1
|
Azizi N, Issaiy M, Jalali AH, Kolahi S, Naghibi H, Zarei D, Firouznia K. Perfusion-weighted MRI patterns in neuropsychiatric systemic lupus erythematosus: a systematic review and meta-analysis. Neuroradiology 2025; 67:109-124. [PMID: 39230717 DOI: 10.1007/s00234-024-03457-1] [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: 06/11/2024] [Accepted: 08/19/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Neuropsychiatric Systemic Lupus Erythematosus (NPSLE) is a complex manifestation of Systemic Lupus Erythematosus (SLE) characterized by a wide range of neurological and psychiatric symptoms. This study aims to elucidate the patterns of Perfusion-Weighted MRI (PWI) in NPSLE patients compared to SLE patients without neuropsychiatric manifestations (non-NPSLE) and healthy controls (HCs). MATERIAL AND METHODS A systematic search was conducted in PubMed/Medline, Embase, Web of Science, and Scopus for studies utilizing PWI in NPSLE patients published through April 14, 2024. Cerebral blood flow (CBF) data from NPSLE, non-NPSLE patients, and HCs were extracted for meta-analysis, using standardized mean difference (SMD) as an estimate measure. For studies lacking sufficient data for inclusion, CBF, cerebral blood volume (CBV), and mean transit time (MTT) were reviewed qualitatively. RESULTS Our review included eight observational studies employing PWI techniques, including dynamic susceptibility contrast (DSC) and arterial spin labeling (ASL). The meta-analysis of NPSLE compared to non-NPSLE incorporated four studies, encompassing 104 NPSLE patients and 90 non-NPSLE patients. The results revealed an SMD of -1.42 (95% CI: -2.85-0.00, I2: 94%) for CBF in NPSLE compared to non-NPSLE. CONCLUSION PWI reveals informative patterns of cerebral perfusion, showing a significant reduction in mean CBF in NPSLE patients compared to non-NPSLE patients. Our qualitative synthesis highlights these changes, particularly in the frontal and temporal lobes. However, the existing data exhibits considerable heterogeneity and limitations.
Collapse
Affiliation(s)
- Narges Azizi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Imam Khomeini Hospital Complex (IKHC), 2nd Floor, Keshavarz Boulevard, Tehran, Iran
| | - Mahbod Issaiy
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Imam Khomeini Hospital Complex (IKHC), 2nd Floor, Keshavarz Boulevard, Tehran, Iran
| | - Amir Hossein Jalali
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Imam Khomeini Hospital Complex (IKHC), 2nd Floor, Keshavarz Boulevard, Tehran, Iran
| | - Shahriar Kolahi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Imam Khomeini Hospital Complex (IKHC), 2nd Floor, Keshavarz Boulevard, Tehran, Iran
| | - Hamed Naghibi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Imam Khomeini Hospital Complex (IKHC), 2nd Floor, Keshavarz Boulevard, Tehran, Iran
| | - Diana Zarei
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Imam Khomeini Hospital Complex (IKHC), 2nd Floor, Keshavarz Boulevard, Tehran, Iran
| | - Kavous Firouznia
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Imam Khomeini Hospital Complex (IKHC), 2nd Floor, Keshavarz Boulevard, Tehran, Iran.
| |
Collapse
|
2
|
van Dinther M, Voorter PHM, Zhang E, van Kuijk SMJ, Jansen JFA, van Oostenbrugge RJ, Backes WH, Staals J. The neurovascular unit and its correlation with cognitive performance in patients with cerebral small vessel disease: a canonical correlation analysis approach. GeroScience 2024; 46:5061-5073. [PMID: 38888875 PMCID: PMC11335703 DOI: 10.1007/s11357-024-01235-8] [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: 09/29/2023] [Accepted: 05/31/2024] [Indexed: 06/20/2024] Open
Abstract
Growing evidence indicates an important role of neurovascular unit (NVU) dysfunction in the pathophysiology of cerebral small vessel disease (cSVD). Individually measurable functions of the NVU have been correlated with cognitive function, but a combined analysis is lacking. We aimed to perform a unified analysis of NVU function and its relation with cognitive performance. The relationship between NVU function in the white matter and cognitive performance (both latent variables composed of multiple measurable variables) was investigated in 73 patients with cSVD (mean age 70 ± 10 years, 41% women) using canonical correlation analysis. MRI-based NVU function measures included (1) the intravoxel incoherent motion derived perfusion volume fraction (f) and microvascular diffusivity (D*), reflecting cerebral microvascular flow; (2) the IVIM derived intermediate volume fraction (fint), indicative of the perivascular clearance system; and (3) the dynamic contrast-enhanced MRI derived blood-brain barrier (BBB) leakage rate (Ki) and leakage volume fraction (VL), reflecting BBB integrity. Cognitive performance was composed of 13 cognitive test scores. Canonical correlation analysis revealed a strong correlation between the latent variables NVU function and cognitive performance (r 0.73; p = 0.02). For the NVU, the dominating variables were D*, fint, and Ki. Cognitive performance was driven by multiple cognitive tests comprising different cognitive domains. The functionality of the NVU is correlated with cognitive performance in cSVD. Instead of focusing on individual pathophysiological mechanisms, future studies should target NVU dysfunction as a whole to acquire a coherent understanding of the complex disease mechanisms that occur in the NVU in cSVD.Trial registration: NTR3786 (Dutch Trial Register).
Collapse
Affiliation(s)
- Maud van Dinther
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands.
- CARIM-School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands.
| | - Paulien H M Voorter
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- MHeNs-School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Eleana Zhang
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Sander M J van Kuijk
- Department of Epidemiology and Medical Technology Assessment (KEMTA), Maastricht University, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- MHeNs-School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Robert J van Oostenbrugge
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM-School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
- MHeNs-School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Walter H Backes
- CARIM-School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- MHeNs-School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Julie Staals
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM-School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
3
|
Kalantari S, Soltani M, Maghbooli M, Khoshe Mehr F, Kalantari Z, Borji S, Memari B, Hossein Heydari A, Elahi R, Bayat M, Salighehrad H. Alteraciones del flujo sanguíneo cerebral medidas con RM-ASL como predictor de demencia vascular en la enfermedad isquémica de pequeño vaso. RADIOLOGIA 2024. [DOI: 10.1016/j.rx.2024.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Wang Y, Wang T, Yu Z, Wang J, Liu F, Ye M, Fang X, Liu Y, Liu J. Alterations in structural integrity of superior longitudinal fasciculus III associated with cognitive performance in cerebral small vessel disease. BMC Med Imaging 2024; 24:138. [PMID: 38858645 PMCID: PMC11165890 DOI: 10.1186/s12880-024-01324-2] [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: 12/01/2023] [Accepted: 06/05/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND This study aimed to investigate the alterations in structural integrity of superior longitudinal fasciculus subcomponents with increasing white matter hyperintensity severity as well as the relationship to cognitive performance in cerebral small vessel disease. METHODS 110 cerebral small vessel disease study participants with white matter hyperintensities were recruited. According to Fazekas grade scale, white matter hyperintensities of each subject were graded. All subjects were divided into two groups. The probabilistic fiber tracking method was used for analyzing microstructure characteristics of superior longitudinal fasciculus subcomponents. RESULTS Probabilistic fiber tracking results showed that mean diffusion, radial diffusion, and axial diffusion values of the left arcuate fasciculus as well as the mean diffusion value of the right arcuate fasciculus and left superior longitudinal fasciculus III in high white matter hyperintensities rating group were significantly higher than those in low white matter hyperintensities rating group (p < 0.05). The mean diffusion value of the left superior longitudinal fasciculus III was negatively related to the Montreal Cognitive Assessment score of study participants (p < 0.05). CONCLUSIONS The structural integrity injury of bilateral arcuate fasciculus and left superior longitudinal fasciculus III is more severe with the aggravation of white matter hyperintensities. The structural integrity injury of the left superior longitudinal fasciculus III correlates to cognitive impairment in cerebral small vessel disease.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Radiology, Eye& ENT Hospital of Shanghai Medical School, Fudan University, Shanghai, China
| | - Tianyao Wang
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zekuan Yu
- Academy for Engineering and Technology, Fudan University, Shanghai, 200433, China
| | - Junjie Wang
- Department of Neurosurgery, Beijing Hospital, National Center of Gerontology, Beijing, China
- Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Fang Liu
- Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Mengwen Ye
- Academy for Engineering and Technology, Fudan University, Shanghai, 200433, China
| | - Xianjin Fang
- Anhui University of Science and Technology, Anhui, China
| | - Yinhong Liu
- Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Beijing, China.
| | - Jun Liu
- Department of Radiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 XianXia Road, Shanghai, 200050, China.
| |
Collapse
|
5
|
van Dinther M, Hooghiemstra AM, Bron EE, Versteeg A, Leeuwis AE, Kalay T, Moonen JE, Kuipers S, Backes WH, Jansen JFA, van Osch MJP, Biessels G, Staals J, van Oostenbrugge RJ. Lower cerebral blood flow predicts cognitive decline in patients with vascular cognitive impairment. Alzheimers Dement 2024; 20:136-144. [PMID: 37491840 PMCID: PMC10917014 DOI: 10.1002/alz.13408] [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: 03/07/2023] [Revised: 06/26/2023] [Accepted: 07/03/2023] [Indexed: 07/27/2023]
Abstract
INTRODUCTION Chronic cerebral hypoperfusion is one of the assumed pathophysiological mechanisms underlying vascular cognitive impairment (VCI). We investigated the association between baseline cerebral blood flow (CBF) and cognitive decline after 2 years in patients with VCI and reference participants. METHODS One hundred eighty-one participants (mean age 66.3 ± 7.4 years, 43.6% women) underwent arterial spin labeling (ASL) magnetic resonance imaging (MRI) and neuropsychological assessment at baseline and at 2-year follow-up. We determined the association between baseline global and lobar CBF and cognitive decline with multivariable regression analysis. RESULTS Lower global CBF at baseline was associated with more global cognitive decline in VCI and reference participants. This association was most profound in the domain of attention/psychomotor speed. Lower temporal and frontal CBF at baseline were associated with more cognitive decline in memory. DISCUSSION Our study supports the role of hypoperfusion in the pathophysiological and clinical progression of VCI. HIGHLIGHTS Impaired cerebral blood flow (CBF) at baseline is associated with faster cognitive decline in VCI and normal aging. Our results suggest that low CBF precedes and contributes to the development of vascular cognitive impairment. CBF determined by ASL might be used as a biomarker to monitor disease progression or treatment responses in VCI.
