City Medical Centre, Osaka, Japan
| | - Tomoko Ochi
- Department of Radiology, Nara Medical University, Nara, Japan
| | | | | | - Ryohei Matsumoto
- Department of Psychiatry, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Takashi Okada
- Department of Psychiatry, Nara Medical University, Nara, Japan
| |
Collapse
|
4
|
Peterson A, Sathe A, Zaras D, Yang Y, Durant A, Deters KD, Shashikumar N, Pechman KR, Kim ME, Gao C, Mohd Khairi N, Li Z, Yao T, Huo Y, Dumitrescu L, Gifford KA, Wilson JE, Cambronero FE, Risacher SL, Beason-Held LL, An Y, Arfanakis K, Erus G, Davatzikos C, Tosun D, Toga AW, Thompson PM, Mormino EC, Habes M, Wang D, Zhang P, Schilling K, Albert M, Kukull W, Biber SA, Landman BA, Johnson SC, Schneider J, Barnes LL, Bennett DA, Jefferson AL, Resnick SM, Saykin AJ, Hohman TJ, Archer DB. Sex and APOE ε4 allele differences in longitudinal white matter microstructure in multiple cohorts of aging and Alzheimer's disease. Alzheimers Dement 2024. [PMID: 39711105 DOI: 10.1002/alz.14343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/08/2024] [Accepted: 09/27/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION The effects of sex and apolipoprotein E (APOE)-Alzheimer's disease (AD) risk factors-on white matter microstructure are not well characterized. METHODS Diffusion magnetic resonance imaging data from nine well-established longitudinal cohorts of aging were free water (FW)-corrected and harmonized. This dataset included 4741 participants (age = 73.06 ± 9.75) with 9671 imaging sessions over time. FW and FW-corrected fractional anisotropy (FAFWcorr) were used to assess differences in white matter microstructure by sex and APOE ε4 carrier status. RESULTS Sex differences in FAFWcorr in projection tracts and APOE ε4 differences in FW limbic and occipital transcallosal tracts were most pronounced. DISCUSSION There are prominent differences in white matter microstructure by sex and APOE ε4 carrier status. This work adds to our understanding of disparities in AD. Additional work to understand the etiology of these differences is warranted. HIGHLIGHTS Sex and apolipoprotein E (APOE) ε4 carrier status relate to white matter microstructural integrity. Females generally have lower free water-corrected fractional anisotropy compared to males. APOE ε4 carriers tended to have higher free water than non-carriers.
Collapse
Affiliation(s)
- Amalia Peterson
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Aditi Sathe
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Dimitrios Zaras
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Yisu Yang
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alaina Durant
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kacie D Deters
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Niranjana Shashikumar
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kimberly R Pechman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Michael E Kim
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, USA
| | - Chenyu Gao
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Nazirah Mohd Khairi
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Zhiyuan Li
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Tianyuan Yao
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, USA
| | - Yuankai Huo
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, USA
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Katherine A Gifford
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jo Ellen Wilson
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Psychiatry and Behavioral Sciences, Center for Cognitive Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Veteran's Affairs, Geriatric Research, Education and Clinical Center, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Francis E Cambronero
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Shannon L Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Lori L Beason-Held
- Laboratory for Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Yang An
- Laboratory for Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Konstantinos Arfanakis
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, USA
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
- Department of Diagnostic Radiology, Rush University Medical Center, Chicago, Illinois, USA
| | - Guray Erus
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christos Davatzikos
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Arthur W Toga
- Laboratory of Neuroimaging, USC Stevens Institute of Neuroimaging and Informatics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging and Informatics, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Mohamad Habes
- Neuroimage Analytics Laboratory and Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Di Wang
- Neuroimage Analytics Laboratory and Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Panpan Zhang
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kurt Schilling
- Department of Radiology & Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Walter Kukull
- National Alzheimer's Coordinating Center, University of Washington, Seattle, Washington, USA
| | - Sarah A Biber
- National Alzheimer's Coordinating Center, University of Washington, Seattle, Washington, USA
| | - Bennett A Landman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, USA
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Radiology & Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Julie Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Angela L Jefferson
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, USA
| | - Susan M Resnick
- Laboratory for Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Derek B Archer
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
5
|
Cai J, Rong H, Chen J, Deng Z, Chen S, Liao H, Pan D, Chen Y, Shi Z, Li Y, Li H, Xu Y, Tang Y. Association of immune-mediated diseases with the risk of dementia and brain structure in UK Biobank participants. Age Ageing 2024; 53:afae274. [PMID: 39714283 DOI: 10.1093/ageing/afae274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Immunity and inflammation may be essential to the pathogenesis of dementia. However, the association of immune-mediated diseases with the risk of incident dementia has not been well characterised. OBJECTIVES We aimed to investigate the prospective association of 27 immune-mediated diseases and incident dementia risk and to explore the underlying mechanisms driven by brain structures. METHODS We included 487 459 UK Biobank participants aged 37-73 years without dementia at enrolment. Immune-mediated diseases and dementia cases were ascertained according to the International Classification of Diseases codes. Time-varying Cox proportional hazards regression and general linear regression models were used to examine the association of immune-mediated disease with incident dementia risk and brain morphometric measures, respectively. RESULTS Over a median follow-up of 12.3 years, 1654 cases of incident dementia were documented in 86 243 patients with immune-mediated diseases. Overall, immune-mediated diseases were associated with a higher all-cause dementia risk (hazard ratio [HR], 1.24; 95% confidence interval, 1.17-1.32). Five out of 27 immune-mediated diseases were associated with an increased risk of dementia individually. Comorbidity of multiple immune-mediated diseases further increased the risk. Moreover, the immune-mediated disease was associated with smaller total surface areas of both left (β, -286.51; SE, 102.58; P = .014) and right hemispheres (β, -298.56; SE, 103.96; P = .016), greater white matter hyperintensities volume (β, 1.02; SE, 0.13; P < .001) and less healthy white matter microstructures. CONCLUSIONS Immune-mediated diseases were associated with an increased risk of incident dementia, and the association of those diseases with brain structural abnormalities might provide clues to the underlying mechanisms.
Collapse
Affiliation(s)
- Jinhua Cai
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Heng Rong
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jiongxue Chen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhenhong Deng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Sitai Chen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Huanquan Liao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of Neurology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Dong Pan
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Yanting Chen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhongshan Shi
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yi Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Honghong Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yongteng Xu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yamei Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
6
|
Asadollahi E, Trevisiol A, Saab AS, Looser ZJ, Dibaj P, Ebrahimi R, Kusch K, Ruhwedel T, Möbius W, Jahn O, Lee JY, Don AS, Khalil MA, Hiller K, Baes M, Weber B, Abel ED, Ballabio A, Popko B, Kassmann CM, Ehrenreich H, Hirrlinger J, Nave KA. Oligodendroglial fatty acid metabolism as a central nervous system energy reserve. Nat Neurosci 2024; 27:1934-1944. [PMID: 39251890 PMCID: PMC11452346 DOI: 10.1038/s41593-024-01749-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/05/2024] [Indexed: 09/11/2024]
Abstract
Brain function requires a constant supply of glucose. However, the brain has no known energy stores, except for glycogen granules in astrocytes. In the present study, we report that continuous oligodendroglial lipid metabolism provides an energy reserve in white matter tracts. In the isolated optic nerve from young adult mice of both sexes, oligodendrocytes survive glucose deprivation better than astrocytes. Under low glucose, both axonal ATP levels and action potentials become dependent on fatty acid β-oxidation. Importantly, ongoing oligodendroglial lipid degradation feeds rapidly into white matter energy metabolism. Although not supporting high-frequency spiking, fatty acid β-oxidation in mitochondria and oligodendroglial peroxisomes protects axons from conduction blocks when glucose is limiting. Disruption of the glucose transporter GLUT1 expression in oligodendrocytes of adult mice perturbs myelin homeostasis in vivo and causes gradual demyelination without behavioral signs. This further suggests that the imbalance of myelin synthesis and degradation can underlie myelin thinning in aging and disease.
Collapse
Affiliation(s)
- Ebrahim Asadollahi
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany.
| | - Andrea Trevisiol
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- University of Toronto, Sunnybrook Health Sciences Centre, Department of Physical Sciences, North York, Ontario, Canada
| | - Aiman S Saab
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Zoe J Looser
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Payam Dibaj
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- Center for Rare Diseases Göttingen, Department of Pediatrics and Pediatric Neurology, Georg August University Göttingen, Göttingen, Germany
| | - Reyhane Ebrahimi
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
| | - Kathrin Kusch
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- University of Göttingen Medical School, Institute for Auditory Neuroscience and Inner Ear Lab, Göttingen, Germany
| | - Torben Ruhwedel
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
| | - Wiebke Möbius
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
| | - Olaf Jahn
- Max Planck Institute for Multidisciplinary Sciences, Department of Molecular Neurobiology, Neuroproteomics Group, Göttingen, Germany
- University Medical Center Göttingen, Department of Psychiatry and Psychotherapy, Translational Neuroproteomics Group, Göttingen, Germany
| | - Jun Yup Lee
- School of Medical Sciences and Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Anthony S Don
- School of Medical Sciences and Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Michelle-Amirah Khalil
- Department for Bioinformatics and Biochemistry, Braunschweig Integrated Center of System Biology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Karsten Hiller
- Department for Bioinformatics and Biochemistry, Braunschweig Integrated Center of System Biology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Myriam Baes
- Lab of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Bruno Weber
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Brian Popko
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Celia M Kassmann
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
| | - Hannelore Ehrenreich
- Max Planck Institute for Multidisciplinary Sciences, Clinical Neuroscience, Göttingen, Germany
- Central Institute of Mental Health, Mannheim, Germany
| | - Johannes Hirrlinger
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Klaus-Armin Nave
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany.
| |
Collapse
|
7
|
Syed Nasser N, Venugopal VK, Veenstra C, Johansson P, Rajan S, Mahajan K, Naik S, Masand R, Yadav P, Khanduri S, Singhal S, Bhargava R, Kabra U, Gupta S, Saggar K, Varaprasad B, Aggrawal K, Rao A, K S M, Dakhole A, Kelkar A, Benjamin G, Sodani V, Goyal P, Mahajan H. Age-stratified Assessment of Brain Volumetric Segmentation on the Indian Population Using Quantitative Magnetic Resonance Imaging. Clin Neuroradiol 2024; 34:541-551. [PMID: 38253891 DOI: 10.1007/s00062-023-01374-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 12/16/2023] [Indexed: 01/24/2024]
Abstract
BACKGROUND AND PURPOSE Automated methods for quantifying brain tissue volumes have gained clinical interest for their objective assessment of neurological diseases. This study aimed to establish reference curves for brain volumes and fractions in the Indian population using Synthetic MRI (SyMRI), a quantitative imaging technique providing multiple contrast-weighted images through fast postprocessing. METHODS The study included a cohort of 314 healthy individuals aged 15-65 years from multiple hospitals/centers across India. The SyMRI-quantified brain volumes and fractions, including brain parenchymal fraction (BPF), gray matter fraction (GMF), white matter fraction (WMF), and myelin. RESULTS Normative age-stratified quantification curves were created based on the obtained data. The results showed significant differences in brain volumes between the sexes, but not after normalization by intracranial volume. CONCLUSION The findings provide normative data for the Indian population and can be used for comparative analysis of brain structure values. Furthermore, our data indicate that the use of fractions rather than absolute volumes in normative curves, such as BPF, GMF, and WMF, can mitigate sex and population differences as they account for individual differences in head size or brain volume.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Swati Naik
- Batra Hospital & Medical Research Centre, New Delhi, India
| | | | - Pratiksha Yadav
- Dr. D. Y. Patil Medical College, Hospital and Research Centre, Pune, India
| | | | | | | | | | | | - Kavita Saggar
- Dayanand Medical College & Hospital, Ludhiana, India
| | | | | | | | - Manoj K S
- Metro Scans and Laboratory, Thiruvananthapuram, India
| | | | | | | | | | | | | |
Collapse
|
8
|
Gao H, Liu X, Venkat P, Findeis E, Zacharek A, Powell B, Mccann M, Kim H, Zhang Z, Chopp M. Treatment of vascular dementia in female rats with AV-001, an Angiopoietin-1 mimetic peptide, improves cognitive function. Front Neurosci 2024; 18:1408205. [PMID: 39050669 PMCID: PMC11266070 DOI: 10.3389/fnins.2024.1408205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Background Vascular dementia (VaD) is a complex neurodegenerative disorder. We previously found that treatment of VaD in middle-aged male rats subjected to multiple microinfarction (MMI) with AV-001, a Tie2 receptor agonist, significantly improves cognitive function. Age and sex affect the development and response of VaD to therapeutic intervention. Thus, the present study investigated the therapeutic effect of AV-001 on VaD in aged female rats subjected to MMI. Methods Female 18-month-old Wistar rats were subjected to MMI by injecting either 1,000 (low dose, LD-MMI) or 6,000 (high dose, HD-MMI) cholesterol crystals of size 70-100 μm into the right internal carotid artery. AV-001 (1 μg/Kg, i.p.) was administered once daily after MMI for 1 month, with treatment initiated 1 day after MMI. A battery of behavioral tests to examine sensorimotor and cognitive functions was performed at 21-28 days after MMI. All rats were sacrificed at 1 month after MMI. Results Aged female rats subjected to LD-MMI exhibit severe neurological deficits, memory impairment, and significant white matter (WM) and oligodendrogenesis injury in the corpus callosum compared with control rats. HD-MMI in aged female rats induces significant anxiety- and depression-like behaviors, which were not detected in LD-MMI aged female rats. Also, HD-MMI induces significantly increased WM injury compared to LD-MMI. AV-001 treatment of LD-MMI and HD-MMI increases oligodendrogenesis, myelin and axon density in the corpus callosum and striatal WM bundles, promotes WM integrity and attenuates neurological and cognitive deficits. Additionally, both LD-MMI and HD-MMI rats exhibit a significant increase, while AV-001 significantly decreases the levels of inflammatory factors in the cerebrospinal fluid (CSF). Conclusion MMI reduces oligodendrogenesis, and induces demyelination, axonal injury and WM injury, and causes memory impairment, while HD-MMI induces increased WM injury and further depression-like behaviors compared to LD-MMI rats. AV-001 has a therapeutic effect on aged female rats with MMI by reducing WM damage and improving neuro-cognitive outcomes.
Collapse
Affiliation(s)
- Huanjia Gao
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Xianshuang Liu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Elizabeth Findeis
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Brianna Powell
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Mikkala Mccann
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Harold Kim
- Vasomune Therapeutics Inc., Toronto, ON, Canada
| | - Zhenggang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| |
Collapse
|
9
|
Lee JH, Kim JY, Ryu K, Al-Masni MA, Kim TH, Han D, Kim HG, Kim DH. JUST-Net: Jointly unrolled cross-domain optimization based spatio-temporal reconstruction network for accelerated 3D myelin water imaging. Magn Reson Med 2024; 91:2483-2497. [PMID: 38342983 DOI: 10.1002/mrm.30021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 02/13/2024]
Abstract
PURPOSE We introduced a novel reconstruction network, jointly unrolled cross-domain optimization-based spatio-temporal reconstruction network (JUST-Net), aimed at accelerating 3D multi-echo gradient-echo (mGRE) data acquisition and improving the quality of resulting myelin water imaging (MWI) maps. METHOD An unrolled cross-domain spatio-temporal reconstruction network was designed. The main idea is to combine frequency and spatio-temporal image feature representations and to sequentially implement convolution layers in both domains. The k-space subnetwork utilizes shared information from adjacent frames, whereas the image subnetwork applies separate convolutions in both spatial and temporal dimensions. The proposed reconstruction network was evaluated for both retrospectively and prospectively accelerated acquisition. Furthermore, it was assessed in simulation studies and real-world cases with k-space corruptions to evaluate its potential for motion artifact reduction. RESULTS The proposed JUST-Net enabled highly reproducible and accelerated 3D mGRE acquisition for whole-brain MWI, reducing the acquisition time from fully sampled 15:23 to 2:22 min within a 3-min reconstruction time. The normalized root mean squared error of the reconstructed mGRE images increased by less than 4.0%, and the correlation coefficients for MWI showed a value of over 0.68 when compared to the fully sampled reference. Additionally, the proposed method demonstrated a mitigating effect on both simulated and clinical motion-corrupted cases. CONCLUSION The proposed JUST-Net has demonstrated the capability to achieve high acceleration factors for 3D mGRE-based MWI, which is expected to facilitate widespread clinical applications of MWI.
Collapse
Affiliation(s)
- Jae-Hun Lee
- Department of Electrical and Electronic Engineering, Yonsei University, Seoul, Republic of Korea
- Artificial Intelligence and Robotics Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Jae-Yoon Kim
- Department of Electrical and Electronic Engineering, Yonsei University, Seoul, Republic of Korea
| | - Kanghyun Ryu
- Artificial Intelligence and Robotics Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Mohammed A Al-Masni
- Department of Artificial Intelligence, Sejong University, Seoul, Republic of Korea
| | - Tae Hyung Kim
- Department of Computer Engineering, Hongik University, Seoul, Republic of Korea
| | - Dongyeob Han
- Siemens Healthineers Ltd, Seoul, Republic of Korea
| | - Hyun Gi Kim
- Department of Radiology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong-Hyun Kim
- Department of Electrical and Electronic Engineering, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
10
|
Mariani JN, Mansky B, Madsen PM, Salinas D, Kesmen D, Huynh NPT, Kuypers NJ, Kesel ER, Bates J, Payne C, Chandler-Militello D, Benraiss A, Goldman SA. Repression of developmental transcription factor networks triggers aging-associated gene expression in human glial progenitor cells. Nat Commun 2024; 15:3873. [PMID: 38719882 PMCID: PMC11079006 DOI: 10.1038/s41467-024-48118-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 04/18/2024] [Indexed: 05/12/2024] Open
Abstract
Human glial progenitor cells (hGPCs) exhibit diminished expansion competence with age, as well as after recurrent demyelination. Using RNA-sequencing to compare the gene expression of fetal and adult hGPCs, we identify age-related changes in transcription consistent with the repression of genes enabling mitotic expansion, concurrent with the onset of aging-associated transcriptional programs. Adult hGPCs develop a repressive transcription factor network centered on MYC, and regulated by ZNF274, MAX, IKZF3, and E2F6. Individual over-expression of these factors in iPSC-derived hGPCs lead to a loss of proliferative gene expression and an induction of mitotic senescence, replicating the transcriptional changes incurred during glial aging. miRNA profiling identifies the appearance of an adult-selective miRNA signature, imposing further constraints on the expansion competence of aged GPCs. hGPC aging is thus associated with acquisition of a MYC-repressive environment, suggesting that suppression of these repressors of glial expansion may permit the rejuvenation of aged hGPCs.
Collapse
Affiliation(s)
- John N Mariani
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| | - Benjamin Mansky
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Pernille M Madsen
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health, Copenhagen, 2200, Denmark
| | - Dennis Salinas
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Deniz Kesmen
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Nguyen P T Huynh
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health, Copenhagen, 2200, Denmark
| | - Nicholas J Kuypers
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Erin R Kesel
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Janna Bates
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Casey Payne
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Devin Chandler-Militello
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Abdellatif Benraiss
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health, Copenhagen, 2200, Denmark.
| |
Collapse
|
11
|
Song H, Tomasevich A, Paolini A, Browne KD, Wofford KL, Kelley B, Kantemneni E, Kennedy J, Qiu Y, Schneider ALC, Dolle JP, Cullen DK, Smith DH. Sex differences in the extent of acute axonal pathologies after experimental concussion. Acta Neuropathol 2024; 147:79. [PMID: 38705966 PMCID: PMC11070329 DOI: 10.1007/s00401-024-02735-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/07/2024]
Abstract
Although human females appear be at a higher risk of concussion and suffer worse outcomes than males, underlying mechanisms remain unclear. With increasing recognition that damage to white matter axons is a key pathologic substrate of concussion, we used a clinically relevant swine model of concussion to explore potential sex differences in the extent of axonal pathologies. At 24 h post-injury, female swine displayed a greater number of swollen axonal profiles and more widespread loss of axonal sodium channels than males. Axon degeneration for both sexes appeared to be related to individual axon architecture, reflected by a selective loss of small caliber axons after concussion. However, female brains had a higher percentage of small caliber axons, leading to more extensive axon loss after injury compared to males. Accordingly, sexual dimorphism in axonal size is associated with more extensive axonal pathology in females after concussion, which may contribute to worse outcomes.
Collapse
Affiliation(s)
- Hailong Song
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Alexandra Tomasevich
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Andrew Paolini
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Kevin D Browne
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
- Center for Neurotrauma, Neurodegeneration and Restoration, Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104, USA
| | - Kathryn L Wofford
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
- Center for Neurotrauma, Neurodegeneration and Restoration, Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104, USA
| | - Brian Kelley
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Eashwar Kantemneni
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Justin Kennedy
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Yue Qiu
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Andrea L C Schneider
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Epidemiology, Biostatistics, and Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jean-Pierre Dolle
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - D Kacy Cullen
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
- Center for Neurotrauma, Neurodegeneration and Restoration, Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Douglas H Smith
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA.
| |
Collapse
|
12
|
Pavuluri K, Huston J, Ehman RL, Manduca A, Jack CR, Senjem ML, Vemuri P, Murphy MC. Associations between vascular health, brain stiffness and global cognitive function. Brain Commun 2024; 6:fcae073. [PMID: 38505229 PMCID: PMC10950054 DOI: 10.1093/braincomms/fcae073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/19/2023] [Accepted: 02/27/2024] [Indexed: 03/21/2024] Open
Abstract
Vascular brain injury results in loss of structural and functional connectivity and leads to cognitive impairment. Its various manifestations, including microinfarcts, microhaemorrhages and white matter hyperintensities, result in microstructural tissue integrity loss and secondary neurodegeneration. Among these, tissue microstructural alteration is a relatively early event compared with atrophy along the aging and neurodegeneration continuum. Understanding its association with cognition may provide the opportunity to further elucidate the relationship between vascular health and clinical outcomes. Magnetic resonance elastography offers a non-invasive approach to evaluate tissue mechanical properties, providing a window into the microstructural integrity of the brain. This retrospective study evaluated brain stiffness as a potential biomarker for vascular brain injury and its role in mediating the impact of vascular dysfunction on cognitive impairment. Seventy-five participants from the Mayo Clinic Study of Aging underwent brain imaging using a 3T MR imager with a spin-echo echo-planar imaging sequence for magnetic resonance elastography and T1- and T2-weighted pulse sequences. This study evaluated the effects of vascular biomarkers (white matter hyperintensities and cardiometabolic condition score) on brain stiffness using voxelwise analysis. Partial correlation analysis explored associations between brain stiffness, white matter hyperintensities, cardiometabolic condition and global cognition. Mediation analysis determined the role of stiffness in mediating the relationship between vascular biomarkers and cognitive performance. Statistical significance was set at P-values < 0.05. Diagnostic accuracy of magnetic resonance elastography stiffness for white matter hyperintensities and cardiometabolic condition was evaluated using receiver operator characteristic curves. Voxelwise linear regression analysis indicated white matter hyperintensities negatively correlate with brain stiffness, specifically in periventricular regions with high white matter hyperintensity levels. A negative association between cardiovascular risk factors and stiffness was also observed across the brain. No significant patterns of stiffness changes were associated with amyloid load. Global stiffness (µ) negatively correlated with both white matter hyperintensities and cardiometabolic condition when all other covariables including amyloid load were controlled. The positive correlation between white matter hyperintensities and cardiometabolic condition weakened and became statistically insignificant when controlling for other covariables. Brain stiffness and global cognition were positively correlated, maintaining statistical significance after adjusting for all covariables. These findings suggest mechanical alterations are associated with cognitive dysfunction and vascular brain injury. Brain stiffness significantly mediated the indirect effects of white matter hyperintensities and cardiometabolic condition on global cognition. Local cerebrovascular diseases (assessed by white matter hyperintensities) and systemic vascular risk factors (assessed by cardiometabolic condition) impact brain stiffness with spatially and statistically distinct effects. Global brain stiffness is a significant mediator between vascular disease measures and cognitive function, highlighting the value of magnetic resonance elastography-based mechanical assessments in understanding this relationship.
Collapse
Affiliation(s)
| | - John Huston
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Richard L Ehman
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Armando Manduca
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthew L Senjem
- Department of Information Technology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | |
Collapse
|
13
|
Zhang H, Yang Y, Zhang J, Huang L, Niu Y, Chen H, Liu Q, Wang R. Oligodendrocytes Play a Critical Role in White Matter Damage of Vascular Dementia. Neuroscience 2024; 538:1-10. [PMID: 37913862 DOI: 10.1016/j.neuroscience.2023.10.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/17/2023] [Accepted: 10/21/2023] [Indexed: 11/03/2023]
Abstract
With the deepening of population aging, the treatment of cognitive impairment and dementia is facing increasing challenges. Vascular dementia (VaD) is a cognitive dysfunction caused by brain blood flow damage and one of the most common causes of dementia after Alzheimer's disease. White matter damage in patients with chronic ischemic dementia often occurs before cognitive impairment, and its pathological changes include leukoaraiosis, myelin destruction and oligodendrocyte death. The pathophysiology of vascular dementia is complex, involving a variety of neuronal and vascular lesions. The current proposed mechanisms include calcium overload, oxidative stress, nitrative stress and inflammatory damage, which can lead to hypoxia-ischemia and demyelination. Oligodendrocytes are the only myelinating cells in the central nervous system and closely associated with VaD. In this review article, we intend to further discuss the role of oligodendrocytes in white matter and myelin injury in VaD and the development of anti-myelin injury target drugs.