Collapse
Affiliation(s)
- Maud van Dinther
- Department of NeurologyMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Astrid M. Hooghiemstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Esther E. Bron
- Department of Radiology & Nuclear MedicineErasmus MC—University Medical Center RotterdamRotterdamThe Netherlands
| | - Adriaan Versteeg
- Department of Radiology & Nuclear MedicineErasmus MC—University Medical Center RotterdamRotterdamThe Netherlands
| | - Anna E. Leeuwis
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Old Age PsychiatryGGZ inGeestAmsterdamThe Netherlands
| | - Tugba Kalay
- Department of NeurologySt. Antonius ZiekenhuisNieuwegeinThe Netherlands
| | - Justine E. Moonen
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Sanne Kuipers
- Department of NeurologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Walter H. Backes
- Department of Radiology and Nuclear MedicineMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Jacobus F. A. Jansen
- Department of Radiology and Nuclear MedicineMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Mathias J. P. van Osch
- C.J. Gorter MRI Center, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Geert‐Jan Biessels
- Department of NeurologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Julie Staals
- Department of NeurologyMaastricht University Medical CenterMaastrichtThe Netherlands
| | | | | |
Collapse
|
6
|
Li W, Jiang J, Yin X, Zhang Y, Zou X, Sun M, Jia J, Ma B, Xu J. Mediation of Regional Cerebral Blood Flow in the Relationship between Specific Gut Microbiota and Cognition in Vascular Cognitive Impairment. J Alzheimers Dis 2024; 97:435-445. [PMID: 38108351 DOI: 10.3233/jad-230709] [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] [Indexed: 12/19/2023]
Abstract
BACKGROUND Gut microbiota could affect the onset and development of vascular cognitive impairment (VCI) through modulating metabolic and immune pathways. However, the vascular mechanisms involved remain unclear. OBJECTIVE To investigate the gut microbiota associated with VCI and examine the mediating effects of regional cerebral blood flow (CBF) to explore potential therapeutic targets for VCI. METHODS This prospective study enrolled patients with VCI (n = 16) and healthy controls (n = 18) from the Chinese Imaging, Biomarkers, and Lifestyle study between January 1 and June 30, 2022. The gut microbiota composition and diversity were determined by 16 S ribosomal RNA gene sequencing. The association between gut microbiota and Montreal Cognitive Assessment (MoCA) scores was determined using Spearman's correlation analysis. Regional CBF was calculated using pseudo-continuous arterial spin labeling. The mediating effects of regional CBF on the relationship between specific gut microbiota and cognition in VCI were investigated using mediation analysis. RESULTS Compared to healthy controls, patients with VCI had significantly greater abundance of Bifidobacterium, Veillonella, R uminococcus gnavus , Fusobacterium, and Erysipelatoclostridium and smaller abundance of Collinsella. The abundance of Ruminococcus gnavus was negatively associated with MoCA scores in patients with VCI, with the CBF in the left hypothalamus, right hypothalamus, and left amygdala accounting for 63.96%, 48.22%, and 36.51%, respectively, of this association after adjusting for confounders. CONCLUSIONS Ruminococcus gnavus is associated with cognition in VCI, which is strongly mediated by CBF in the bilateral hypothalamus and left amygdala. These findings highlight the potential regulatory roles of nutrition and metabolism-related areas of the brain in VCI.
Collapse
Affiliation(s)
- Wenyi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jiwei Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | | | - Yuan Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xinying Zou
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Mengfan Sun
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jianjun Jia
- Department of Geriatric Neurology, The Second Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Baiping Ma
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
7
|
Ruan Z, Sun D, Zhou X, Yu M, Li S, Sun W, Li Y, Gao L, Xu H. Altered neurovascular coupling in patients with vascular cognitive impairment: a combined ASL-fMRI analysis. Front Aging Neurosci 2023; 15:1224525. [PMID: 37416325 PMCID: PMC10320594 DOI: 10.3389/fnagi.2023.1224525] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/08/2023] [Indexed: 07/08/2023] Open
Abstract
Background and objective This study aims to examine the role of neurovascular coupling (NVC) in vascular cognitive impairment (VCI) by investigating the relationship between white matter lesion (WML) burden, NVC, and cognitive deficits. Additionally, we aim to explore the potential of NVC as a tool for understanding the neural mechanisms underlying VCI. Methods This study included thirty-eight small vessel disease cognitive impairment (SVCI) patients, 34 post-stroke cognitive impairment (PSCI) patients, and 43 healthy controls (HC). Comprehensive assessments, including neuroimaging and neuropsychological testing, were conducted to evaluate cognitive function. WML burden was measured and correlated with NVC coefficients to examine the relationship between white matter pathology and NVC. Mediation analysis was employed to explore the link relationship between NVC, WML burden, and cognitive function. Results The present study showed that NVC was significantly reduced in the SVCI and PSCI groups compared with HCs at both whole-brain and brain region level. The analysis revealed notable findings regarding NVC in relation to WML burden and cognitive function in VCI patients. Specifically, reduced NVC coefficients were observed within higher order brain systems responsible for cognitive control and emotion regulation. Mediation analysis demonstrated that NVC played a mediating role in the relationship between WML burden and cognitive impairment. Conclusion This study reveals the mediating role of NVC in the relationship between WML burden and cognitive function in VCI patients. The results demonstrate the potential of the NVC as an accurate measure of cognitive impairment and its ability to identify specific neural circuits affected by WML burden.
Collapse
Affiliation(s)
- Zhao Ruan
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Dong Sun
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiaoli Zhou
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Minhua Yu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Sirui Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wenbo Sun
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yidan Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lei Gao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
8
|
Huang D, Guo Y, Guan X, Pan L, Zhu Z, Chen Z, Dijkhuizen RM, Duering M, Yu F, Boltze J, Li P. Recent advances in arterial spin labeling perfusion MRI in patients with vascular cognitive impairment. J Cereb Blood Flow Metab 2023; 43:173-184. [PMID: 36284489 PMCID: PMC9903225 DOI: 10.1177/0271678x221135353] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/01/2022] [Accepted: 09/21/2022] [Indexed: 01/24/2023]
Abstract
Cognitive impairment (CI) is a major health concern in aging populations. It impairs patients' independent life and may progress to dementia. Vascular cognitive impairment (VCI) encompasses all cerebrovascular pathologies that contribute to cognitive impairment (CI). Moreover, the majority of CI subtypes involve various aspects of vascular dysfunction. Recent research highlights the critical role of reduced cerebral blood flow (CBF) in the progress of VCI, and the detection of altered CBF may help to detect or even predict the onset of VCI. Arterial spin labeling (ASL) is a non-invasive, non-ionizing perfusion MRI technique for assessing CBF qualitatively and quantitatively. Recent methodological advances enabling improved signal-to-noise ratio (SNR) and data acquisition have led to an increase in the use of ASL to assess CBF in VCI patients. Combined with other imaging modalities and biomarkers, ASL has great potential for identifying early VCI and guiding prediction and prevention strategies. This review focuses on recent advances in ASL-based perfusion MRI for identifying patients at high risk of VCI.
Collapse
Affiliation(s)
- Dan Huang
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunlu Guo
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyu Guan
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijun Pan
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyu Zhu
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeng’ai Chen
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Marco Duering
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Germany
- Medical Image Analysis Center (MIAC) and qbig, Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Fang Yu
- Department of Anesthesiology, Westchester Medical Center, New York Medical College, NY, USA
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Peiying Li
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Altered Cerebral Blood Flow in the Progression of Chronic Kidney Disease. J Pers Med 2023; 13:jpm13010142. [PMID: 36675803 PMCID: PMC9863421 DOI: 10.3390/jpm13010142] [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: 11/27/2022] [Revised: 12/25/2022] [Accepted: 12/27/2022] [Indexed: 01/13/2023] Open
Abstract
Background: In chronic kidney disease (CKD), cognitive impairment is a definite complication. However, the mechanisms of how CKD leads to cognitive impairment are not clearly known. Methods: Cerebral blood flow (CBF) information was collected from 37 patients with CKD (18 in stage 3; 19 in stage 4) and 31 healthy controls (HCs). For CKD patients, we also obtained laboratory results as well as neuropsychological tests. We conducted brain perfusion imaging studies using arterial spin labeling and calculated the relationship between regional CBF changes and various clinical indicators and neuropsychological tests. We also generated receiver operator characteristic (ROC) curves to explore whether CBF value changes in certain brain regions can be used to identify CKD. Results: Compared with HCs, CBF decreased in the right insula and increased in the left hippocampus in the CKD4 group; through partial correlation analysis, we found that CBF in the right insula was negatively correlated with the number connection test A (NCT-A) (r = −0.544, p = 0.024); CBF in the left hippocampus was positively correlated with blood urea nitrogen (r = 0.649, p = 0.005) and negatively correlated with serum calcium level (r = −0.646, p = 0.005). By comparing the ROC curve area, it demonstrated that altered CBF values in the right insula (AUC = 0.861, p < 0.01) and left hippocampus (AUC = 0.862, p < 0.01) have a good ability to identify CKD. Conclusions: Our study found that CBF alterations in the left hippocampus and the right insula brain of adult patients with stage 4 CKD were correlated with disease severity or laboratory indicators. These findings provide further insight into the relationship between altered cerebral perfusion and cognitive impairment in patients with non-end-stage CKD as well as, additional information the underlying neuropathophysiological mechanisms.