Collapse
Affiliation(s)
- Hexin Zhang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Yanrong Yang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Jingjing Zhang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Li Huang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Yang Niu
- Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia medical University, Yinchuan 750004, Ningxia, China
| | - Hua Chen
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Qibing Liu
- Department of Pharmacy, The First Affiliated Hospital of Hainan Medical University, Haikou 570100, China
| | - Rui Wang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| |
Collapse
|
14
|
Wang Q, Schindler SE, Chen G, Mckay NS, McCullough A, Flores S, Liu J, Sun Z, Wang S, Wang W, Hassenstab J, Cruchaga C, Perrin RJ, Fagan AM, Morris JC, Wang Y, Benzinger TLS. Investigating White Matter Neuroinflammation in Alzheimer Disease Using Diffusion-Based Neuroinflammation Imaging. Neurology 2024; 102:e208013. [PMID: 38315956 PMCID: PMC10890836 DOI: 10.1212/wnl.0000000000208013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/13/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Alzheimer disease (AD) is primarily associated with accumulations of amyloid plaques and tau tangles in gray matter, however, it is now acknowledged that neuroinflammation, particularly in white matter (WM), significantly contributes to the development and progression of AD. This study aims to investigate WM neuroinflammation in the continuum of AD and its association with AD pathologies and cognition using diffusion-based neuroinflammation imaging (NII). METHODS This is a cross-sectional, single-center, retrospective evaluation conducted on an observational study of 310 older research participants who were enrolled in the Knight Alzheimer's Disease Research Center cohort. Hindered water ratio (HR), an index of WM neuroinflammation, was quantified by a noninvasive diffusion MRI method, NII. The alterations of NII-HR were investigated at different AD stages, classified based on CSF concentrations of β-amyloid (Aβ) 42/Aβ40 for amyloid and phosphorylated tau181 (p-tau181) for tau. On the voxel and regional levels, the relationship between NII-HR and CSF markers of amyloid, tau, and neuroinflammation were examined, as well as cognition. RESULTS This cross-sectional study included 310 participants (mean age 67.1 [±9.1] years), with 52 percent being female. Subgroups included 120 individuals (38.7%) with CSF measures of soluble triggering receptor expressed on myeloid cells 2, 80 participants (25.8%) with CSF measures of chitinase-3-like protein 1, and 110 individuals (35.5%) with longitudinal cognitive measures. The study found that cognitively normal individuals with positive CSF Aβ42/Aβ40 and p-tau181 had higher HR than healthy controls and those with positive CSF Aβ42/Aβ40 but negative p-tau181. WM tracts with elevated NII-HR in individuals with positive CSF Aβ42/Aβ40 and p-tau181 were primarily located in the posterior brain regions while those with elevated NII-HR in individuals with positive CSF Aβ42/Aβ40 and p-tau181 connected the posterior and anterior brain regions. A significant negative correlation between NII-HR and CSF Aβ42/Aβ40 was found in individuals with positive CSF Aβ42/Aβ40. Baseline NII-HR correlated with baseline cognitive composite score and predicted longitudinal cognitive decline. DISCUSSION Those findings suggest that WM neuroinflammation undergoes alterations before the onset of AD clinical symptoms and that it interacts with amyloidosis. This highlights the potential value of noninvasive monitoring of WM neuroinflammation in AD progression and treatment.
Collapse
Affiliation(s)
- Qing Wang
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Suzanne E Schindler
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Gengsheng Chen
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Nicole S Mckay
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Austin McCullough
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Shaney Flores
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Jingxia Liu
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Zhexian Sun
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Sicheng Wang
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Wenshang Wang
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Jason Hassenstab
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Carlos Cruchaga
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Richard J Perrin
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Anne M Fagan
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - John C Morris
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Yong Wang
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| | - Tammie L S Benzinger
- From the Mallinckrodt Institute of Radiology (Q.W., G.C., N.S.M., A.M., S.F., Y.W., T.L.S.B.), Knight Alzheimer Disease Research Center (Q.W., S.E.S., G.C., N.S.M., A.M., J.H., R.J.P., A.M.F., J.C.M., T.L.S.B.), Department of Neurology (S.E.S., J.H., C.C., A.M.F., J.C.M.), Department of Surgery (J.L.), Department of Biomedical Engineering (Z.S.), Department of Electrical and System Engineering (S.W., W.W., Y.W.), Department of Psychiatry (C.C.), Department of Pathology & Immunology (R.J.P.), Department of Obstetrics & Gynecology (Y.W.), and Department of Neurosurgery (T.L.S.B.), Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
15
|
Wang H, Zhang Z, Hongpaisan J. PKCε activator protects hippocampal microvascular disruption and memory defect in 3×Tg-Alzheimer's disease mice with cerebral microinfarcts. Front Aging Neurosci 2023; 15:1272361. [PMID: 38187357 PMCID: PMC10768563 DOI: 10.3389/fnagi.2023.1272361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/23/2023] [Indexed: 01/09/2024] Open
Abstract
Background Current evidence suggests that microvessel disease is involved in Alzheimer's disease (AD). Cerebrovascular disease correlates with cardiovascular disease and is complicated in ≈40% of AD patients. The protein kinase C (PKC) ε activator DCPLA can stimulate human antigen (Hu) R that prevents degradation and promotes the translation of mitochondrial Mn-superoxide dismutase (MnSOD) and vascular endothelial growth factor-A (VEGF) mRNAs. Methods To induce brain microinfarcts, we injected triple transgenic (3×Tg) and wild-type (WT) control mice with microbeads (20 μm caliber) into common carotid arteries, with or without the DCPLA-ME (methyl-ester) for 2 weeks. After water maze training, mice at 16 months old were examined for confocal immunohistochemistry at a single cell or microvessel level in the hippocampal CA1 area, important for spatial memory storage, and in the dorsal hippocampus by western blots. Results In 3×Tg mice without cerebral microinfarcts, an accelerating age-related increase in (mild) oxidative stress and hypoxia inducible factor (HIF)-1α, but a reduction in VEGF, mitochondrial transcription factor A (TFAM), and MnSOD were associated with capillary loss. The change was less pronounced in arterioles. However, in 3×Tg mice with cerebral microinfarcts, increasing arteriolar diameter and their wall cells were related with the strong oxidative DNA damage 8-hydroxy-2'-deoxyguanosine (8-OHdG), apoptosis (cleaved caspase 3), and sustained hypoxia (increased HIF-1α and VEGF/PKCε/extracellular signal regulated kinase or ERK pathway). Microocclusion enhanced the loss of the synaptic marker spinophilin, astrocytic number, and astrocyte-vascular coupling areas and demyelination of axons. DCPLA-ME prevented spatial memory defect; strong oxidative stress-related apoptosis; sustained hypoxia (by reducing HIF-1α and VEGF); and exaggerated cell repair in arteriolar walls, pericapillary space dilation, neuro-glial-vascular disruption, and demyelination. Conclusion In conclusion, in 3×Tg mice with cerebral microinfarcts, sustained hypoxia (increased HIF-1α and VEGF signals) is dominant with arteriolar wall thickening, and DCPLA has a protective effect on sustained hypoxia.
Collapse
Affiliation(s)
| | | | - Jarin Hongpaisan
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
16
|
Obis E, Sol J, Andres-Benito P, Martín-Gari M, Mota-Martorell N, Galo-Licona JD, Piñol-Ripoll G, Portero-Otin M, Ferrer I, Jové M, Pamplona R. Lipidomic Alterations in the Cerebral Cortex and White Matter in Sporadic Alzheimer's Disease. Aging Dis 2023; 14:1887-1916. [PMID: 37196109 PMCID: PMC10529741 DOI: 10.14336/ad.2023.0217] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/17/2023] [Indexed: 05/19/2023] Open
Abstract
Non-targeted LC-MS/MS-based lipidomic analysis was conducted in post-mortem human grey matter frontal cortex area 8 (GM) and white matter of the frontal lobe centrum semi-ovale (WM) to identify lipidome fingerprints in middle-aged individuals with no neurofibrillary tangles and senile plaques, and cases at progressive stages of sporadic Alzheimer's disease (sAD). Complementary data were obtained using RT-qPCR and immunohistochemistry. The results showed that WM presents an adaptive lipid phenotype resistant to lipid peroxidation, characterized by a lower fatty acid unsaturation, peroxidizability index, and higher ether lipid content than the GM. Changes in the lipidomic profile are more marked in the WM than in GM in AD with disease progression. Four functional categories are associated with the different lipid classes affected in sAD: membrane structural composition, bioenergetics, antioxidant protection, and bioactive lipids, with deleterious consequences affecting both neurons and glial cells favoring disease progression.
Collapse
Affiliation(s)
- Elia Obis
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Joaquim Sol
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
- Catalan Institute of Health (ICS), Lleida, Spain, Research Support Unit (USR), Fundació Institut Universitari per a la Recerca en Atenció Primària de Salut Jordi Gol i Gurina (IDIAP JGol), Lleida, Spain.
| | - Pol Andres-Benito
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.
- Bellvitge University Hospital-Bellvitge Biomedical Research Institute (IDIBELL), E-08907 Hospitalet de Llobregat, Barcelona, Spain.
| | - Meritxell Martín-Gari
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Natàlia Mota-Martorell
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - José Daniel Galo-Licona
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius, Clinical Neuroscience Research, Santa Maria University Hospital, IRBLleida, Lleida, Spain.
| | - Manuel Portero-Otin
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Isidro Ferrer
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.
- Bellvitge University Hospital-Bellvitge Biomedical Research Institute (IDIBELL), E-08907 Hospitalet de Llobregat, Barcelona, Spain.
- Department of Pathology and Experimental Therapeutics, University of Barcelona, L’Hospitalet de Llobregat, Barcelona, Spain.
| | - Mariona Jové
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Reinald Pamplona
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| |
Collapse
|
17
|
Hou J, Zhou Y, Cai Z, Terekhova M, Swain A, Andhey PS, Guimaraes RM, Ulezko Antonova A, Qiu T, Sviben S, Strout G, Fitzpatrick JAJ, Chen Y, Gilfillan S, Kim DH, Van Dyken SJ, Artyomov MN, Colonna M. Transcriptomic atlas and interaction networks of brain cells in mouse CNS demyelination and remyelination. Cell Rep 2023; 42:112293. [PMID: 36952346 PMCID: PMC10511667 DOI: 10.1016/j.celrep.2023.112293] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/04/2022] [Accepted: 03/06/2023] [Indexed: 03/24/2023] Open
Abstract
Demyelination is a hallmark of multiple sclerosis, leukoencephalopathies, cerebral vasculopathies, and several neurodegenerative diseases. The cuprizone mouse model is widely used to simulate demyelination and remyelination occurring in these diseases. Here, we present a high-resolution single-nucleus RNA sequencing (snRNA-seq) analysis of gene expression changes across all brain cells in this model. We define demyelination-associated oligodendrocytes (DOLs) and remyelination-associated MAFBhi microglia, as well as astrocytes and vascular cells with signatures of altered metabolism, oxidative stress, and interferon response. Furthermore, snRNA-seq provides insights into how brain cell types connect and interact, defining complex circuitries that impact demyelination and remyelination. As an explicative example, perturbation of microglia caused by TREM2 deficiency indirectly impairs the induction of DOLs. Altogether, this study provides a rich resource for future studies investigating mechanisms underlying demyelinating diseases.
Collapse
Affiliation(s)
- Jinchao Hou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yingyue Zhou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhangying Cai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marina Terekhova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Amanda Swain
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Prabhakar S Andhey
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rafaela M Guimaraes
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Ribeirão Preto Medical School, University of São Paulo - Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Alina Ulezko Antonova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tian Qiu
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sanja Sviben
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gregory Strout
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James A J Fitzpatrick
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO 63110, USA; Departments of Cell Biology and Physiology and Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Yun Chen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Susan Gilfillan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Do-Hyun Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven J Van Dyken
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
18
|
Maitre M, Jeltsch-David H, Okechukwu NG, Klein C, Patte-Mensah C, Mensah-Nyagan AG. Myelin in Alzheimer's disease: culprit or bystander? Acta Neuropathol Commun 2023; 11:56. [PMID: 37004127 PMCID: PMC10067200 DOI: 10.1186/s40478-023-01554-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with neuronal and synaptic losses due to the accumulation of toxic amyloid β (Αβ) peptide oligomers, plaques, and tangles containing tau (tubulin-associated unit) protein. While familial AD is caused by specific mutations, the sporadic disease is more common and appears to result from a complex chronic brain neuroinflammation with mitochondriopathies, inducing free radicals' accumulation. In aged brain, mutations in DNA and several unfolded proteins participate in a chronic amyloidosis response with a toxic effect on myelin sheath and axons, leading to cognitive deficits and dementia. Αβ peptides are the most frequent form of toxic amyloid oligomers. Accumulations of misfolded proteins during several years alters different metabolic mechanisms, induce chronic inflammatory and immune responses with toxic consequences on neuronal cells. Myelin composition and architecture may appear to be an early target for the toxic activity of Aβ peptides and others hydrophobic misfolded proteins. In this work, we describe the possible role of early myelin alterations in the genesis of neuronal alterations and the onset of symptomatology. We propose that some pathophysiological and clinical forms of the disease may arise from structural and metabolic disorders in the processes of myelination/demyelination of brain regions where the accumulation of non-functional toxic proteins is important. In these forms, the primacy of the deleterious role of amyloid peptides would be a matter of questioning and the initiating role of neuropathology would be primarily the fact of dysmyelination.
Collapse
Affiliation(s)
- Michel Maitre
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France.
| | - Hélène Jeltsch-David
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
- Biotechnologie et signalisation cellulaire, UMR 7242 CNRS, Université de Strasbourg, 300 Boulevard Sébastien Brant CS 10413, Illkirch cedex, 67412, France
| | - Nwife Getrude Okechukwu
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| | - Christian Klein
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| | - Christine Patte-Mensah
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| | - Ayikoe-Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| |
Collapse
|
19
|
Kallianpur KJ, Masaki KH, Chen R, Willcox BJ, Allsopp RC, Davy P, Dodge HH. Weak Social Networks in Late Life Predict Incident Alzheimer's Disease: The Kuakini Honolulu-Asia Aging Study. J Gerontol A Biol Sci Med Sci 2023; 78:663-672. [PMID: 36208464 PMCID: PMC10061568 DOI: 10.1093/gerona/glac215] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND We assessed 10-year longitudinal associations between late-life social networks and incidence of all-cause dementia (ACD), Alzheimer's disease (AD), and vascular dementia (VaD) in Japanese-American men. METHODS We prospectively analyzed, from baseline (1991-1993) through 1999-2000, 2636 initially nondemented Kuakini Honolulu-Asia Aging Study participants who remained dementia-free during the first 3 years of follow-up. Global cognition was evaluated by the Cognitive Abilities Screening Instrument (CASI); depressive symptoms by the 11-item Center for Epidemiologic Studies Depression (CES-D) Scale; and social networks by the Lubben Social Network Scale (LSNS). Median split of LSNS scores defined weak/strong social network groups. A panel of neurologists and geriatricians diagnosed and classified dementia; AD and VaD diagnoses comprised cases in which AD or VaD, respectively, were considered the primary cause of dementia. RESULTS Median (range) baseline age was 77 (71-93) years. Participants with weak (LSNS score ≤29) versus strong (>29) social networks had higher age-adjusted incidence (in person-years) of ACD (12.6 vs. 8.7; p = .014) and AD (6.7 vs. 4.0; p = .007) but not VaD (2.4 vs. 1.4; p = .15). Kaplan-Meier curves showed a lower likelihood of survival free of ACD (log-rank p < .0001) and AD (p = .0006) for men with weak networks. In Cox proportional hazards models adjusting for age, education, APOE ɛ4, prevalent stroke, depressive symptoms, and CASI score (all at baseline), weak networks predicted increased incidence of ACD (hazard ratio [HR] = 1.52, p = .009) and AD (HR = 1.67, p = .014) but not VaD (p > .2). CONCLUSION Weak social networks may heighten the risk of dementia and AD, underscoring the need to promote social connectedness in older adults.
Collapse
Affiliation(s)
- Kalpana J Kallianpur
- Kuakini Center for Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, USA
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, University of Hawaii, Honolulu, Hawaii, USA
| | - Kamal H Masaki
- Kuakini Center for Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, USA
- Department of Geriatric Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Randi Chen
- Kuakini Center for Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, USA
| | - Bradley J Willcox
- Kuakini Center for Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, USA
- Department of Geriatric Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Richard C Allsopp
- Kuakini Center for Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, USA
| | - Philip Davy
- Kuakini Center for Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, USA
| | - Hiroko H Dodge
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
- Layton Aging and Alzheimer’s Disease Center, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
20
|
Hu Y, Zhang M, Liu B, Tang Y, Wang Z, Wang T, Zheng J, Zhang J. Honokiol prevents chronic cerebral hypoperfusion induced astrocyte A1 polarization to alleviate neurotoxicity by targeting SIRT3-STAT3 axis. Free Radic Biol Med 2023; 202:62-75. [PMID: 36997099 DOI: 10.1016/j.freeradbiomed.2023.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 03/11/2023] [Accepted: 03/20/2023] [Indexed: 04/01/2023]
Abstract
Alzheimer's Dementia (AD) and Vascular Dementia (VaD) are two main types of dementias for which no specific treatment is available. Chronic Cerebral Hypoperfusion (CCH) is a pathogenesis underlying AD and VaD that promotes neuroinflammatory responses and oxidative stress. Honokiol (HNK) is a natural compound isolated from magnolia leaves that can easily cross blood brain barrier and has anti-inflammatory and antioxidant effects. In the present study, the effects of HNK on astrocyte polarization and neurological damage in in vivo and in vitro models of chronic cerebral hypoperfusion were explored. We found that HNK was able to inhibit the phosphorylation and nuclear translocation of STAT3, A1 polarization, and reduce conditioned medium's neuronal toxicity of astrocyte under chronic hypoxia induced by cobalt chloride; STAT3 phosphorylation inhibitor C188-9 was able to mimic the above effects of HNK, suggesting that HNK may inhibit chronic hypoxia-induced A1 polarization in astrocytes via STAT3. SIRT3 inhibitor 3-TYP reversed, while Sirt3 overexpression mimicked the inhibitory effects of HNK on oxidative stress, STAT3 phosphorylation and nuclear translocation, A1 polarization and neuronal toxicity of astrocyte under chronic hypoxic conditions. For in vivo research, continuous intraperitoneal injection of HNK (1mg/kg) for 21 days ameliorated the decrease in SIRT3 activity and oxidative stress, inhibited astrocytic STAT3 nuclear translocation and A1 polarization, and prevented neuron and synaptic loss in the hippocampal of CCH rats. Besides, HNK application improved the spatial memory impairment of CCH rats, as assessed with Morris Water Maze. In conclusion, these results suggest that the phytochemical HNK can inhibit astrocyte A1 polarization via regulating SIRT3-STAT3 axis, thus improving CCH-induced neurological damage. These results highlight HNK as novel treatment for dementia with underlying vascular mechanisms.
Collapse
Affiliation(s)
- Yuan Hu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China.
| | - Miao Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Bihan Liu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Yingying Tang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Zhuo Wang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Tao Wang
- Department of Neurology, First Clinical Medical College of China Three Gorges University, Yichang, Hubei, 443003, China
| | - Jiaxin Zheng
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China.
| |
Collapse
|
21
|
Boa Sorte Silva NC, Dao E, Liang Hsu C, Tam RC, Lam K, Alkeridy W, Laule C, Vavasour IM, Stein RG, Liu-Ambrose T. Myelin and Physical Activity in Older Adults With Cerebral Small Vessel Disease and Mild Cognitive Impairment. J Gerontol A Biol Sci Med Sci 2023; 78:545-553. [PMID: 35876839 DOI: 10.1093/gerona/glac149] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Myelin loss is a feature of cerebral small vessel disease (cSVD). Although physical activity levels may exert protective effects over cSVD pathology, its specific relationship with myelin content in people living with the cSVD is unknown. Thus, we investigated whether physical activity levels are associated with myelin in community-dwelling older adults with cSVD and mild cognitive impairment. METHODS Cross-sectional data from 102 individuals with cSVD and mild cognitive impairment were analyzed (mean age [SD] = 74.7 years [5.5], 63.7% female). Myelin was measured using a magnetic resonance gradient and spin echo sequence. Physical activity was estimated using the Physical Activity Scale for the Elderly. Hierarchical regression models adjusting for total intracranial volume, age, sex, body mass index, and education were conducted to determine the associations between myelin content and physical activity. Significant models were further adjusted for white matter hyperintensity volume. RESULTS In adjusted models, greater physical activity was linked to higher myelin content in the whole-brain white matter (R2change = .04, p = .048). Greater physical activity was also associated with myelin content in the sagittal stratum (R2change = .08, p = .004), anterior corona radiata (R2change = .04, p = .049), and genu of the corpus callosum (R2change = .05, p = .018). Adjusting for white matter hyperintensity volume did not change any of these associations. CONCLUSIONS Physical activity may be a strategy to maintain myelin in older adults with cSVD and mild cognitive impairment. Future randomized controlled trials of exercise are needed to determine whether exercise increases myelin content.
Collapse
Affiliation(s)
- Nárlon C Boa Sorte Silva
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Elizabeth Dao
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Radiology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - Chun Liang Hsu
- Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
| | - Roger C Tam
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,School of Biomedical Engineering, Faculty of Applied Science and Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kevin Lam
- Department of Medicine, Division of Neurology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Walid Alkeridy
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Medicine, King Saud University, College of Medicine, Riyadh, Saudi Arabia.,Department of Medicine, Division of Geriatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cornelia Laule
- Department of Radiology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Physics and Astronomy, University of British Columbia, Vancouver, British Columbia, Canada
| | - Irene M Vavasour
- Department of Radiology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ryan G Stein
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Teresa Liu-Ambrose
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
22
|
Endothelial Dysfunction in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:ijms24032909. [PMID: 36769234 PMCID: PMC9918222 DOI: 10.3390/ijms24032909] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
The cerebral vascular system stringently regulates cerebral blood flow (CBF). The components of the blood-brain barrier (BBB) protect the brain from pathogenic infections and harmful substances, efflux waste, and exchange substances; however, diseases develop in cases of blood vessel injuries and BBB dysregulation. Vascular pathology is concurrent with the mechanisms underlying aging, Alzheimer's disease (AD), and vascular dementia (VaD), which suggests its involvement in these mechanisms. Therefore, in the present study, we reviewed the role of vascular dysfunction in aging and neurodegenerative diseases, particularly AD and VaD. During the development of the aforementioned diseases, changes occur in the cerebral blood vessel morphology and local cells, which, in turn, alter CBF, fluid dynamics, and vascular integrity. Chronic vascular inflammation and blood vessel dysregulation further exacerbate vascular dysfunction. Multitudinous pathogenic processes affect the cerebrovascular system, whose dysfunction causes cognitive impairment. Knowledge regarding the pathophysiology of vascular dysfunction in neurodegenerative diseases and the underlying molecular mechanisms may lead to the discovery of clinically relevant vascular biomarkers, which may facilitate vascular imaging for disease prevention and treatment.
Collapse
|
23
|
Srivishagan S, Kumaralingam L, Thanikasalam K, Pinidiyaarachchi UAJ, Ratnarajah N. Discriminative patterns of white matter changes in Alzheimer's. Psychiatry Res Neuroimaging 2023; 328:111576. [PMID: 36495726 DOI: 10.1016/j.pscychresns.2022.111576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/12/2022] [Accepted: 11/22/2022] [Indexed: 12/02/2022]
Abstract
Changes in structural connectivity of the Alzheimer's brain have not been widely studied utilizing cutting-edge methodologies. This study develops an efficient structural connectome-based convolutional neural network (CNN) to classify the AD and uses explanations of CNNs' choices in classification to pinpoint the discriminative changes in white matter connectivity in AD. A CNN architecture has been developed to classify normal control (NC) and AD subjects from the weighted structural connectome. Then, the CNN classification decision is visually analyzed using gradient-based localization techniques to identify the discriminative changes in white matter connectivity in Alzheimer's. The cortical regions involved in the identified discriminative structural connectivity changes in AD are highly covered in the temporal/subcortical regions. A specific pattern is identified in the discriminative changes in structural connectivity of AD, where the white matter changes are revealed within the temporal/subcortical regions and from the temporal/subcortical regions to the frontal and parietal regions in both left and right hemispheres. The proposed approach has the potential to comprehensively analyze the discriminative structural connectivity differences in AD, change the way of detecting biomarkers, and help clinicians better understand the structural changes in AD and provide them with more confidence in automated diagnostic systems.
Collapse
Affiliation(s)
- Subaramya Srivishagan
- Department of Physical Science, Faculty of Applied Science, University of Vavuniya, Vavuniya, Sri Lanka; PGIS, University of Peradeniya, Peradeniya, Sri Lanka
| | - Logiraj Kumaralingam
- Department of Computer Science, Faculty of Science, University of Jaffna, Jaffna, Sri Lanka
| | - Kokul Thanikasalam
- Department of Computer Science, Faculty of Science, University of Jaffna, Jaffna, Sri Lanka
| | - U A J Pinidiyaarachchi
- Department of Statistics and Computer Science, Faculty of Science, University of Peradeniya, Peradeniya, Sri Lanka
| | - Nagulan Ratnarajah
- Department of Physical Science, Faculty of Applied Science, University of Vavuniya, Vavuniya, Sri Lanka.
| |
Collapse
|
24
|
Wang W, Li Y, Meng X. Vitamin D and neurodegenerative diseases. Heliyon 2023; 9:e12877. [PMID: 36820164 PMCID: PMC9938420 DOI: 10.1016/j.heliyon.2023.e12877] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/23/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
Neurodegenerative diseases, featured by progressive loss of structure or function of neurons, are considered incurable at present. Movement disorders like tremor and postural instability, cognitive or behavioral disorders such as memory impairment are the most common symptoms of them and the growing patient population of neurodegenerative diseases poses a serious threat to public health and a burden on economic development. Hence, it is vital to prevent the occurrence of the diseases and delay their progress. Vitamin D can be transformed into a hormone in vivo with both genomic and non-genomic actions, exerting diverse physiological effects. Cumulative evidence indicates that vitamin D can ameliorate neurodegeneration by regulating pertinent molecules and signaling pathways including maintaining Ca2+ homeostasis, reducing oxidative stress, inhibiting inflammation, suppressing the formation and aggregation of the pathogenic protein, etc. This review updates discoveries of molecular mechanisms underlying biological functions of vitamin D in neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, multiple sclerosis, and vascular dementia. Clinical trials investigating the influence of vitamin D supplementation in patients with neurodegenerative diseases are also summarized. The synthesized information will probably provoke an enhanced understanding of the neuroprotective roles of vitamin D in the nervous system and provide therapeutic options for patients with neurodegenerative diseases in the future.