Collapse
|
10
|
Zhou J, He J, Wang W. Application of 3D-ASL in hemodynamic analysis and prognosis evaluation of vascular cognitive impairment. Am J Transl Res 2022; 14:7960-7968. [PMID: 36505282 PMCID: PMC9730099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/30/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To investigate the application of magnetic resonance 3D arterial spin labeling (3D-ASL) imaging in the hemodynamic analysis and prognostic assessment of vascular cognitive impairment (VCI). METHODS Using a retrospective research method, 108 patients with ischemic cerebrovascular disease diagnosed in the Department of Neurology of Lianyungang Hospital of Traditional Chinese Medicine from January 2021 to April 2022 were chose as the research subjects. The Montreal cognitive assessment (MoCA) was used to evaluate cognitive function. The patients were divided into a VCI group (n=54, 28 males and 26 females) and a normal cognitive function group (NCF group, n=54, 30 males and 24 females). The 3D-ASL cerebral perfusion imaging was performed on the two groups of patients using different post label delay (PLD) (1525 ms, 2525 ms). The cerebral blood flow (CBF) values of bilateral frontal lobe, temporal lobe, temporal parietal junction, parietal lobe, and hippocampus were analyzed under different PLDs in the two groups. The two sets of MoCA scale scores were compared. The receiver operating characteristic curve (ROC) of CBF of VCI patients was drawn, and the area under curve (AUC), specificity and sensitivity under different PLDs was compared. RESULTS There was no statistical significance between the two groups in terms of sex, average age, hypertension, diabetes, coronary heart disease, smoking history, and drinking history (P>0.05). CBF 1525 values and CBF 2525 values in the bilateral frontal lobes, temporal lobes, temporoparietal junction, parietal lobes, and hippocampus were significantly reduced in the VCI group under different PLD (all P<0.05). There was no significant difference in the CBF 1525 value and CBF 2525 value of the bilateral frontal lobe and temporal lobe in the VCI group (all P<0.05). The language, delayed memory, executive ability, attention and calculation ability, naming, abstract thinking, orientation, and total scores of the VCI group were significantly lower than those of the NCF group (all P<0.05). The ROC analysis revealed that the AUC, specificity, and sensitivity of CBF (bilateral frontal, temporal, temporoparietal junction, parietal, and hippocampus) at PLD 1525 ms were lower than those of CBF at PLD 2525 ms (P<0.05). CONCLUSION Non-invasive 3D-ASL technology can be used to detect cerebral hemodynamics and predict prognosis in VCI patients. PLD 1525 ms was more sensitive to detect cerebral hypoperfusion. PLD 2525 ms showed a more accurate hypoperfusion range. This guides and adjusts treatment methods.
Collapse
Affiliation(s)
- Jianguo Zhou
- Department of Radiology, Lianyungang Hospital of Traditional Chinese MedicineLianyungang 222004, Jiangsu, China
| | - Jiangtao He
- Department of Radiology, The Fourth People’s Hospital of Lianyungang, Affiliated Hospital of Nanjing Medical University Kangda CollegeLianyungang 222000, Jiangsu, China
| | - Wei Wang
- Department of Radiology, Xuyi People’s HospitalHuai’an 211700, Jiangsu, China
| |
Collapse
|
11
|
Zhang W, Li M, Zhou X, Huang C, Wan K, Li C, Yin J, Zhao W, Zhang C, Zhu X, Sun Z. Altered serum amyloid beta and cerebral perfusion and their associations with cognitive function in patients with subcortical ischemic vascular disease. Front Neurosci 2022; 16:993767. [PMID: 36312019 PMCID: PMC9608371 DOI: 10.3389/fnins.2022.993767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/08/2022] [Indexed: 01/07/2024] Open
Abstract
Subcortical ischemic vascular disease (SIVD) is one of the important causes of cognitive dysfunction, altered amyloid-beta (Aβ) and cerebral perfusion may be involved in the pathophysiological mechanism of SIVD and are closely related to cognitive function. We aimed to investigate altered serum Aβ and cerebral perfusion in patients with SIVD and their correlation with cognitive function. Seventy-four healthy controls (HCs) and 74 SIVD patients, including 38 SIVD patients with no cognitive impairment (SIVD-NCI) and 36 SIVD patients with mild cognitive impairment (SIVD-MCI) underwent the measurement of serum Aβ40 and Aβ42 levels, pseudo-continuous arterial spin labeling MRI scanning, and cognitive evaluation. Compared to the healthy controls (HCs), the level of serum Aβ40 and Aβ40/42 ratio increased and Aβ42 decreased in SIVD patients. The serum Aβ40 level and Aβ40/42 ratio in patients with SIVD-MCI were significantly higher than those in the HCs and SIVD-NCI, and the level of Aβ42 in the SIVD-MCI was lower than the HCs. In addition, the serum Aβ40/42 ratio provided high diagnostic accuracy for SIVD and SIVD-MCI, it was further identified as an independent risk factor for cognitive impairment. Patients with SIVD-NCI and SIVD-MCI exhibited both increased and decreased cerebral blood flow (CBF) in regional. The Aβ40/42 ratio was associated with global CBF, while altered global and regional CBF was associated with cognitive deficits. In addition, white matter hyperintensities volume (WMHV) correlated with Aβ40/42 ratio, CBF, and cognition. The relationship between Aβ40/42 ratio and cognition was partially mediated by altered CBF. Based on these results, we conclude that the serum Aβ40/42 ratio may be a potential biomarker that can complement current methods for the prediction and diagnosis of cognitive impairment in SIVD patients. In addition, serum Aβ may play a role in cognitive function by regulating CBF, which provides new insights into the intervention, treatment, and prevention of cognitive impairment in SIVD.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mingxu Li
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xia Zhou
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chaojuan Huang
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ke Wan
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chenchen Li
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiabin Yin
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenming Zhao
- Department of Radiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cun Zhang
- Department of Radiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaoqun Zhu
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhongwu Sun
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
12
|
Gyanwali B, Tan CS, Petr J, Escobosa LLT, Vrooman H, Chen C, Mutsaerts HJ, Hilal S. Arterial Spin-Labeling Parameters and Their Associations with Risk Factors, Cerebral Small-Vessel Disease, and Etiologic Subtypes of Cognitive Impairment and Dementia. AJNR Am J Neuroradiol 2022; 43:1418-1423. [PMID: 36562454 PMCID: PMC9575536 DOI: 10.3174/ajnr.a7630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 07/01/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND PURPOSE Cerebral small-vessel disease may alter cerebral blood flow (CBF) leading to brain changes and, hence, cognitive impairment and dementia. CBF and the spatial coefficient of variation can be measured quantitatively by arterial spin-labeling. We aimed to investigate the associations of demographics, vascular risk factors, location, and severity of cerebral small-vessel disease as well as the etiologic subtypes of cognitive impairment and dementia with CBF and the spatial coefficient of variation. MATERIALS AND METHODS Three hundred ninety patients with a diagnosis of no cognitive impairment, cognitive impairment no dementia, vascular cognitive impairment no dementia, Alzheimer disease, and vascular dementia were recruited from the memory clinic. Cerebral microbleeds and lacunes were categorized into strictly lobar, strictly deep, and mixed-location and enlarged perivascular spaces into the centrum semiovale and basal ganglia. Total and region-specific white matter hyperintensity volumes were segmented using FreeSurfer. CBF (n = 333) and the spatial coefficient of variation (n = 390) were analyzed with ExploreASL from 2D-EPI pseudocontinuous arterial spin-labeling images in white matter (WM) and gray matter (GM). To analyze the effect of demographic and vascular risk factors as well as the location and severity of cerebral small-vessel disease markers on arterial spin-labeling parameters, we constructed linear regression models, whereas logistic regression models were used to determine the association between arterial spin-labeling parameters and cognitive impairment no dementia, vascular cognitive impairment no dementia, Alzheimer disease, and vascular dementia. RESULTS Increasing age, male sex, hypertension, hyperlipidemia, history of heart disease, and smoking were associated with lower CBF and a higher spatial coefficient of variation. Higher numbers of lacunes and cerebral microbleeds were associated with lower CBF and a higher spatial coefficient of variation. Location-specific analysis showed mixed-location lacunes and cerebral microbleeds were associated with lower CBF. Higher total, anterior, and posterior white matter hyperintensity volumes were associated with a higher spatial coefficient of variation. No association was observed between enlarged perivascular spaces and arterial spin-labeling parameters. A higher spatial coefficient of variation was associated with the diagnosis of vascular cognitive impairment no dementia, Alzheimer's disease, and vascular dementia. CONCLUSIONS Reduced CBF and an increased spatial coefficient of variation were associated with cerebral small-vessel disease, and more specifically lacunes, whereas cerebral microbleeds and white matter hyperintensities were associated with WM-CBF and GM spatial coefficient of variation. The spatial coefficient of variation was associated with cognitive impairment and dementia, suggesting that hypoperfusion might be the key underlying mechanism for vascular brain damage.