Collapse
|
25
|
Hou J, Chen Y, Grajales-Reyes G, Colonna M. TREM2 dependent and independent functions of microglia in Alzheimer's disease. Mol Neurodegener 2022; 17:84. [PMID: 36564824 PMCID: PMC9783481 DOI: 10.1186/s13024-022-00588-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/02/2022] [Indexed: 12/25/2022] Open
Abstract
Microglia are central players in brain innate immunity and have been the subject of extensive research in Alzheimer's disease (AD). In this review, we aim to summarize the genetic and functional discoveries that have advanced our understanding of microglia reactivity to AD pathology. Given the heightened AD risk posed by rare variants of the microglial triggering receptor expressed on myeloid cells 2 (TREM2), we will focus on the studies addressing the impact of this receptor on microglia responses to amyloid plaques, tauopathy and demyelination pathologies in mouse and human. Finally, we will discuss the implications of recent discoveries on microglia and TREM2 biology on potential therapeutic strategies for AD.
Collapse
Affiliation(s)
- Jinchao Hou
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Yun Chen
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA ,grid.4367.60000 0001 2355 7002Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Gary Grajales-Reyes
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Marco Colonna
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
26
|
Nguyen AT, Kouri N, Labuzan SA, Przybelski SA, Lesnick TG, Raghavan S, Reid RI, Reichard RR, Knopman DS, Petersen RC, Jack CR, Mielke MM, Dickson DW, Graff-Radford J, Murray ME, Vemuri P. Neuropathologic scales of cerebrovascular disease associated with diffusion changes on MRI. Acta Neuropathol 2022; 144:1117-1125. [PMID: 35841412 PMCID: PMC9637622 DOI: 10.1007/s00401-022-02465-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 01/26/2023]
Abstract
Summarizing the multiplicity and heterogeneity of cerebrovascular disease (CVD) features into a single measure has been difficult in both neuropathology and imaging studies. The objective of this work was to evaluate the association between neuroimaging surrogates of CVD and two available neuropathologic CVD scales in those with both antemortem imaging CVD measures and postmortem CVD evaluation. Individuals in the Mayo Clinic Study of Aging with MRI scans within 5 years of death (N = 51) were included. Antemortem CVD measures were computed from diffusion MRI (dMRI), FLAIR, and T2* GRE imaging modalities and compared with postmortem neuropathologic findings using Kalaria and Strozyk Scales. Of all the neuroimaging measures, both regional and global dMRI measures were associated with Kalaria and Strozyk Scales (p < 0.05) and modestly correlated with global cognitive performance. The major conclusions from this study were: (i) microstructural white matter injury measurements using dMRI may be meaningful surrogates of neuropathologic CVD scales, because they aid in capturing diffuse (and early) changes to white matter and secondary neurodegeneration due to lesions; (ii) vacuolation in the corpus callosum may be associated with white matter changes measured on antemortem dMRI imaging; (iii) Alzheimer's disease neuropathologic change did not associate with neuropathologic CVD scales; and (iv) future work should be focused on developing better quantitative measures utilizing dMRI to optimally assess CVD-related neuropathologic changes.
Collapse
Affiliation(s)
- Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Naomi Kouri
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Sydney A Labuzan
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Timothy G Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Sheelakumari Raghavan
- Department of Radiology, Mayo Clinic and Foundation, 200 First Street SW, Rochester, MN, 55905, USA
| | - Robert I Reid
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - R Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Clifford R Jack
- Department of Radiology, Mayo Clinic and Foundation, 200 First Street SW, Rochester, MN, 55905, USA
| | - Michelle M Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | | | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic and Foundation, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
27
|
Wakhloo D, Oberhauser J, Madira A, Mahajani S. From cradle to grave: neurogenesis, neuroregeneration and neurodegeneration in Alzheimer's and Parkinson's diseases. Neural Regen Res 2022; 17:2606-2614. [PMID: 35662189 PMCID: PMC9165389 DOI: 10.4103/1673-5374.336138] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/16/2021] [Accepted: 12/02/2021] [Indexed: 11/29/2022] Open
Abstract
Two of the most common neurodegenerative disorders - Alzheimer's and Parkinson's diseases - are characterized by synaptic dysfunction and degeneration that culminate in neuronal loss due to abnormal protein accumulation. The intracellular aggregation of hyper-phosphorylated tau and the extracellular aggregation of amyloid beta plaques form the basis of Alzheimer's disease pathology. The major hallmark of Parkinson's disease is the loss of dopaminergic neurons in the substantia nigra pars compacta, following the formation of Lewy bodies, which consists primarily of alpha-synuclein aggregates. However, the discrete mechanisms that contribute to neurodegeneration in these disorders are still poorly understood. Both neuronal loss and impaired adult neurogenesis have been reported in animal models of these disorders. Yet these findings remain subject to frequent debate due to a lack of conclusive evidence in post mortem brain tissue from human patients. While some publications provide significant findings related to axonal regeneration in Alzheimer's and Parkinson's diseases, they also highlight the limitations and obstacles to the development of neuroregenerative therapies. In this review, we summarize in vitro and in vivo findings related to neurogenesis, neuroregeneration and neurodegeneration in the context of Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- Debia Wakhloo
- Deparment of Neuropathology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Jane Oberhauser
- Deparment of Neuropathology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Angela Madira
- Deparment of Neuropathology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Sameehan Mahajani
- Deparment of Neuropathology, Stanford University, School of Medicine, Stanford, CA, USA
| |
Collapse
|
28
|
Hase Y, Ameen‐Ali KE, Waller R, Simpson JE, Stafford C, Mahesh A, Ryan L, Pickering L, Bodman C, Hase M, Boche D, Horsburgh K, Wharton SB, Kalaria RN. Differential perivascular microglial activation in the deep white matter in vascular dementia developed post-stroke. Brain Pathol 2022; 32:e13101. [PMID: 35748290 PMCID: PMC9616090 DOI: 10.1111/bpa.13101] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/30/2022] [Indexed: 11/28/2022] Open
Abstract
With the hypothesis that perivascular microglia are involved as neuroinflammatory components of the gliovascular unit contributing to white matter hyperintensities on MRI and pathophysiology, we assessed their status in stroke survivors who develop dementia. Immunohistochemical and immunofluorescent methods were used to assess the distribution and quantification of total and perivascular microglial cell densities in 68 brains focusing on the frontal lobe WM and overlying neocortex in post-stroke dementia (PSD), post-stroke non-dementia (PSND) and similar age control subjects. We primarily used CD68 as a marker of phagocytic microglia, as well as other markers of microglia including Iba-1 and TMEM119, and the myeloid cell marker TREM2 to assess dementia-specific changes. We first noted greater total densities of CD68+ and TREM2+ cells per mm2 in the frontal WM compared to the overlying cortex across the stroke cases and controls (p = 0.001). PSD subjects showed increased percentage of activated perivascular CD68+ cells distinct from ramified or primed microglia in the WM (p < 0.05). However, there was no apparent change in perivascular TREM2+ cells. Total densities of TREM2+ cells were only ~10% of CD68+ cells but there was high degree of overlap (>70%) between them in both the WM and the cortex. CD68 and Iba-1 or CD68 and TMEM119 markers were colocalised by ~55%. Within the deep WM, ~30% of CD68+ cells were co-localised with fragments of degraded myelin basic protein. Among fragmented CD68+ cells in adjacent WM of PSD subjects, >80% of the cells expressed cleaved caspase-3. Our observations suggest although the overall repertoire of perivascular microglial cells is not changed in the parenchyma, PSD subjects accrue more perivascular-activated CD68+ microglia rather than TREM2+ cells. This implies there is a subset of CD68+ cells, which are responsible for the differential response in perivascular inflammation within the gliovascular unit of the deep WM.
Collapse
Affiliation(s)
- Yoshiki Hase
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Kamar E. Ameen‐Ali
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
- Institute of Neuroscience and PsychologyUniversity of Glasgow, Queen Elizabeth University HospitalGlasgowUK
| | - Rachel Waller
- Sheffield Institute for Translational NeuroscienceUniversity of SheffieldSheffieldUK
| | - Julie E. Simpson
- Sheffield Institute for Translational NeuroscienceUniversity of SheffieldSheffieldUK
| | - Charlotte Stafford
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Ayushi Mahesh
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Lucy Ryan
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Lucy Pickering
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Caroline Bodman
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Mai Hase
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Delphine Boche
- Clinical and Experimental Sciences, Faculty of MedicineUniversity of Southampton, Southampton General HospitalSouthamptonUK
| | - Karen Horsburgh
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Stephen B. Wharton
- Sheffield Institute for Translational NeuroscienceUniversity of SheffieldSheffieldUK
| | - Raj N. Kalaria
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
29
|
Yang Y, Zhao X, Zhu Z, Zhang L. Vascular dementia: A microglia's perspective. Ageing Res Rev 2022; 81:101734. [PMID: 36113763 DOI: 10.1016/j.arr.2022.101734] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/28/2022] [Accepted: 09/11/2022] [Indexed: 01/31/2023]
Abstract
Vascular dementia (VaD) is a second most common form of age-related dementia. It is characterized by cognitive impairment associated with vascular pathology, symptoms mainly caused by cerebral damage due to inadequate blood flow to the brain. The pathogenesis of VaD is complex, and a growing body of literature emphasizes on the involvement of microglia in disease development and progression. Here, we review the current knowledge on the role of microglia in regulating neuroinflammation under the pathogenesis of VaD. The commonly used animal and cell models for understanding the disease pathogenesis were summarized. The mechanisms by which microglia contribute to VaD are multifactorial, and we specifically focus on some of the predominant functions of microglia, including chemotaxis, secretory property, phagocytosis, and its crosstalk with other neurovascular unit cells. Finally, potential therapeutic strategies targeting microglia-modulated neuroinflammation are discussed.
Collapse
Affiliation(s)
- Yi Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China.
| | - Xinyuan Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China
| | - Zirui Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China
| | - Lihui Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China.
| |
Collapse
|
30
|
Tsentidou G, Moraitou D, Tsolaki M. Emotion Recognition in a Health Continuum: Comparison of Healthy Adults of Advancing Age, Community Dwelling Adults Bearing Vascular Risk Factors and People Diagnosed with Mild Cognitive Impairment. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:13366. [PMID: 36293946 PMCID: PMC9602834 DOI: 10.3390/ijerph192013366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/11/2022] [Accepted: 10/14/2022] [Indexed: 06/16/2023]
Abstract
The identification of basic emotions plays an important role in social relationships and behaviors linked to survival. In neurodegenerative conditions such as Alzheimer's disease (AD), the ability to recognize emotions may already be impaired at early stages of the disease, such as the stage of Mild Cognitive Impairment (MCI). However, as regards vascular pathologies related to cognitive impairment, very little is known about emotion recognition in people bearing vascular risk factors (VRF). Therefore, the aim of the present study was to examine emotion recognition ability in the health continuum "healthy advancing age-advancing age with VRF-MCI". The sample consisted of 106 adults divided in three diagnostic groups; 43 adults with MCI, 41 adults bearing one or more VRF, and 22 healthy controls of advancing age (HC). Since HC were more educated and younger than the other two groups, the age-group and level of educational were taken into account in the statistical analyses. A dynamic visual test was administered to examine recognition of basic emotions and emotionally neutral conditions. The results showed only a significant diagnostic group x educational level interaction as regards total emotion recognition ability, F (4, 28.910) = 4.117 p = 0.004 η2 = 0.166. High educational level seems to contribute to a high-level-emotion-recognition-performance both in healthy adults of advancing age and in adults bearing vascular risk factors. Medium educational level appears to play the same role only in healthy adults. Neither educational level can help MCI people to enhance their significantly lower emotion recognition ability.
Collapse
Affiliation(s)
- Glykeria Tsentidou
- Laboratory of Psychology, Department of Experimental and Cognitive Psychology, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Laboratory of Neurodegenerative Diseases, Center for Interdisciplinary Research and Innovation (CIRI), Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Despina Moraitou
- Laboratory of Psychology, Department of Experimental and Cognitive Psychology, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Laboratory of Neurodegenerative Diseases, Center for Interdisciplinary Research and Innovation (CIRI), Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Greek Association of Alzheimer’s Disease and Related Disorders (GAADRD), 54643 Thessaloniki, Greece
| | - Magdalini Tsolaki
- Laboratory of Neurodegenerative Diseases, Center for Interdisciplinary Research and Innovation (CIRI), Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Greek Association of Alzheimer’s Disease and Related Disorders (GAADRD), 54643 Thessaloniki, Greece
| |
Collapse
|
31
|
The impact of cerebral vasomotor reactivity on cerebrovascular diseases and cognitive impairment. J Neural Transm (Vienna) 2022; 129:1321-1330. [PMID: 36205784 PMCID: PMC9550758 DOI: 10.1007/s00702-022-02546-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022]
Abstract
The regulation of cerebral blood flow (CBF) is a complex and tightly controlled function ensuring delivery of oxygen and nutrients and removal of metabolic wastes from brain tissue. Cerebral vasoreactivity (CVR) refers to the ability of the nervous system to regulate CBF according to metabolic demands or changes in the microenvironment. This can be assessed through a variety of nuclear medicine and imaging techniques and protocols. Several studies have investigated the association of CVR with physiological and pathological conditions, with particular reference to the relationship with cognitive impairment and cerebrovascular disorders (CVD). A better understanding of the interaction between CVR and cognitive dysfunction in chronic and particularly acute CVD could help improving treatment and rehabilitation strategies in these patients. In this paper, we reviewed current knowledge on CVR alterations in the context of acute and chronic CVD and cognitive dysfunction. Alterations in CVR and hemodynamics have been described in patients with both neurodegenerative and vascular cognitive impairment, and the severity of these alterations seems to correlate with CVR derailment. Furthermore, an increased risk of cognitive impairment progression has been associated with alterations in CVR parameters and hemodynamics. Few studies have investigated these associations in acute cerebrovascular disorders and the results are inconsistent; thus, further research on this topic is encouraged.
Collapse
|
32
|
Chen TY, Chan PC, Tsai CF, Wei CY, Chiu PY. White matter hyperintensities in dementia with Lewy bodies are associated with poorer cognitive function and higher dementia stages. Front Aging Neurosci 2022; 14:935652. [PMID: 36092817 PMCID: PMC9459160 DOI: 10.3389/fnagi.2022.935652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
Purpose White matter hyperintensities (WMHs) are frequently found in elderly individuals with or without dementia. However, the association between WMHs and clinical presentations of dementia with Lewy bodies (DLB) has rarely been studied. Methods We conducted a retrospective analysis of patients with DLB registered in a dementia database. WMHs were rated visually using the Fazekas scale, and its associated factors including dementia severity, cognitive functions, neuropsychiatric symptoms, and core clinical features were compared among different Fazekas scores. Domains in the Clinical Dementia Rating (CDR), Cognitive abilities Screening Instruments (CASI), and Neuropsychiatric Inventory (NPI) were compared among different Fazekas groups after adjusting for age, sex, education, and disease duration. Results Among the 449 patients, 76, 207, 110, and 56 had Fazekas score of 0, 1, 2, and 3, respectively. There was a positive association between dementia severity and WMHs severity, and the mean sums of boxes of the Clinical Dementia Rating (CDR-SB) were 5.9, 7.8, 9.5, and 11.2 (f = 16.84, p < 0.001) for the Fazekas scale scores 0, 1, 2, and 3, respectively. There was a negative association between cognitive performance and WMHs severity, and the mean CASI were 57.7, 45.4, 4.06, and 33.4 (f = 14.22, p < 0.001) for the Fazekas scale scores 0, 1, 2, and 3, respectively. However, WMHs were not associated with the core clinical features of DLB. After adjustment, all cognitive domains in CDR increased as the Fazekas score increased. In addition, performance on all cognitive domains in CASI decreased as the Fazekas score increased (all p < 0.001). Among neuropsychiatric symptoms, delusions, euphoria, apathy, aberrant motor behavior, and sleep disorders were significantly worse in the higher Fazekas groups compared to those in the group with Fazekas score of 0 after adjustment. Conclusion WMHs in DLB might contribute to deterioration of cognitive function, neuropsychiatric symptoms, and dementia stages. However, core clinical features were not significantly influenced by WMHs in DLB.
Collapse
Affiliation(s)
- Tai-Yi Chen
- Department of Radiology, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Po-Chi Chan
- Department of Neurology, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Ching-Fang Tsai
- Tainan Sin-Lau Hospital, The Presbyterian Church in Taiwan, Tainan, Taiwan
| | - Cheng-Yu Wei
- Department of Neurology, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Pai-Yi Chiu
- Department of Neurology, Show Chwan Memorial Hospital, Changhua, Taiwan
- Department of Applied Mathematics, Tunghai University, Taichung, Taiwan
- *Correspondence: Pai-Yi Chiu,
| |
Collapse
|
33
|
Yu W, Li Y, Hu J, Wu J, Huang Y. A Study on the Pathogenesis of Vascular Cognitive Impairment and Dementia: The Chronic Cerebral Hypoperfusion Hypothesis. J Clin Med 2022; 11:jcm11164742. [PMID: 36012981 PMCID: PMC9409771 DOI: 10.3390/jcm11164742] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
The pathogenic mechanisms underlying vascular cognitive impairment and dementia (VCID) remain controversial due to the heterogeneity of vascular causes and complexity of disease neuropathology. However, one common feature shared among all these vascular causes is cerebral blood flow (CBF) dysregulation, and chronic cerebral hypoperfusion (CCH) is the universal consequence of CBF dysregulation, which subsequently results in an insufficient blood supply to the brain, ultimately contributing to VCID. The purpose of this comprehensive review is to emphasize the important contributions of CCH to VCID and illustrate the current findings about the mechanisms involved in CCH-induced VCID pathological changes. Specifically, evidence is mainly provided to support the molecular mechanisms, including Aβ accumulation, inflammation, oxidative stress, blood-brain barrier (BBB) disruption, trophic uncoupling and white matter lesions (WMLs). Notably, there are close interactions among these multiple mechanisms, and further research is necessary to elucidate the hitherto unsolved questions regarding these interactions. An enhanced understanding of the pathological features in preclinical models could provide a theoretical basis, ultimately achieving the shift from treatment to prevention.
Collapse
Affiliation(s)
- Weiwei Yu
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
| | - Yao Li
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
| | - Jun Hu
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
| | - Jun Wu
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
- Correspondence: (J.W.); (Y.H.); Tel.: +86-0755-8392-2833 (J.W.); +86-010-83572857 (Y.H.)
| | - Yining Huang
- Department of Neurology, Peking University First Hospital, 8 Xishiku Street Xicheng District, Beijing 100034, China
- Correspondence: (J.W.); (Y.H.); Tel.: +86-0755-8392-2833 (J.W.); +86-010-83572857 (Y.H.)
| |
Collapse
|
34
|
Jenkins TA. Metabolic Syndrome and Vascular-Associated Cognitive Impairment: a Focus on Preclinical Investigations. Curr Diab Rep 2022; 22:333-340. [PMID: 35737273 PMCID: PMC9314301 DOI: 10.1007/s11892-022-01475-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW Metabolic syndrome is associated with an increased risk of vascular cognitive impairment or, in the more extreme, vascular dementia. Animal models are used to investigate the relationship between pathology and behaviour. This review summarizes the latest understanding of the role of the hippocampus and prefrontal cortex in vascular cognitive impairment, the influence of inflammation in this association while also commenting on some of the latest interventions proposed. RECENT FINDINGS Models of vascular cognitive impairment and vascular dementia, whether they develop from an infarct or non-infarct base, demonstrate increased neuroinflammation, reduced neuronal function and deficits in prefrontal and hippocampal-associated cognitive domains. Promising new research shows agents and environmental interventions that inhibit central oxidative stress and inflammation can reverse both pathology and cognitive dysfunction. While preclinical studies suggest that reversal of deficits in vascular cognitive impairment models is possible, replication in patients still needs to be demonstrated.
Collapse
Affiliation(s)
- Trisha A Jenkins
- Human Biosciences, School of Health and Biomedical Sciences, STEM College, RMIT University, Plenty Road, Bundoora, VIC, 3083, Australia.
| |
Collapse
|
35
|
Silva NCBS, Dao E, Hsu CL, Tam RC, Stein R, Alkeridy W, Laule C, Vavasour IM, Liu-Ambrose T. Myelin Content and Gait Impairment in Older Adults with Cerebral Small Vessel Disease and Mild Cognitive Impairment. Neurobiol Aging 2022; 119:56-66. [DOI: 10.1016/j.neurobiolaging.2022.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/19/2022] [Accepted: 03/15/2022] [Indexed: 11/25/2022]
|
36
|
Mitroi DN, Tian M, Kawaguchi R, Lowry WE, Carmichael ST. Single-nucleus transcriptome analysis reveals disease- and regeneration-associated endothelial cells in white matter vascular dementia. J Cell Mol Med 2022; 26:3183-3195. [PMID: 35543222 PMCID: PMC9170821 DOI: 10.1111/jcmm.17315] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 02/01/2022] [Accepted: 03/12/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Vascular dementia (VaD) is the accumulation of vascular lesions in the subcortical white matter of the brain. These lesions progress and there is no direct medical therapy. AIMS To determine the specific cellular responses in VaD so as to provide molecular targets for therapeutic development. MATERIALS AND METHODS Single-nucleus transcriptome analysis was performed in human periventricular white matter (PVWM) samples of VaD and normal control (NC) subjects. RESULTS Differential analysis shows that cell type-specific transcriptomic changes in VaD are associated with the disruption of specific biological processes, including angiogenesis, immune activation, axonal injury and myelination. Each cell type in the neurovascular unit within white matter has a specific alteration in gene expression in VaD. In a central cell type for this disease, subcluster analysis of endothelial cells (EC) indicates that VaD contains a disease-associated EC subcluster that expresses genes associated with programmed cell death and a response to protein folding. Two other subpopulations of EC in VaD express molecular systems associated with regenerative processes in angiogenesis, and in axonal sprouting and oligodendrocyte progenitor cell maturation. CONCLUSION This comprehensive molecular profiling of brain samples from patients with VaD reveals previously unknown molecular changes in cells of the neurovascular niche, and an attempt at regeneration in injured white matter.
Collapse
Affiliation(s)
- Daniel N. Mitroi
- Department of Psychiatry and Biobehavioral SciencesDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Min Tian
- Department of Psychiatry and Biobehavioral SciencesDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Riki Kawaguchi
- Department of Psychiatry and Biobehavioral SciencesDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - William E. Lowry
- Department of Molecular, Cell and Developmental BiologyUCLALos AngelesCaliforniaUSA
| | - S. Thomas Carmichael
- Department of Psychiatry and Biobehavioral SciencesDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| |
Collapse
|
37
|
Wang J, Zhou Y, He Y, Li Q, Zhang W, Luo Z, Xue R, Lou M. Impact of different white matter hyperintensities patterns on cognition: A cross-sectional and longitudinal study. Neuroimage Clin 2022; 34:102978. [PMID: 35255417 PMCID: PMC8897653 DOI: 10.1016/j.nicl.2022.102978] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVES White matter hyperintensities (WMH) are highly prevalent in older adults and considered to be a contributor to cognition impairment. However, the strategic WMH lesion distribution related to cognitive impairment is still debated. The aim of this study was to characterize the spatial patterns of WMH associated with cognitive impairment and explore its risk factors. METHODS We retrospectively analyzed patients who underwent T2 fluid attenuated inversion recovery (FLAIR) and mini-mental state examination (MMSE) in two centers. WHM was classified into four patterns based on T2 FLAIR as follows: (1) multiple subcortical spots (multi-spots); (2) peri-basal ganglia (peri-BG); (3) anterior subcortical patches (anterior SC patches); and (4) posterior subcortical patches (posterior SC patches). We cross-sectionally and longitudinally estimated associations between different WMH patterns and all-cause dementia and cognitive decline. Multivariable logistic regression analysis was followed to identify risk factors of WMH patterns related to cognitive impairment. RESULTS A total of 442 patients with WMH were enrolled, with average age of 71.6 ± 11.3 years, and MMSE score of 24.1 ± 5.4. Among them, 281 (63.6%), 66 (14.9%), 163 (36.9%) and 197 (44.6%) patients presented multi-spots, peri-BG, anterior SC patches and posterior SC patches, respectively. Patients with anterior SC patches were more likely to have all-cause dementia in cross-sectional study (OR 2.002; 95% CI 1.098-3.649; p = 0.024), and have cognitive decline in longitudinal analysis (OR 3.029; 95% CI 1.270-7.223; p = 0.012). Four patterns of WMH referred to different cognitive domains, and anterior SC patches had the most significant and extensive impact on cognition after Bonferroni multiple comparison correction (all p < 0.0125). In addition, older age (OR 1.054; 95% CI 1.027-1.082; p < 0.001), hypertension (OR 1.956; 95% CI 1.145-3.341; p = 0.014), higher percentage of neutrophils (OR 1.046; 95% CI 1.014-1.080; p = 0.005) and lower concentration of hemoglobin (OR 0.983; 95% CI 0.967-1.000; p = 0.044) were risk factors for the presence of anterior SC patches. CONCLUSIONS Different patterns of subcortical leukoaraiosis visually identified on MRI might have different impacts on cognitive impairment. Further studies should be undertaken to validate this simple visual classification of WMH in different population.