Collapse
Affiliation(s)
- B Gyanwali
- From the Memory Aging and Cognition Centre (B.G., C.C., S.H.), National University Health System, Singapore
| | - C S Tan
- Saw Swee Hock School of Public Health (C.S.T., L.L.T.E., S.H.), National University of Singapore, and National University Health System, Singapore
| | - J Petr
- Helmholtz-Zentrum Dresden-Rossendorf (J.P.), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - L L T Escobosa
- Saw Swee Hock School of Public Health (C.S.T., L.L.T.E., S.H.), National University of Singapore, and National University Health System, Singapore
| | - H Vrooman
- Department of Radiology and Nuclear Medicine (H.V.), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - C Chen
- From the Memory Aging and Cognition Centre (B.G., C.C., S.H.), National University Health System, Singapore
- Department of Pharmacology (C.C., S.H.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - H J Mutsaerts
- Department of Radiology (H.J.M.), VU University Medical Center, Amsterdam, the Netherlands
- Department of Radiology (H.J.M.), Brain Center Rudolf Magnus, University Medical Center, Utrecht, the Netherlands
| | - S Hilal
- From the Memory Aging and Cognition Centre (B.G., C.C., S.H.), National University Health System, Singapore
- Saw Swee Hock School of Public Health (C.S.T., L.L.T.E., S.H.), National University of Singapore, and National University Health System, Singapore
- Department of Pharmacology (C.C., S.H.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
13
|
Mahammedi A, Wang LL, Williamson BJ, Khatri P, Kissela B, Sawyer RP, Shatz R, Khandwala V, Vagal A. Small Vessel Disease, a Marker of Brain Health: What the Radiologist Needs to Know. AJNR Am J Neuroradiol 2022; 43:650-660. [PMID: 34620594 PMCID: PMC9089248 DOI: 10.3174/ajnr.a7302] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 07/05/2021] [Indexed: 11/07/2022]
Abstract
Small vessel disease, a disorder of cerebral microvessels, is an expanding epidemic and a common cause of stroke and dementia. Despite being almost ubiquitous in brain imaging, the clinicoradiologic association of small vessel disease is weak, and the underlying pathogenesis is poorly understood. The STandards for ReportIng Vascular changes on nEuroimaging (STRIVE) criteria have standardized the nomenclature. These include white matter hyperintensities of presumed vascular origin, recent small subcortical infarcts, lacunes of presumed vascular origin, prominent perivascular spaces, cerebral microbleeds, superficial siderosis, cortical microinfarcts, and brain atrophy. Recently, the rigid categories among cognitive impairment, vascular dementia, stroke, and small vessel disease have become outdated, with a greater emphasis on brain health. Conventional and advanced small vessel disease imaging markers allow a comprehensive assessment of global brain heath. In this review, we discuss the pathophysiology of small vessel disease neuroimaging nomenclature by means of the STRIVE criteria, clinical implications, the role of advanced imaging, and future directions.
Collapse
Affiliation(s)
- A Mahammedi
- From the Departments of Neuroradiology (A.M., L.L.W., B.J.W., V.K., A.V.)
| | - L L Wang
- From the Departments of Neuroradiology (A.M., L.L.W., B.J.W., V.K., A.V.)
| | - B J Williamson
- From the Departments of Neuroradiology (A.M., L.L.W., B.J.W., V.K., A.V.)
| | - P Khatri
- Neurology (P.K., B.K., R.P.S., R.S.) University of Cincinnati Medical Center, Cincinnati, Ohio
| | - B Kissela
- Neurology (P.K., B.K., R.P.S., R.S.) University of Cincinnati Medical Center, Cincinnati, Ohio
| | - R P Sawyer
- Neurology (P.K., B.K., R.P.S., R.S.) University of Cincinnati Medical Center, Cincinnati, Ohio
| | - R Shatz
- Neurology (P.K., B.K., R.P.S., R.S.) University of Cincinnati Medical Center, Cincinnati, Ohio
| | - V Khandwala
- From the Departments of Neuroradiology (A.M., L.L.W., B.J.W., V.K., A.V.)
| | - A Vagal
- From the Departments of Neuroradiology (A.M., L.L.W., B.J.W., V.K., A.V.)
| |
Collapse
|
14
|
Zhang L, Li Y, Bian L, Luo Q, Zhang X, Zhao B. Cognitive Impairment of Patient With Neurological Cerebrovascular Disease Using the Artificial Intelligence Technology Guided by MRI. Front Public Health 2022; 9:813641. [PMID: 35310781 PMCID: PMC8927700 DOI: 10.3389/fpubh.2021.813641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
This study was to explore the application of MRI based on artificial intelligence technology combined with neuropsychological assessment to the cognitive impairment of patients with neurological cerebrovascular diseases. A total of 176 patients were divided into a control group, a vascular cognitive impairment non-dementia (VCIND) group, a vascular dementia (VD) group, and an Alzheimer's disease (AD) group. All patients underwent MRI and neuropsychological evaluation and examination, and an improved fuzzy C-means (FCM) clustering algorithm was proposed for MRI processing. It was found that the segmentation accuracy (SA) and similarity (KI) data of the improved FCM algorithm used in this study were higher than those of the standard FCM algorithm, bias-corrected FCM (BCFCM) algorithm, and rough FCM (RFCM) algorithm (p < 0.05). In the activities of daily living (ADL), the values in the VCIND group (23.55 ± 6.12) and the VD group (28.56 ± 3.1) were higher than that in the control group (19.17 ± 3.67), so the hippocampal volume was negatively correlated with the ADL (r = −0.872, p < 0.01). In the VCIND group (52.4%), VD group (31%), and AD group (26.1%), the proportion of patients with the lacunar infarction distributed on both sides of the brain and the number of multiple cerebral infarction lesions (76.2, 71.4, and 71.7%, respectively) were significantly higher than those in the control group (23.9 and 50%). In short, the improved FCM algorithm showed a higher segmentation effect and SA for MRI of neurological cerebrovascular disease. In addition, the distribution, number, white matter lesions, and hippocampal volume of lacunar cerebral infarction were related to the cognitive impairment of patients with cerebrovascular diseases.
Collapse
Affiliation(s)
- Lifang Zhang
- Department of Neurology, Changzhi People's Hospital, Changzhi Medical College, Changzhi, China
- Department of Mental Health, Changzhi Medical College, Changzhi, China
- *Correspondence: Lifang Zhang
| | - Yanran Li
- Department of Radiology, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Lin Bian
- Department of Neurology, Changzhi People's Hospital, Changzhi Medical College, Changzhi, China
| | - Qingrong Luo
- Department of Neurology, Changzhi People's Hospital, Changzhi Medical College, Changzhi, China
| | - Xiaoxi Zhang
- Department of Mental Health, Changzhi Medical College, Changzhi, China
| | - Bing Zhao
- Department of Neurology, Changzhi People's Hospital, Changzhi Medical College, Changzhi, China
| |
Collapse
|
15
|
Tu MC, Chung HW, Hsu YH, Yang JJ, Wu WC. Stage-Dependent Cerebral Blood Flow and Leukoaraiosis Couplings in Subcortical Ischemic Vascular Disease and Alzheimer's Disease. J Alzheimers Dis 2022; 86:729-739. [PMID: 35124651 PMCID: PMC9028753 DOI: 10.3233/jad-215405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background: Alzheimer’s disease (AD) and subcortical ischemic vascular disease (SIVD) have both been associated with white matter hyperintensities (WMHs) and altered cerebral blood flow (CBF) although the etiology of AD is still unclear. Objective: To test the hypothesis that CBF and WMHs have differential effects on cognition and that the relationship between CBF and WMHs changes with the subtypes and stages of dementia. Methods: Forty-two patients with SIVD, 50 patients with clinically-diagnosed AD, and 30 cognitively-normal subjects were included. Based on the Clinical Dementia Rating (CDR), the patients were dichotomized into early-stage (CDR = 0.5) and late-stage (CDR = 1 or 2) groups. CBF and WMH metrics were derived from magnetic resonance imaging and correlated with cognition. Results: Hierarchical linear regression revealed that CBF metrics had distinct contribution to global cognition, memory, and attention, whereas WMH metrics had distinct contribution to executive function (all p < 0.05). In SIVD, the WMHs in frontotemporal areas correlated with the CBF in bilateral thalami at the early stage; the correlation then became between the WMHs in basal ganglia and the CBF in frontotemporal areas at the late stage. A similar corticosubcortical coupling was observed in AD but involved fewer areas. Conclusion: A stage-dependent coupling between CBF and WMHs was identified in AD and SIVD, where the extent of cortical WMHs correlated with subcortical CBF for CDR = 0.5, whereas the extent of subcortical WMHs correlated with cortical CBF for CDR = 1–2.
Collapse
Affiliation(s)
- Min-Chien Tu
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan.,Department of Neurology, Taichung Tzu Chi Hospital, Taichung, Taiwan.,Department of Neurology, Tzu Chi University, Hualien, Taiwan
| | - Hsiao-Wen Chung
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Yen-Hsuan Hsu
- Department of Psychology, National Chung Cheng University, Chiayi, Taiwan.,Center for Innovative Research on Aging Society, National Chung Cheng University, Chiayi, Taiwan
| | - Jir-Jei Yang
- Department of Radiology, Taichung Tzu Chi Hospital, Taichung, Taiwan
| | - Wen-Chau Wu
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan.,Institute of Medical Device and Imaging, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
16
|
Xu W, Song Y, Chen S, Xue C, Hu G, Qi W, Ma W, Lin X, Chen J. An ALE Meta-Analysis of Specific Functional MRI Studies on Subcortical Vascular Cognitive Impairment. Front Neurol 2021; 12:649233. [PMID: 34630270 PMCID: PMC8492914 DOI: 10.3389/fneur.2021.649233] [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] [Received: 01/06/2021] [Accepted: 07/28/2021] [Indexed: 11/23/2022] Open
Abstract
Background: Subcortical vascular cognitive impairment (sVCI), caused by cerebral small vessel disease, accounts for the majority of vascular cognitive impairment, and is characterized by an insidious onset and impaired memory and executive function. If not recognized early, it inevitably develops into vascular dementia. Several quantitative studies have reported the consistent results of brain regions in sVCI patients that can be used to predict dementia conversion. The purpose of the study was to explore the exact abnormalities within the brain in sVCI patients by combining the coordinates reported in previous studies. Methods: The PubMed, Embase, and Web of Science databases were thoroughly searched to obtain neuroimaging articles on the amplitude of low-frequency fluctuation, regional homogeneity, and functional connectivity in sVCI patients. According to the activation likelihood estimation (ALE) algorithm, a meta-analysis based on coordinate and functional connectivity modeling was conducted. Results: The quantitative meta-analysis included 20 functional imaging studies on sVCI patients. Alterations in specific brain regions were mainly concentrated in the frontal lobes including the middle frontal gyrus, superior frontal gyrus, medial frontal gyrus, and precentral gyrus; parietal lobes including the precuneus, angular gyrus, postcentral gyrus, and inferior parietal lobule; occipital lobes including the lingual gyrus and cuneus; temporal lobes including the fusiform gyrus and middle temporal gyrus; and the limbic system including the cingulate gyrus. These specific brain regions belonged to important networks known as the default mode network, the executive control network, and the visual network. Conclusion: The present study determined specific abnormal brain regions in sVCI patients, and these brain regions with specific changes were found to belong to important brain functional networks. The findings objectively present the exact abnormalities within the brain, which help further understand the pathogenesis of sVCI and identify them as potential imaging biomarkers. The results may also provide a basis for new approaches to treatment.
Collapse
Affiliation(s)
- Wenwen Xu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Song
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Shanshan Chen
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Chen Xue
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Guanjie Hu
- Institute of Brain Functional Imaging, Nanjing Medical University, Nanjing, China
| | - Wenzhang Qi
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Wenying Ma
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Xingjian Lin
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jiu Chen
- Institute of Brain Functional Imaging, Nanjing Medical University, Nanjing, China.,Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
17
|
Liu S, Hou B, You H, Zhang Y, Zhu Y, Ma C, Zuo Z, Feng F. The Association Between Perivascular Spaces and Cerebral Blood Flow, Brain Volume, and Cardiovascular Risk. Front Aging Neurosci 2021; 13:599724. [PMID: 34531732 PMCID: PMC8438293 DOI: 10.3389/fnagi.2021.599724] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Basal ganglia perivascular spaces are associated with cognitive decline and cardiovascular risk factors. There is a lack of studies on the cardiovascular risk burden of basal ganglia perivascular spaces (BG-PVS) and their relationship with gray matter volume (GMV) and GM cerebral blood flow (CBF) in the aging brain. Here, we investigated these two issues in a large sample of cognitively intact older adults. Methods: A total of 734 volunteers were recruited. MRI was performed with 3.0 T using a pseudo-continuous arterial spin labeling (pCASL) sequence and a sagittal isotropic T1-weighted sequence for CBF and GMV analysis. The images obtained from 406 participants were analyzed to investigate the relationship between the severity of BG-PVS and GMV/CBF. False discovery rate-corrected P-values (PFDR) of <0.05 were considered significant. The images obtained from 254 participants were used to study the relationship between the severity of BG-PVS and cardiovascular risk burden. BG-PVS were rated using a 5-grade score. The severity of BG-PVS was classified as mild (grade <3) and severe (grade ≥3). Cardiovascular risk burden was assessed with the Framingham General Cardiovascular Risk Score (FGCRS). Results: Severe basal ganglia perivascular spaces were associated with significantly smaller GMV and CBF in multiple cortical regions (PFDR <0.05), and were associated with significantly larger volume in the bilateral caudate nucleus, pallidum, and putamen (PFDR <0.05). The participants with severe BG-PVS were more likely to have a higher cardiovascular risk burden than the participants with mild BG-PVS (60.71% vs. 42.93%; P =0.02). Conclusion: In cognitively intact older adults, severe BG-PVS are associated with smaller cortical GMV and CBF, larger subcortical GMV, and higher cardiovascular risk burden.
Collapse
Affiliation(s)
- Sirui Liu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Hou
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui You
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiwei Zhang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicheng Zhu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Zhentao Zuo
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,Sino-Danish College, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Feng Feng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Wang Y, Lu P, Zhan Y, Wu X, Qiu Y, Wang Z, Xu Q, Zhou Y. The Contribution of White Matter Diffusion and Cortical Perfusion Pathology to Vascular Cognitive Impairment: A Multimode Imaging-Based Machine Learning Study. Front Aging Neurosci 2021; 13:687001. [PMID: 34426730 PMCID: PMC8379092 DOI: 10.3389/fnagi.2021.687001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/02/2021] [Indexed: 11/13/2022] Open
Abstract
Widespread impairments in white matter and cerebrovascular integrity have been consistently implicated in the pathophysiology of patients with small vessel disease (SVD). However, the neural circuit mechanisms that underlie the developing progress of clinical cognitive symptoms remain largely elusive. Here, we conducted cross-modal MRI scanning including diffusion tensor imaging and arterial spin labeling in a cohort of 113 patients with SVD, which included 74 patients with vascular mild cognitive impairment (vMCI) and 39 patients without vMCI symptoms, and hence developed multimode imaging-based machine learning models to identify markers that discriminated SVD subtypes. Diffusion and perfusion features, respectively, extracted from individual white matter and gray matter regions were used to train three sets of classifiers in a nested 10-fold fashion: diffusion-based, perfusion-based, and combined diffusion-perfusion-based classifiers. We found that the diffusion-perfusion combined classifier achieved the highest accuracy of 72.57% with leave-one-out cross-validation, with the diffusion features largely spanning the capsular lateral pathway of the cholinergic tracts, and the perfusion features mainly distributed in the frontal-subcortical-limbic areas. Furthermore, diffusion-based features within vMCI group were associated with performance on executive function tests. We demonstrated the superior accuracy of using diffusion-perfusion combined multimode imaging features for classifying vMCI subtype out of a cohort of patients with SVD. Disruption of white matter integrity might play a critical role in the progression of cognitive impairment in patients with SVD, while malregulation of coritcal perfusion needs further study.
Collapse
Affiliation(s)
- Yao Wang
- Department of Radiology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Peiwen Lu
- Department of Neurology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yafeng Zhan
- Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Xiaowei Wu
- Department of Radiology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yage Qiu
- Department of Radiology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zheng Wang
- Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Qun Xu
- Department of Neurology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Zhou
- Department of Radiology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
19
|
Qiao Y, He X, Zhang J, Liang Y, Shao W, Zhang Z, Zhang S, Peng D. The Associations Between White Matter Disruptions and Cognitive Decline at the Early Stage of Subcortical Vascular Cognitive Impairment: A Case-Control Study. Front Aging Neurosci 2021; 13:681208. [PMID: 34408641 PMCID: PMC8364958 DOI: 10.3389/fnagi.2021.681208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/04/2021] [Indexed: 11/13/2022] Open
Abstract
Objective Emerging evidence suggests that white matter (WM) disruption is associated with the incidence of subcortical vascular cognitive impairment (SVCI). However, our knowledge regarding this relationship in the early stage of SVCI is limited. We aimed to investigate the associations between WM disruptions and cognitive declines at the early stage of SVCI. Method We performed a case–control study, involving 22 cases and 19 controls. The cases were patients at the early stage of SVCI, which was defined as subcortical ischemic vascular disease with normal global cognitive measures (pre-SVCI). The controls were healthy people matched by age, sex, and education years. We assessed the differences in a battery of neuropsychological tests between the two groups, investigated the diffusion changes in 40 WM tracts among the participants via an atlas-based segmentation strategy, and compared the differences between the cases and controls by multiple linear regression analysis. We then evaluated the relationships between diffusion indices and cognitive assessment scores by Pearson’s correlation. Results The pre-SVCI group exhibited significant differences in the Montreal cognitive assessment (MoCA), Rey–Osterrieth Complex Figure (R-O)-copy, and Trail Making Test (TMT)-B test compared with the controls. Compared with the controls, some long associative and projective bundles, such as the right anterior corona radiata (ACR), the right inferior fronto-occipital fasciculus (IFOF), and the left external capsule (EC), were extensively damaged in cases after Bonferroni correction (p < 0.05/40). Damages to specific fibers, such as the right ACR, IFOF, and posterior thalamic radiation (PTR), exhibited significant correlations with declines in MoCA, R-O delay, and the Mini-Mental State Examination (MMSE), respectively, after Bonferroni correction (p < 0.05/14). Conclusion Long WM tracts, especially those in the right hemisphere, were extensively damaged in the pre-SVCI patients and correlated with declines in executive functions and spatial processing. Patients of pre-SVCI are likely at an ultra-early stage of SVCI, and there is a very high risk of this condition becoming SVCI.
Collapse
Affiliation(s)
- Yanan Qiao
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Xuwen He
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Junying Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Ying Liang
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Wen Shao
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Zhanjun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Sihang Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Dantao Peng
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
20
|
Liu X, Cheng R, Chen L, Gong J, Luo T, Lv F. Altered Neurovascular Coupling in Subcortical Ischemic Vascular Disease. Front Aging Neurosci 2021; 13:598365. [PMID: 34054499 PMCID: PMC8149589 DOI: 10.3389/fnagi.2021.598365] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/18/2021] [Indexed: 11/18/2022] Open
Abstract
Patients with subcortical ischemic vascular disease (SIVD) exhibit a high risk of cognitive impairment that might be caused by neurologic deficits and vascular injuries. However, the mechanism remains unknown. In current study, 24 normal controls (NC) and 54 SIVD patients, including 26 SIVD patients with no cognitive impairment (SIVD-NCI) and 28 SIVD patients with mild cognitive impairment (SIVD-MCI) underwent the resting-state functional MRI (rs-fMRI) and neuropsychological assessments. We combined regional homogeneity (ReHo) and cerebral blood flow (CBF) by using the global ReHo-CBF correlations coefficient and the ReHo/CBF ratio to detect the inner link between neuronal activity and vascular responses. Correlations between the ReHo/CBF ratio and neuropsychological assessments were explored in patients with SIVD. As a result, we identified significantly decreased global ReHo-CBF coupling in the SIVD-NCI group and SIVD- MCI group with respect to the NC. The SIVD-MCI group showed more serious decoupling of the global ReHo-CBF correlation. We also found a significantly abnormal ReHo/CBF ratio predominantly located in cognitive-related brain regions, including the left insula, right middle temporal gyrus, right precuneus, left precentral gyrus, and left inferior parietal lobule but not the supramarginal and angular gyri. The SIVD-MCI group showed more severe disorders of neurovascular coupling than the other two groups. Moreover, the ReHo/CBF ratio in the left precentral gyrus of the SIVD-NCI group exhibited a positive correlation with the MMSE scores. These findings suggested that patients with SIVD show abnormal neurovascular coupling at the early stage of the disease and during disease development. It might be associated with disease severity and cognitive impairment. Neurovascular decoupling in brain may be a possible neuropathological mechanism of SIVD.