Collapse
Affiliation(s)
- Junjun Wang
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China; Department of Neurology, Zhejiang Hospital, #12 Lingyin Road, Hangzhou, China
| | - Ying Zhou
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China
| | - Yaode He
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China
| | - Qingqing Li
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China
| | - Wenhua Zhang
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China
| | - Zhongyu Luo
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China
| | - Rui Xue
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China
| | - Min Lou
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China.
| |
Collapse
|
38
|
Li J, Zhu Z, Li Y, Chen Y, Hu X, Liu Y, Shi Y, Hu Y, Bi Y, Xu X, Zheng M, Cheng L, Jing J. D-4F, an apolipoprotein A-I mimetic, promotes the clearance of myelin debris and the reduction of foamy macrophages after spinal cord injury. Bioengineered 2022; 13:11794-11809. [PMID: 35546071 PMCID: PMC9276047 DOI: 10.1080/21655979.2022.2073063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
After spinal cord injury (SCI), a large number of blood-derived macrophages infiltrate the lesion site and phagocytose myelin debris to become foamy macrophages, which leads to chronic inflammation. The drug D-4F, an apolipoprotein A-I peptidomimetic made of D-amino acids, has been reported to promote the lipid metabolism of foamy macrophages in atherosclerosis. However, the role and mechanism of D-4F in SCI are still unclear. In this study, we found that D-4F can promote the removal of myelin debris, reduce the formation of foamy macrophages in the lesion core and promote neuroprotection and recovery of motor function after SCI. These beneficial functions of D-4F may be related to its ability to upregulate the expression of ATP-binding cassette transporter A1 (ABCA1), the main transporter that mediates lipid efflux in foamy macrophages because inhibiting the activity of ABCA1 can reverse the effect of D-4F in vitro. In conclusion, D-4F may be a promising candidate for treating SCI by promoting the clearance of myelin debris by foamy macrophages via the ABCA1 pathway.
Collapse
Affiliation(s)
- Jinxin Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhenyu Zhu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yiteng Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yihao Chen
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xuyang Hu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yanchang Liu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yi Shi
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yao Hu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yihui Bi
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xinzhong Xu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Meige Zheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Li Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Juehua Jing
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
39
|
Torres-Simón L, Doval S, Nebreda A, Llinas SJ, Marsh EB, Maestú F. Understanding brain function in vascular cognitive impairment and dementia with EEG and MEG: A systematic review. Neuroimage Clin 2022; 35:103040. [PMID: 35653914 PMCID: PMC9163840 DOI: 10.1016/j.nicl.2022.103040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/09/2022] [Accepted: 05/06/2022] [Indexed: 11/22/2022]
Abstract
Vascular Cognitive Impairment (VCI) is the second most prevalent dementia after Alzheimer's Disease (AD), and cerebrovascular disease (CBVD) is a major comorbid contributor to the progression of most neurodegenerative diseases. Early differentiation of cognitive impairment is critical given both the high prevalence of CBVD, and that its risk factors are modifiable. The ability for electroencephalogram (EEG) and magnetoencephalogram (MEG) to detect changes in brain functioning for other dementias suggests that they may also be promising biomarkers for early VCI. The present systematic review aims to summarize the literature regarding electrophysiological patterns of mild and major VCI. Despite considerable heterogeneity in clinical definition and electrophysiological methodology, common patterns exist when comparing patients with VCI to healthy controls (HC) and patients with AD, though there is a low specificity when comparing between VCI subgroups. Similar to other dementias, slowed frequency patterns and disrupted inter- and intra-hemispheric connectivity are repeatedly reported for VCI patients, as well as longer latencies and smaller amplitudes in evoked responses. Further study is needed to fully establish MEG and EEG as clinically useful biomarkers, including a clear definition of VCI and standardized methodology, allowing for comparison across groups and consolidation of multicenter efforts.
Collapse
Affiliation(s)
- Lucía Torres-Simón
- Center of Cognitive and Computational Neuroscience; Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain.
| | - Sandra Doval
- Center of Cognitive and Computational Neuroscience; Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain
| | - Alberto Nebreda
- Center of Cognitive and Computational Neuroscience; Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain
| | - Sophia J Llinas
- Department of Neurology, the Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Elisabeth B Marsh
- Department of Neurology, the Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Fernando Maestú
- Center of Cognitive and Computational Neuroscience; Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
40
|
Xie S, Yang J, Huang S, Fan Y, Xu T, He J, Guo J, Ji X, Wang Z, Li P, Chen J, Zhang Y. Disrupted myelination network in the cingulate cortex of Parkinson's disease. IET Syst Biol 2022; 16:98-119. [PMID: 35394697 PMCID: PMC9290774 DOI: 10.1049/syb2.12043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/31/2022] [Accepted: 03/21/2022] [Indexed: 12/13/2022] Open
Abstract
The cingulate cortex is part of the conserved limbic system, which is considered as a hub of emotional and cognitive control. Accumulating evidence suggested that involvement of the cingulate cortex is significant for cognitive impairment of Parkinson's disease (PD). However, mechanistic studies of the cingulate cortex in PD pathogenesis are limited. Here, transcriptomic and regulatory network analyses were conducted for the cingulate cortex in PD. Enrichment and clustering analyses showed that genes involved in regulation of membrane potential and glutamate receptor signalling pathway were upregulated. Importantly, myelin genes and the oligodendrocyte development pathways were markedly downregulated, indicating disrupted myelination in PD cingulate cortex. Cell‐type‐specific signatures revealed that myelinating oligodendrocytes were the major cell type damaged in the PD cingulate cortex. Furthermore, downregulation of myelination pathways in the cingulate cortex were shared and validated in another independent RNAseq cohort of dementia with Lewy bodies (DLB). In combination with ATACseq data, gene regulatory networks (GRNs) were further constructed for 32 transcription factors (TFs) and 466 target genes among differentially expressed genes (DEGs) using a tree‐based machine learning algorithm. Several transcription factors, including Olig2, Sox8, Sox10, E2F1, and NKX6‐2, were highlighted as key nodes in a sub‐network, which control many overlapping downstream targets associated with myelin formation and gliogenesis. In addition, the authors have validated a subset of DEGs by qPCRs in two PD mouse models. Notably, seven of these genes,TOX3, NECAB2 NOS1, CAPN3, NR4A2, E2F1 and FOXP2, have been implicated previously in PD or neurodegeneration and are worthy of further studies as novel candidate genes. Together, our findings provide new insights into the role of remyelination as a promising new approach to treat PD after demyelination.
Collapse
Affiliation(s)
- Song Xie
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiajun Yang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Shenghui Huang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yuanlan Fan
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Tao Xu
- Molecular Neuropharmacology Lab, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,The Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang Province, China
| | - Jiangshuang He
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiahao Guo
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xiang Ji
- Department of Mathematics, School of Science & Engineering, Tulane University, New Orleans, Louisiana, USA
| | - Zhibo Wang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Peijun Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiangfan Chen
- Molecular Neuropharmacology Lab, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,The Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang Province, China
| | - Yi Zhang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,The Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang Province, China.,Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
41
|
Yu H, Shi J, Lin Y, Zhang Y, Luo Q, Huang S, Wang S, Wei J, Huang J, Li C, Ji L. Icariin Ameliorates Alzheimer's Disease Pathology by Alleviating Myelin Injury in 3 × Tg-AD Mice. Neurochem Res 2022; 47:1049-1059. [PMID: 35037164 DOI: 10.1007/s11064-021-03507-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 11/09/2021] [Accepted: 12/07/2021] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by excessive deposition of β amyloid (Aβ), hyperphosphorylation of tau protein, and neuronal cell death. Recent studies have shown that myelin cell damage, which leads to cognitive dysfunction, occurs before AD-related pathological changes. Here, we examine the effect of icariin (ICA), a prenylated flavonol glycoside, in improving cognitive function in AD model mice. ICA has been reported to exhibit cardiovascular protective functions and antiaging effects. In this study, we used 3 × Tg-AD mice as an AD model. The Morris water maze and Y maze tests were performed to assess the learning and memory of the mice. Immunofluorescence analysis of Aβ1-42 deposition and myelin basic protein (MBP) expression in the mouse hippocampus was performed. Tau protein phosphorylation and MBP protein expression in the hippocampus were further analyzed by Western blotting. Myelin damage in the mouse optic nerve was evaluated by electron microscopy, and LFB staining was performed to assess myelin morphology in the mouse corpus callosum. MBP, Mpp5, and Egr2 transcript levels were quantified by qPCR. We observed that ICA treatment improved the learning and memory of 3 × Tg-AD mice and reduced Aβ deposition and tau protein phosphorylation in the hippocampus. Moreover, this treatment protocol increased myelin-related gene expression and reduced myelin damage.
Collapse
Affiliation(s)
- Hongxia Yu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jianhong Shi
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yiyou Lin
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yehui Zhang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qihang Luo
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Suo Huang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Sichen Wang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jiale Wei
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Junhao Huang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Changyu Li
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Liting Ji
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
42
|
Mossanen Parsi M, Duval C, Ariëns RAS. Vascular Dementia and Crosstalk Between the Complement and Coagulation Systems. Front Cardiovasc Med 2021; 8:803169. [PMID: 35004913 PMCID: PMC8733168 DOI: 10.3389/fcvm.2021.803169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/29/2021] [Indexed: 01/12/2023] Open
Abstract
Vascular Dementia (VaD) is a neurocognitive disorder caused by reduced blood flow to the brain tissue, resulting in infarction, and is the second most common type of dementia. The complement and coagulation systems are evolutionary host defence mechanisms activated by acute tissue injury to induce inflammation, clot formation and lysis; recent studies have revealed that these systems are closely interlinked. Overactivation of these systems has been recognised to play a key role in the pathogenesis of neurological disorders such as Alzheimer's disease and multiple sclerosis, however their role in VaD has not yet been extensively reviewed. This review aims to bridge the gap in knowledge by collating current understanding of VaD to enable identification of complement and coagulation components involved in the pathogenesis of this disorder that may have their effects amplified or supressed by crosstalk. Exploration of these mechanisms may unveil novel therapeutic targets or biomarkers that would improve current treatment strategies for VaD.
Collapse
Affiliation(s)
| | | | - Robert A. S. Ariëns
- Discovery and Translational Science Department, School of Medicine, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
43
|
Hijazi Z, Yassi N, O'Brien JT, Watson R. The influence of cerebrovascular disease in dementia with Lewy bodies and Parkinson's disease dementia. Eur J Neurol 2021; 29:1254-1265. [PMID: 34923713 DOI: 10.1111/ene.15211] [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: 11/03/2021] [Accepted: 12/08/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Lewy body dementia (LBD), including dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD), is a common form of neurodegenerative dementia. The frequency and influence of comorbid cerebrovascular disease is not understood but has potentially important clinical management implications. METHODS A systematic literature search was conducted (Medline and Embase) for studies including participants with DLB and/or PDD assessing cerebrovascular lesions (imaging and pathological studies). They included white matter changes, cerebral amyloid angiopathy (CAA), cerebral microbleeds (CMB), macroscopic infarcts, micro-infarcts and intracerebral haemorrhage. RESULTS Of 4411 articles, 63 studies were included. Cerebrovascular lesions commonly studied included white matter changes (41 studies) and CMB (18 studies). There was an increased severity of white matter changes on magnetic resonance imaging (visualized as white matter hyperintensities, WMH), but not neuropathology, in LBD compared to PD without dementia and age-matched controls. CMB prevalence in DLB was highly variable but broadly similar to Alzheimer's disease (AD) (0-48%), with a lobar predominance. No relationship was found between large cortical or small subcortical infarcts or intracerebral haemorrhage and presence of LBD. CONCLUSION The underlying mechanisms of WMH in LBD require further exploration, as their increased severity in LBD was not supported by neuropathological examination of white matter. CMB in LBD had a similar prevalence as AD. There is a need for larger studies assessing the influence of cerebrovascular lesions on clinical symptoms, disease progression and outcomes.