Collapse
Affiliation(s)
- Xiaoshuang Liu
- Department of Radiology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Runtian Cheng
- Department of Radiology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Chen
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Sichuan, China
| | - Junwei Gong
- Department of Radiology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tianyou Luo
- Department of Radiology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fajin Lv
- Department of Radiology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Jann K, Shao X, Ma SJ, Cen SY, D'Orazio L, Barisano G, Yan L, Casey M, Lamas J, Staffaroni AM, Kramer JH, Ringman JM, Wang DJJ. Evaluation of Cerebral Blood Flow Measured by 3D PCASL as Biomarker of Vascular Cognitive Impairment and Dementia (VCID) in a Cohort of Elderly Latinx Subjects at Risk of Small Vessel Disease. Front Neurosci 2021; 15:627627. [PMID: 33584191 PMCID: PMC7873482 DOI: 10.3389/fnins.2021.627627] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/07/2021] [Indexed: 01/16/2023] Open
Abstract
Cerebral small vessel disease (cSVD) affects arterioles, capillaries, and venules and can lead to cognitive impairments and clinical symptomatology of vascular cognitive impairment and dementia (VCID). VCID symptoms are similar to Alzheimer’s disease (AD) but the neurophysiologic alterations are less well studied, resulting in no established biomarkers. The purpose of this study was to evaluate cerebral blood flow (CBF) measured by 3D pseudo-continuous arterial spin labeling (pCASL) as a potential biomarker of VCID in a cohort of elderly Latinx subjects at risk of cSVD. Forty-five elderly Latinx subjects (12 males, 69 ± 7 years) underwent repeated MRI scans ∼6 weeks apart. CBF was measured using 3D pCASL in the whole brain, white matter and 4 main vascular territories (leptomeningeal anterior, middle, and posterior cerebral artery (leptoACA, leptoMCA, leptoPCA), as well as MCA perforator). The test-retest repeatability of CBF was assessed by intra-class correlation coefficient (ICC) and within-subject coefficient of variation (wsCV). Absolute and relative CBF was correlated with gross cognitive measures and domain specific assessment of executive and memory function, vascular risks, and Fazekas scores and volumes of white matter hyperintensity (WMH). Neurocognitive evaluations were performed using Montreal Cognitive Assessment (MoCA) and neuropsychological test battery in the Uniform Data Set v3 (UDS3). Good to excellent test-retest repeatability was achieved (ICC = 0.77–0.85, wsCV 3–9%) for CBF measurements in the whole brain, white matter, and 4 vascular territories. Relative CBF normalized by global mean CBF in the leptoMCA territory was positively correlated with the executive function composite score, while relative CBF in the leptoMCA and MCA perforator territory was positively correlated with MoCA scores, controlling for age, gender, years of education, and testing language. Relative CBF in WM was negatively correlated with WMH volume and MoCA scores, while relative leptoMCA CBF was positively correlated with WMH volume. Reliable 3D pCASL CBF measurements were achieved in the cohort of elderly Latinx subjects. Relative CBF in the leptomeningeal and perforator MCA territories were the most likely candidate biomarker of VCID. These findings need to be replicated in larger cohorts with greater variability of stages of cSVD.
Collapse
Affiliation(s)
- Kay Jann
- Laboratory of FMRI Technology, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Xingfeng Shao
- Laboratory of FMRI Technology, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Samantha J Ma
- Laboratory of FMRI Technology, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Steven Y Cen
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Lina D'Orazio
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Giuseppe Barisano
- Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Lirong Yan
- Laboratory of FMRI Technology, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Marlena Casey
- Laboratory of FMRI Technology, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jesse Lamas
- Laboratory of FMRI Technology, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Adam M Staffaroni
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Joel H Kramer
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - John M Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Danny J J Wang
- Laboratory of FMRI Technology, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
22
|
DeCarli C, Villeneuve S, Maillard P, Harvey D, Singh B, Carmichael O, Fletcher E, Olichney J, Farias S, Jagust W, Reed B, Mungas D. Vascular Burden Score Impacts Cognition Independent of Amyloid PET and MRI Measures of Alzheimer's Disease and Vascular Brain Injury. J Alzheimers Dis 2020; 68:187-196. [PMID: 30775991 DOI: 10.3233/jad-180965] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND/OBJECTIVE To determine the impact of vascular burden on rates of decline in episodic memory and executive function. We hypothesize that greater vascular burden will have an additive negative impact on cognition after accounting for baseline cognitive impairment, positron emission tomography (PET) amyloid burden, and magnetic resonance imaging (MRI) measures. METHODS Individuals were followed an average of 5 years with serial cognitive assessments. Predictor variables include vascular burden score (VBS), quantitative brain MRI assessment, and amyloid imaging. Subjects consisted of 65 individuals, 53% of whom were male, aged 73.2±7.2 years on average with an average of 15.5±3.3 years of educational achievement. RESULTS Baseline cognitive impairment was significantly associated poorer episodic memory (p < 0.0001), smaller hippocampal volume (p < 0.0001), smaller brain volume (p = 0.0026), and greater global Pittsburg Imaging Compound B (PiB) index (p = 0.0008). Greater amyloid burden was associated with greater decline in episodic memory over time (β= -0.20±0.07, p < 0.005). VBS was significantly associated with the level of executive function performance (β= -0.14±0.05, p < 0.005) and there was a significant negative interaction between VBS, cognitive impairment, and PiB index (β= -0.065±0.03, p = 0.03). CONCLUSIONS Our results find a significant influence of VBS independent of standard MRI measures and cerebral amyloid burden on executive function. In addition, VBS reduced the amount of cerebral amyloid burden needed to result in cognitive impairment. We conclude that the systemic effects of vascular disease as reflected by the VBS independently influence cognitive ability.
Collapse
Affiliation(s)
- Charles DeCarli
- UC Davis Department of Neurology and Center for Neuroscience, Davis, CA, USA
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, McGill University, Montreal, Canada
| | - Pauline Maillard
- UC Davis Department of Neurology and Center for Neuroscience, Davis, CA, USA
| | - Danielle Harvey
- Division of Biostatistics, School of Medicine, University of California at Davis, Davis, CA, USA
| | - Baljeet Singh
- UC Davis Department of Neurology and Center for Neuroscience, Davis, CA, USA
| | - Owen Carmichael
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Evan Fletcher
- UC Davis Department of Neurology and Center for Neuroscience, Davis, CA, USA
| | - John Olichney
- UC Davis Department of Neurology and Center for Neuroscience, Davis, CA, USA
| | - Sarah Farias
- UC Davis Department of Neurology and Center for Neuroscience, Davis, CA, USA
| | - William Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley CA, USA
| | - Bruce Reed
- Center for Scientific Review, Division of Neuroscience, Development and Aging, NIH, Bethesda, MD, USA
| | - Dan Mungas
- UC Davis Department of Neurology and Center for Neuroscience, Davis, CA, USA
| |
Collapse
|
23
|
Abstract
: The persistence of HIV in the central nervous system is somewhat controversial particularly in the context of HIV viral suppression from combined antiretroviral therapy. Further, its significance in relation to HIV pathogenesis in the context of HIV-associated neurocognitive disorders, systemic HIV pathogenesis, and eradication in general, but especially from the brain, are even more contentious. This review will discuss each of these aspects in detail, highlighting new data, particularly from recent conference presentations.
Collapse
|
24
|
Li W, Wei D, Liang J, Xie X, Song K, Huang L. Comprehensive Evaluation of White Matter Damage and Neuron Death and Whole-Transcriptome Analysis of Rats With Chronic Cerebral Hypoperfusion. Front Cell Neurosci 2019; 13:310. [PMID: 31379504 PMCID: PMC6653095 DOI: 10.3389/fncel.2019.00310] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 06/25/2019] [Indexed: 12/27/2022] Open
Abstract
Background/Aims Chronic cerebral hypoperfusion (CCH) is induced by chronic deficit of brain perfusion, contributes to a persistent or progressive cognitive dysfunction, which is characterized by diverse neuropathological manifestations. There are currently no effective medications available. White matter damage (WMD) and cortical neuron death may be caused by CCH, which are related to cognitive impairment, while the underlying molecular mechanisms remain unclear. In the study, a database of the transcriptome level was built to determine potential biomarkers in cortex of CCH. Methods CCH was induced in male Sprague-Dawley rats by permanent occlusion of the bilateral common carotid arteries. Rats were randomly divided into three groups: Sham-operated group (n = 24), the 4th and 8th week of CCH groups (total = 56, n = 28 for each group). Cognitive function was evaluated using the Morris water maze task. WMD and neuron damage were detected using diffusion tensor imaging and histological analysis, respectively. Western blotting analysis of various markers was used to examine neuronal death. Whole-transcriptome microarray was performed to assess mRNA, circRNA, and lncRNA expression profiles at 4th and 8th weeks after CCH. Diversified bioinformatic tools were performed to analyze and predict the key biological processes and signaling pathways of differentially expressed RNAs and co-expressed potential target genes. Co-expression networks of mRNA–circRNA–miRNA and lncRNA–mRNA were constructed. Results Compared to the sham group, cognitive impairment, disintegration of white matter, blood-brain barrier damage and neuron death were induced by CCH. Neuron death including apoptosis and necroptosis might occur in the cortex of CCH. We constructed the regulatory networks of whole-transcriptomic including differentially expressed mRNAs, circRNAs, and lncRNAs, and related biological functions and pathways involved in neurological disease, cell death and survival, energy and metabolism, et al. Our results also indicated that Cyr61 mRNA may play a role in the CCH-related cortical neuronal death. Conclusion WMD and cortical neuronal death are worthy of attention in the pathogenesis of CCH. Additionally, the present results provide potential evidence at the whole-transcription level for CCH, offering candidate biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Wenxian Li
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Department of Neurology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Di Wei
- Department of Urology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jianye Liang
- Medical Imaging Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xiaomei Xie
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Kangping Song
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Li'an Huang
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
25
|
Du J, Xu Q. Neuroimaging studies on cognitive impairment due to cerebral small vessel disease. Stroke Vasc Neurol 2019; 4:99-101. [PMID: 31338220 PMCID: PMC6613873 DOI: 10.1136/svn-2018-000209] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/23/2019] [Accepted: 03/18/2019] [Indexed: 01/17/2023] Open
Abstract
Vascular cognitive impairment (VCI) is a major contributor to age-related dementing illnesses which imposes a tremendous burden on families and society. It is a heterogeneous group of brain disorders. However, cerebral small vessel disease (CSVD) accounts for about 50%-70% of VCI, which represented a more homogeneous subtype of VCI. Advanced multimodal neuroimaging techniques like brain network connectome analyses are currently applied to explore the underlying mechanism of VCI. Some progress in the field of structural and functional brain network researches on a poststroke longitudinal CSVD cohort (Renji CSVD Cohort Study) was reported. Global and regional brain network characters were compared between patients with CSVD and healthy control. It suggested that distributed brain structural network disruption may play a pivot role in the cognitive decline. The results showed that brain structural network characters have distinctive differentiating capacity on the cognition of patients with CSVD.
Collapse
Affiliation(s)
- Jing Du
- Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Qun Xu
- Health Manage Center, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| |
Collapse
|
26
|
Liu X, Cheng R, Chen L, Luo T, Lv F, Gong J, Jiang P. Alterations of White Matter Integrity in Subcortical Ischemic Vascular Disease with and Without Cognitive Impairment: a TBSS Study. J Mol Neurosci 2019; 67:595-603. [PMID: 30685818 DOI: 10.1007/s12031-019-01266-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 01/17/2019] [Indexed: 10/27/2022]
Abstract
Patients with subcortical ischemic vascular disease (SIVD) may exhibit a high risk of cognitive impairment (CI) by disruption of white matter (WM) integrity. Diffusion tensor imaging (DTI) is recommended as a sensitive method to explore whole brain WM alterations at an asymptomatic stage of the disease, which might be correlated with underlying cognitive disorders. We aim to investigate alterations in WM microstructures and evaluate the relationships between the mean values of diffusion metrics (FA, MD, AD, and RD) and cognitive assessments in SIVD patients. Fifty SIVD patients with (SVCI, N = 25) and without (pre-SVCI, N = 25) cognitive impairments and normal controls (NC, N = 23) underwent DTI and neuropsychological examinations. DTI data were analyzed via TBSS to detect significant changes in WM tracts. Spearman correlation analysis was performed to evaluate relationships between the mean values of diffusion indices and the cognitive assessments. In general, extensive symmetrically altered areas that involved approximately the entire cerebral WM were noted in the pre-SVCI group but were less distinct than that noted in the SVCI group compared with NCs. The genu of corpus callosum exhibited the most damaged WM fiber. Throughout WM, FA was decreased, whereas MD, AD, and RD were increased. Some specific WM tracts in patient groups were significantly correlated with the severity of white matter hyperintensity (WMH), cognitive assessments about executive functions and processing speed. WM integrity has already been damaged at the pre-SVCI stage, which would be associate with future cognitive dysfunction. DTI could potentially establish early biomarkers to detect underlying mechanisms of SIVD.
Collapse
Affiliation(s)
- Xiaoshuang Liu
- The Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Runtian Cheng
- The Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Chen
- The Department of Radiology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Tianyou Luo
- The Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - FaJin Lv
- The Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junwei Gong
- The Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peiling Jiang
- The Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
27
|
Saad M, Bilal M, Gabr W, Elnaby AA. The Diagnostic Role of Brain MRI in Detection of Multiple Sclerosis Related Cognitive Impairment. JOURNAL OF BEHAVIORAL AND BRAIN SCIENCE 2019; 09:313-324. [DOI: 10.4236/jbbs.2019.98023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
28
|
Cuberas-Borrós G, Roca I, Boada M, Tárraga L, Hernández I, Buendia M, Rubio L, Torres G, Bittini Á, Guzmán-de-Villoria JA, Pujadas F, Torres M, Núñez L, Castell J, Páez A. Longitudinal Neuroimaging Analysis in Mild-Moderate Alzheimer's Disease Patients Treated with Plasma Exchange with 5% Human Albumin. J Alzheimers Dis 2018; 61:321-332. [PMID: 29154283 PMCID: PMC5734124 DOI: 10.3233/jad-170693] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background: Recently, modifications of Aβ1-42 levels in CSF and plasma associated with improvement in memory and language functions have been observed in patients with mild-moderate Alzheimer’s disease (AD) treated with plasma exchange (PE) with albumin replacement. Objective: To detect structural and functional brain changes in PE-treated AD patients as part of a Phase II clinical trial. Methods: Patients received between 3 and 18 PE with albumin (Albutein® 5%, Grifols) or sham-PE (controls) for 21 weeks (divided in one intensive and two maintenance periods) followed by 6-month follow-up. Brain perfusion assessed by SPECT scans using an automated software (NeuroGam®) and brain structural changes assessed by MRI were performed at weeks 0 (baseline), 21, and 44 (with additional SPECT at weeks 9 and 33). Statistical parametric mapping (voxel-based analysis, SPM) and Z-scores calculations were applied to investigate changes to baseline. Results: 42 patients were recruited (39 evaluable; 37 analyzed: 18 PE-treated; 19 controls). There was a trend toward decreasing hippocampi and total intracranial volume for both patient groups during the study (p < 0.05). After six months, PE-treated patients had less cerebral perfusion loss than controls in frontal, temporal, and parietal areas, and perfusion stabilization in Brodmann area BA38-R during the PE-treatment period (p < 0.05). SPM analysis showed stabilization or absence of progression of perfusion loss in PE-treated patients until week 21, not observed in controls. Conclusions: Mild-moderate AD patients showed decreased brain volume and impairment of brain perfusion as expected for the progression of the disease. PE-treatment with albumin replacement favored the stabilization of perfusion.
Collapse
Affiliation(s)
- Gemma Cuberas-Borrós
- Department of Nuclear Medicine, Institut de Diagnòstic per la Imatge (IDI), Hospital General Universitari Vall d'Hebrón, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Isabel Roca
- Department of Nuclear Medicine, Institut de Diagnòstic per la Imatge (IDI), Hospital General Universitari Vall d'Hebrón, Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Nuclear Medicine, Gammagrafía Corachan, Barcelona, Spain
| | - Mercè Boada
- Department of Neurology, Hospital General Universitari Vall d'Hebrón, Barcelona, Spain.,Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Lluís Tárraga
- Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Isabel Hernández
- Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Mar Buendia
- Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Lourdes Rubio
- Department of Nuclear Medicine, Gammagrafía Corachan, Barcelona, Spain
| | - Gustavo Torres
- Department of Nuclear Medicine, Gammagrafía Corachan, Barcelona, Spain
| | - Ángel Bittini
- Department of Nuclear Medicine, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | - Francesc Pujadas
- Department of Neurology, Hospital General Universitari Vall d'Hebrón, Barcelona, Spain
| | - Mireia Torres
- Department of Clinical, Instituto Grifols, S.A., Barcelona, Spain
| | - Laura Núñez
- Department of Clinical, Instituto Grifols, S.A., Barcelona, Spain
| | - Joan Castell
- Department of Nuclear Medicine, Institut de Diagnòstic per la Imatge (IDI), Hospital General Universitari Vall d'Hebrón, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antonio Páez
- Department of Clinical, Instituto Grifols, S.A., Barcelona, Spain
| |
Collapse
|
29
|
Ye Q, Bai F. Contribution of diffusion, perfusion and functional MRI to the disconnection hypothesis in subcortical vascular cognitive impairment. Stroke Vasc Neurol 2018; 3:131-139. [PMID: 30294468 PMCID: PMC6169607 DOI: 10.1136/svn-2017-000080] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 01/26/2018] [Accepted: 02/14/2018] [Indexed: 11/29/2022] Open
Abstract
Vascular cognitive impairment (VCI) describes all forms of cognitive impairment caused by any type of cerebrovascular disease. Early identification of VCI is quite difficult due to the lack of both sensitive and specific biomarkers. Extensive damage to the white matter tracts, which connect the cortical and subcortical regions, has been shown in subcortical VCI (SVCI), the most common subtype of VCI that is caused by small vessel disease. Two specific MRI sequences, including diffusion tensor imaging (DTI) and functional MRI (fMRI), have emerged as useful tools for identifying subtle white matter changes and the intrinsic connectivity between distinct cortical regions. This review describes the advantages of these two modalities in SVCI research and the current DTI and fMRI findings on SVCI. Using DTI technique, a variety of studies found that white matter microstructural damages in the anterior and superior areas are more specific to SVCI. Similarly, functional brain abnormalities detected by fMRI have also been mainly shown in anterior brain areas in SVCI. The characteristic distribution of brain abnormalities in SVCI interrupts the prefrontal-subcortical loop that results in cognitive impairments in particular domains, which further confirms the ‘disconnection syndrome’ hypothesis. In addition, another MRI technique, arterial spin labelling (ASL), has been used to describe the disconnection patterns in a variety of conditions by measuring cerebral blood flow. The role of the ASL technique in SVCI research is also assessed. Finally, the review proposes the application of multimodality fusion in the investigation of SVCI pathogenesis.