Collapse
Affiliation(s)
- Zina Hijazi
- Monash University School of Rural Health, Bendigo Hospital, Bendigo, VIC, Australia.,Department of Medicine, Bendigo Hospital, Bendigo, VIC, Australia
| | - Nawaf Yassi
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.,Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Neurology, Melbourne Brain Centre at The Royal Melbourne Hospital, University of Melbourne, Parkville, Australia
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge, Level E4, Box 189, Cambridge, CB2 0QC, UK
| | - Rosie Watson
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.,Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| |
Collapse
|
44
|
Miyoshi E, Morabito S, Swarup V. Systems biology approaches to unravel the molecular and genetic architecture of Alzheimer's disease and related tauopathies. Neurobiol Dis 2021; 160:105530. [PMID: 34634459 PMCID: PMC8616667 DOI: 10.1016/j.nbd.2021.105530] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/30/2021] [Accepted: 10/07/2021] [Indexed: 11/19/2022] Open
Abstract
Over the years, genetic studies have identified multiple genetic risk variants associated with neurodegenerative disorders and helped reveal new biological pathways and genes of interest. However, genetic risk variants commonly reside in non-coding regions and may regulate distant genes rather than the nearest gene, as well as a gene's interaction partners in biological networks. Systems biology and functional genomics approaches provide the framework to unravel the functional significance of genetic risk variants in disease. In this review, we summarize the genetic and transcriptomic studies of Alzheimer's disease and related tauopathies and focus on the advantages of performing systems-level analyses to interrogate the biological pathways underlying neurodegeneration. Finally, we highlight new avenues of multi-omics analysis with single-cell approaches, which provide unparalleled opportunities to systematically explore cellular heterogeneity, and present an example of how to integrate publicly available single-cell datasets. Systems-level analysis has illuminated the function of many disease risk genes, but much work remains to study tauopathies and to understand spatiotemporal gene expression changes of specific cell types.
Collapse
Affiliation(s)
- Emily Miyoshi
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA 92697, USA
| | - Samuel Morabito
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA 92697, USA; Mathematical, Computational and Systems Biology (MCSB) Program, University of California, Irvine, CA 92697, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA 92697, USA.
| |
Collapse
|
45
|
Kalaria RN, Sepulveda-Falla D. Cerebral Small Vessel Disease in Sporadic and Familial Alzheimer Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1888-1905. [PMID: 34331941 PMCID: PMC8573679 DOI: 10.1016/j.ajpath.2021.07.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 06/15/2021] [Accepted: 07/02/2021] [Indexed: 01/26/2023]
Abstract
Alzheimer disease (AD) is the most common cause of dementia. Biological definitions of AD are limited to the cerebral burden of amyloid β plaques, neurofibrillary pathology, and neurodegeneration. However, current evidence suggests that various features of small vessel disease (SVD) are part of and covertly modify both sporadic and familial AD. Neuroimaging studies suggest that white matter hyperintensities explained by vascular mechanisms occurs frequently in the AD spectrum. Recent advances have further emphasized that frontal periventricular and posterior white matter hyperintensities are associated with cerebral amyloid angiopathy in familial AD. Although whether SVD markers precede the classically recognized biomarkers of disease is debatable, post-mortem studies show that SVD pathology incorporating small cortical and subcortical infarcts, microinfarcts, microbleeds, perivascular spacing, and white matter attenuation is commonly found in sporadic as well as in mutation carriers with confirmed familial AD. Age-related cerebral vessel pathologies such as arteriolosclerosis and cerebral amyloid angiopathy modify progression or worsen risk by shifting the threshold for cognitive impairment and AD dementia. The incorporation of SVD as a biomarker is warranted in the biological definition of AD. Therapeutic interventions directly reducing the burden of brain amyloid β have had no major impact on the disease or delaying cognitive deterioration, but lowering the risk of vascular disease seems the only rational approach to tackle both early- and late-onset AD dementia.
Collapse
Affiliation(s)
- Rajesh N Kalaria
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom; Department of Human Anatomy, College of Health Sciences, University of Nairobi, Nairobi, Kenya.
| | - Diego Sepulveda-Falla
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
46
|
Santiago-Mujika E, Luthi-Carter R, Giorgini F, Kalaria RN, Mukaetova-Ladinska EB. Tubulin and Tubulin Posttranslational Modifications in Alzheimer's Disease and Vascular Dementia. Front Aging Neurosci 2021; 13:730107. [PMID: 34776926 PMCID: PMC8586541 DOI: 10.3389/fnagi.2021.730107] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/04/2021] [Indexed: 01/26/2023] Open
Abstract
Alzheimer's disease (AD) and vascular dementia (VaD) are the two most common forms of dementia in older people. Although these two dementia types differ in their etiology, they share many pathophysiological and morphological features, including neuronal loss, which is associated with the microtubule (MT) destabilization. Stabilization of MTs is achieved in different ways: through interactions with MT binding proteins (MTBP) or by posttranslational modifications (PTMs) of tubulin. Polyglutamylation and tyrosination are two foremost PTMs that regulate the interaction between MTs and MTBPs, and play, therefore, a role in neurodegeneration. In this review, we summarize key information on tubulin PTMs in relation to AD and VaD and address the importance of studying further the tubulin code to reveal sites of potential intervention in development of novel and effective dementia therapy.
Collapse
Affiliation(s)
- Estibaliz Santiago-Mujika
- Department of Neuroscience, Behavior and Psychology, University of Leicester, Leicester, United Kingdom
| | - Ruth Luthi-Carter
- Department of Neuroscience, Behavior and Psychology, University of Leicester, Leicester, United Kingdom
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Raj N. Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Elizabeta B. Mukaetova-Ladinska
- Department of Neuroscience, Behavior and Psychology, University of Leicester, Leicester, United Kingdom
- Evington Centre, Leicester General Hospital, Leicester, United Kingdom
| |
Collapse
|
47
|
18F-florbetapir PET as a marker of myelin integrity across the Alzheimer's disease spectrum. Eur J Nucl Med Mol Imaging 2021; 49:1242-1253. [PMID: 34581847 PMCID: PMC8921113 DOI: 10.1007/s00259-021-05493-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 07/08/2021] [Indexed: 01/23/2023]
Abstract
Purpose Recent evidence suggests that PET imaging with amyloid-β (Aβ) tracers can be used to assess myelin integrity in cerebral white matter (WM). Alzheimer’s disease (AD) is characterized by myelin changes that are believed to occur early in the disease course. Nevertheless, the extent to which demyelination, as measured with Aβ PET, contributes to AD progression remains unexplored. Methods Participants with concurrent 18F-florbetapir (FBP) PET, MRI, and cerebrospinal fluid (CSF) examinations were included (241 cognitively normal, 347 Aβ-positive cognitively impaired, and 207 Aβ-negative cognitively impaired subjects). A subset of these participants had also available diffusion tensor imaging (DTI) images (n = 195). We investigated cross-sectional associations of FBP retention in the white matter (WM) with MRI-based markers of WM degeneration, AD clinical progression, and fluid biomarkers. In longitudinal analyses, we used linear mixed models to assess whether FBP retention in normal-appearing WM (NAWM) predicted progression of WM hyperintensity (WMH) burden and clinical decline. Results In AD-continuum individuals, FBP retention in NAWM was (1) higher compared with WMH regions, (2) associated with DTI-based measures of WM integrity, and (3) associated with longitudinal progression of WMH burden. FBP uptake in WM decreased across the AD continuum and with increasingly abnormal CSF biomarkers of AD. Furthermore, FBP retention in the WM was associated with large-calibre axon degeneration as reflected by abnormal plasma neurofilament light chain levels. Low FBP uptake in NAWM predicted clinical decline in preclinical and prodromal AD, independent of demographics, global cortical Aβ, and WMH burden. Most of these associations were also observed in Aβ-negative cognitively impaired individuals. Conclusion These results support the hypothesis that FBP retention in the WM is myelin-related. Demyelination levels progressed across the AD continuum and were associated with clinical progression at early stages, suggesting that this pathologic process might be a relevant degenerative feature in the disease course. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05493-y.
Collapse
|
48
|
Hussain B, Fang C, Chang J. Blood-Brain Barrier Breakdown: An Emerging Biomarker of Cognitive Impairment in Normal Aging and Dementia. Front Neurosci 2021; 15:688090. [PMID: 34489623 PMCID: PMC8418300 DOI: 10.3389/fnins.2021.688090] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
The blood–brain barrier (BBB) plays a vital role in maintaining the specialized microenvironment of the neural tissue. It separates the peripheral circulatory system from the brain parenchyma while facilitating communication. Alterations in the distinct physiological properties of the BBB lead to BBB breakdown associated with normal aging and various neurodegenerative diseases. In this review, we first briefly discuss the aging process, then review the phenotypes and mechanisms of BBB breakdown associated with normal aging that further cause neurodegeneration and cognitive impairments. We also summarize dementia such as Alzheimer's disease (AD) and vascular dementia (VaD) and subsequently discuss the phenotypes and mechanisms of BBB disruption in dementia correlated with cognition decline. Overlaps between AD and VaD are also discussed. Techniques that could identify biomarkers associated with BBB breakdown are briefly summarized. Finally, we concluded that BBB breakdown could be used as an emerging biomarker to assist to diagnose cognitive impairment associated with normal aging and dementia.
Collapse
Affiliation(s)
- Basharat Hussain
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Cheng Fang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Junlei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
49
|
Ferreira S, Pitman KA, Summers BS, Wang S, Young KM, Cullen CL. Oligodendrogenesis increases in hippocampal grey and white matter prior to locomotor or memory impairment in an adult mouse model of tauopathy. Eur J Neurosci 2021; 54:5762-5784. [PMID: 32181929 PMCID: PMC8451881 DOI: 10.1111/ejn.14726] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022]
Abstract
Myelin and axon losses are associated with cognitive decline in healthy ageing but are worse in people diagnosed with tauopathy. To determine whether tauopathy is also associated with enhanced myelin plasticity, we evaluated the behaviour of OPCs in mice that expressed a human pathological variant of microtubule-associated protein tau (MAPTP301S ). By 6 months of age (P180), MAPTP301S mice overexpressed hyperphosphorylated tau and had developed reactive gliosis in the hippocampus but had not developed overt locomotor or memory impairment. By performing cre-lox lineage tracing of adult OPCs, we determined that the number of newborn oligodendrocytes added to the hippocampus, entorhinal cortex and fimbria was equivalent in control and MAPTP301S mice prior to P150. However, between P150 and P180, significantly more new oligodendrocytes were added to these regions in the MAPTP301S mouse brain. This large increase in new oligodendrocyte number was not the result of increased OPC proliferation, nor did it alter oligodendrocyte density in the hippocampus, entorhinal cortex or fimbria, which was equivalent in P180 wild-type and MAPTP301S mice. Furthermore, the proportion of hippocampal and fimbria axons with myelin was unaffected by tauopathy. However, the proportion of myelinated axons that were ensheathed by immature myelin internodes was significantly increased in the hippocampus and fimbria of P180 MAPTP301S mice, when compared with their wild-type littermates. These data suggest that MAPTP301S transgenic mice experience significant oligodendrocyte turnover, with newborn oligodendrocytes compensating for myelin loss early in the development of tauopathy.
Collapse
Affiliation(s)
- Solène Ferreira
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmaniaAustralia
| | - Kimberley A. Pitman
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmaniaAustralia
| | - Benjamin S. Summers
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmaniaAustralia
| | - Shiwei Wang
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmaniaAustralia
| | - Kaylene M. Young
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmaniaAustralia
| | - Carlie L. Cullen
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmaniaAustralia
| |
Collapse
|
50
|
Ding R, Hase Y, Burke M, Foster V, Stevenson W, Polvikoski T, Kalaria RN. Loss with ageing but preservation of frontal cortical capillary pericytes in post-stroke dementia, vascular dementia and Alzheimer's disease. Acta Neuropathol Commun 2021; 9:130. [PMID: 34340718 PMCID: PMC8330023 DOI: 10.1186/s40478-021-01230-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/16/2021] [Indexed: 01/19/2023] Open
Abstract
Cerebral pericytes are an integral component of the neurovascular unit, which governs the blood–brain barrier. There is paucity of knowledge on cortical pericytes across different dementias. We quantified cortical pericytes in capillaries in 124 post-mortem brains from subjects with post-stroke dementia (PSD), vascular dementia (VaD), Alzheimer’s disease (AD) and AD-VaD (Mixed) and, post-stroke non-demented (PSND) stroke survivors as well as normal ageing controls. Collagen 4 (COL4)-positive nucleated pericyte soma were identified as protrusions on capillaries of the frontal cortex. The COL4-positive somata or nodule-like cell bodies were also verified by platelet derived growth factor receptor-β (PDGFR-β) immunohistochemistry. The mean (± SEM) pericyte somata in frontal cortical capillaries in normal young controls (46–65 years of age) was estimated as 5.2 ± 0.2 per mm capillary length. This number was reduced by 45% in older controls (> 78 years) to 2.9 ± 0.1 per mm capillary length (P < 0.001). We further found that the numbers of pericyte cell bodies per COL4 mm2 area or per mm capillary length were not decreased but rather preserved or increased in PSD, AD and Mixed dementia groups compared to similar age older controls (P < 0.01). Consistent with this, we noted that capillary length densities identified by the endothelial marker glucose transporter 1 or COL4 were not different across the dementias compared to older controls. There was a negative correlation with age (P < 0.001) suggesting fewer pericyte somata in older age, although the % COL4 immunoreactive capillary area was increased in older controls compared to young controls. Using a proven reliable method to quantify COL4-positive nucleated pericytes, our observations demonstrate ageing related loss but mostly preserved pericytes in the frontal cortex of vascular and AD dementias. We suggest there is differential regulation of capillary pericytes in the frontal lobe between the cortex and white matter in ageing-related dementias.
Collapse
|