Collapse
Affiliation(s)
- Qing Ye
- Department of Neurology, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Feng Bai
- Department of Neurology, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, China
| |
Collapse
|
30
|
Wallin A, Román GC, Esiri M, Kettunen P, Svensson J, Paraskevas GP, Kapaki E. Update on Vascular Cognitive Impairment Associated with Subcortical Small-Vessel Disease. J Alzheimers Dis 2018; 62:1417-1441. [PMID: 29562536 PMCID: PMC5870030 DOI: 10.3233/jad-170803] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
Subcortical small-vessel disease (SSVD) is a disorder well characterized from the clinical, imaging, and neuropathological viewpoints. SSVD is considered the most prevalent ischemic brain disorder, increasing in frequency with age. Vascular risk factors include hypertension, diabetes, hyperlipidemia, elevated homocysteine, and obstructive sleep apnea. Ischemic white matter lesions are the hallmark of SSVD; other pathological lesions include arteriolosclerosis, dilatation of perivascular spaces, venous collagenosis, cerebral amyloid angiopathy, microbleeds, microinfarcts, lacunes, and large infarcts. The pathogenesis of SSVD is incompletely understood but includes endothelial changes and blood-brain barrier alterations involving metalloproteinases, vascular endothelial growth factors, angiotensin II, mindin/spondin, and the mammalian target of rapamycin pathway. Metabolic and genetic conditions may also play a role but hitherto there are few conclusive studies. Clinical diagnosis of SSVD includes early executive dysfunction manifested by impaired capacity to use complex information, to formulate strategies, and to exercise self-control. In comparison with Alzheimer's disease (AD), patients with SSVD show less pronounced episodic memory deficits. Brain imaging has advanced substantially the diagnostic tools for SSVD. With the exception of cortical microinfarcts, all other lesions are well visualized with MRI. Diagnostic biomarkers that separate AD from SSVD include reduction of cerebrospinal fluid amyloid-β (Aβ)42 and of the ratio Aβ42/Aβ40 often with increased total tau levels. However, better markers of small-vessel function of intracerebral blood vessels are needed. The treatment of SSVD remains unsatisfactory other than control of vascular risk factors. There is an urgent need of finding targets to slow down and potentially halt the progression of this prevalent, but often unrecognized, disorder.
Collapse
Affiliation(s)
- Anders Wallin
- Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg Sweden and Memory Clinic at Department of Neuropsychiatry, Sahlgrenska University, Hospital, Gothenburg, Sweden
| | - Gustavo C. Román
- Department of Neurology, Methodist Neurological Institute, Houston, TX, USA
- Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Margaret Esiri
- Neuropathology Department, West Wing, John Radcliffe Hospital, Oxford, UK
| | - Petronella Kettunen
- Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg Sweden and Memory Clinic at Department of Neuropsychiatry, Sahlgrenska University, Hospital, Gothenburg, Sweden
- Nuffield Department of Clinical Neurosciences, University of Oxford, West Wing, John Radcliffe Hospital, Oxford, UK
| | - Johan Svensson
- Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - George P. Paraskevas
- 1st Department of Neurology, Neurochemistry Unit, National and Kapodistrian University of Athens, Athens, Greece
| | - Elisabeth Kapaki
- 1st Department of Neurology, Neurochemistry Unit, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
31
|
Abstract
Purpose of Review In this review, we summarise the evidence on the association between cardiovascular disease (CVD) and cognitive impairment and explore the role of the nitric oxide (NO) pathway as a causal mechanism. Recent Findings Evidence from epidemiological studies suggests that the presence of CVD and its risk factors in midlife is associated with an increased risk of later life cognitive impairment and dementia. It is unclear what is driving this association but risk may be conveyed via an increase in neurodegeneration (e.g. amyloid deposition), vascular changes (e.g. small vessel disease) and mechanistically due to increased levels of oxidative stress and inflammation as well as changes in NO bioavailability. Summary CVDs and dementia are major challenges to global health worldwide. The NO pathway may be a promising biological candidate for future studies focused on reducing not only CVD but also risk of cognitive decline and dementia.
Collapse
|
32
|
Wang X, Song R, Lu W, Liu Z, Wang L, Zhu X, Liu Y, Sun Z, Li J, Li X. YXQN Reduces Alzheimer's Disease-Like Pathology and Cognitive Decline in APPswePS1dE9 Transgenic Mice. Front Aging Neurosci 2017; 9:157. [PMID: 28603494 PMCID: PMC5440527 DOI: 10.3389/fnagi.2017.00157] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 05/05/2017] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease (AD) is the world's most common form of dementia, in which aggregation of amyloid-β (Aβ) is the hallmark. Unfortunately, few medicines have succeeded to completely cure AD. Yangxue Qingnao (YXQN) is a Chinese traditional medicine, and its pharmacological effect is improving cerebral blood flow. In this study, we firstly demonstrated that YXQN reduced AD-like pathology and cognitive impairment in APPswePS1dE9 (APP/PS1) mice with 2 months administration. Our data showed that YXQN substantially ameliorated behavioral defects in 10-month old APP/PS1 mice using Morris Water Maze and Y-maze tests, in which the cognitive ability of YXQN high-dose group approaches to wild type mice. Next, we focused on the brain pathological alterations in the YXQN group by three experiments, including thioflavin-S, congo-red, and Aβ-immunohistochemistry staining. The results demonstrated that the high-dose of YXQN dramatically suppressed amyloid plaques in the hippocampus and cortex of APP/PS1 mice, which showed a 47-72% reduction in plaque deposits, relative to the vehicle group. In addition, our data verified that YXQN decreased the cerebral amyloid load by attenuating β-secretase BACE1 and γ-secretase PS1 in the pathological processing of APP, and promoting the level of α-secretase ADAM10 in the physiological processing of APP to generate more sAPPα, which combats amyloidosis formation, and also carries out neurotropic and neuroprotective effect. Taken together, our results strongly suggest that YXQN could be a potential medicine for AD, and provide new evidence for further AD drug research and development.
Collapse
Affiliation(s)
- Xiaowan Wang
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal UniversityChangchun, China
| | - Runmin Song
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal UniversityChangchun, China
| | - Wenliang Lu
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal UniversityChangchun, China
- School of Traditional Chinese Pharmacology, Tianjin University of Traditional Chinese Medicine, TianjinChina
| | - Ziyu Liu
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal UniversityChangchun, China
| | - Lichun Wang
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal UniversityChangchun, China
| | - Xiaojuan Zhu
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal UniversityChangchun, China
| | - Yanjun Liu
- Division of Endocrinology, Metabolism and Molecular Medicine, UCLA School of Medicine, Charles R. Drew University of Medicine and Science, Los AngelesCA, United States
| | - Zijie Sun
- Department of Genetics, Stanford University School of Medicine, StanfordCA, United States
| | - Jiang Li
- Dental Hospital, Jilin University, ChangchunChina
| | - Xiaomeng Li
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal UniversityChangchun, China
| |
Collapse
|
33
|
Sun Y, Ge X, Han X, Cao W, Wang Y, Ding W, Cao M, Zhang Y, Xu Q, Zhou Y, Xu J. Characterizing Brain Iron Deposition in Patients with Subcortical Vascular Mild Cognitive Impairment Using Quantitative Susceptibility Mapping: A Potential Biomarker. Front Aging Neurosci 2017; 9:81. [PMID: 28424610 PMCID: PMC5371674 DOI: 10.3389/fnagi.2017.00081] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 03/14/2017] [Indexed: 11/13/2022] Open
Abstract
The presence and pattern of iron accumulation in subcortical vascular mild cognitive impairment (svMCI) and their effects on cognition have rarely been investigated. We aimed to examine brain iron deposition in svMCI subjects using quantitative susceptibility mapping (QSM). Moreover, we aimed to investigate the correlation between brain iron deposition and the severity of cognitive impairment as indicated by z-scores. We recruited 20 subcortical ischemic vascular disease (SIVD) patients who fulfilled the criteria for svMCI. The control group comprised 19 SIVD patients without cognitive impairment. The SIVD and control groups were matched based on age, gender, and years of education. Both groups underwent QSM using a 3.0T MRI system. Susceptibility maps were reconstructed from in vivo data, which were acquired with a three-dimensional spoiled gradient recalled sequence. Then, regions of interest were drawn manually on the map of each subject. The inter-group differences of susceptibility values were explored in deep gray matter nuclei, including the bilateral pulvinar nucleus of the thalamus, head of caudate nucleus, globus pallidus, putamen, hippocampus, substantia nigra, and red nucleus. The correlations between regional iron deposition and composite z-score, memory z-score, language z-score, attention-executive z-score and visuospatial z-score were assessed using partial correlation analysis, with patient age and gender as covariates. Compared with the control, the svMCI group had elevated susceptibility values within the bilateral hippocampus and right putamen. Furthermore, the susceptibility value in the right hippocampus was negatively correlated with memory z-score and positively correlated with language z-score. The susceptibility value in the right putamen was negatively correlated with attention-executive z-score in the svMCI group. However, composite z-score were unrelated to susceptibility values. Our results suggest that brain iron deposition has clinical relevance as a biomarker for cognition. In addition, our results highlight the importance of iron deposition in understanding svMCI-associated cognitive deficits in addition to conventional MRI markers.
Collapse
Affiliation(s)
- Yawen Sun
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Xin Ge
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Xu Han
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Wenwei Cao
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Yao Wang
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Weina Ding
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Mengqiu Cao
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Yong Zhang
- GE Applied Science Laboratory, GE HealthcareShanghai, China
| | - Qun Xu
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Yan Zhou
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Jianrong Xu
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| |
Collapse
|