1
|
He Z, Sun J. The role of the neurovascular unit in vascular cognitive impairment: Current evidence and future perspectives. Neurobiol Dis 2024; 204:106772. [PMID: 39710068 DOI: 10.1016/j.nbd.2024.106772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024] Open
Abstract
Vascular cognitive impairment (VCI) is a progressive cognitive impairment caused by cerebrovascular disease or vascular risk factors. It is the second most common type of cognitive impairment after Alzheimer's disease. The pathogenesis of VCI is complex, and neurovascular unit destruction is one of its important mechanisms. The neurovascular unit (NVU) is responsible for combining blood flow with brain activity and includes endothelial cells, pericytes, astrocytes and many regulatory nerve terminals. The concept of an NVU emphasizes that interactions between different types of cells are essential for maintaining brain homeostasis. A stable NVU is the basis of normal brain function. Therefore, understanding the structure and function of the neurovascular unit and its role in VCI development is crucial for gaining insights into its pathogenesis. This article reviews the structure and function of the neurovascular unit and its contribution to VCI, providing valuable information for early diagnosis and prevention.
Collapse
Affiliation(s)
- Zhidong He
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun 130031, Jilin, China
| | - Jing Sun
- Department of Neurology, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun 130031, Jilin, China..
| |
Collapse
|
2
|
Wang N, Li J, Zhang X, Gao Y, Sui C, Zhang N, Che Y, Liang C, Guo L, Li M. Hippocampal fimbria atrophy and its mediating effect between cerebral small vessel disease and cognitive impairment. Neuroscience 2024; 562:54-62. [PMID: 39461662 DOI: 10.1016/j.neuroscience.2024.10.039] [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: 07/05/2024] [Revised: 10/08/2024] [Accepted: 10/19/2024] [Indexed: 10/29/2024]
Abstract
We aimed to investigate the relationship between the volume reduction in hippocampal (HP) subregions and cognitive impairment in patients with cerebral small vessel disease (CSVD). Clinical, cognitive, and magnetic resonance imaging data were obtained for 315 participants. The CSVD group included 146 participants with a total CSVD score of 1-4. 169 participants with a total CSVD score of zero were used as control group (CSVD-0). The volume differences of 19 HP subregions between CSVD and CSVD-0 groups were analyzed, and we investigated the hazard factors that might cause subregional volume reduction in HP. Mediation analysis was performed to detect the relationship among HP subregional volumes, CSVD burden, and cognitive function. In our results, significant differences can be found in the volumes of CA4 body, presubiculum-head, presubiculum-body, subiculum-body, GC-ML-DG-head, GC-ML-DG-body, fimbria, and HP tail between CSVD group and control group. Regression analysis showed that fimbria was the most impacted HP subregion by CSVD. And mediation analysis revealed fimbria volume was a mediator variable between total CSVD score and MoCA/SCWT score. These results suggest that the volumes of HP subregions, especially the fimbria, may be effective potential biomarkers for early detecting cognitive impairment in CSVD.
Collapse
Affiliation(s)
- Na Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jing-wu Road No. 324, Jinan 250021, Shandong, China.
| | - Jing Li
- Department of Radiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China.
| | - Xinyue Zhang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jing-wu Road No. 324, Jinan 250021, Shandong, China.
| | - Yian Gao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jing-wu Road No. 324, Jinan 250021, Shandong, China.
| | - Chaofan Sui
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jing-wu Road No. 324, Jinan 250021, Shandong, China.
| | - Nan Zhang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jing-wu Road No. 324, Jinan 250021, Shandong, China.
| | - Yena Che
- Department of Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jing-wu Road, Jinan, Shandong 250021, China.
| | - Changhu Liang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jing-wu Road No. 324, Jinan 250021, Shandong, China.
| | - Lingfei Guo
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jing-wu Road No. 324, Jinan 250021, Shandong, China.
| | - Meng Li
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany.
| |
Collapse
|
3
|
Zhang W, Yu X, Wei M, Zhou J, Zhou Y, Zhou X, Zhao K, Zhu X. The influence of alterations in the composition of intestinal microbiota on neurovascular coupling and cognitive dysfunction in individuals afflicted with CSVD. Brain Res Bull 2024; 219:111115. [PMID: 39510273 DOI: 10.1016/j.brainresbull.2024.111115] [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: 07/16/2024] [Revised: 10/17/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
INTRODUCTION An expanding body of research has explored the crucial role of gut microbiota in cerebral small vessel disease (CSVD). The objective of this study is to investigate alterations in the gut microbiota structure among CSVD patients, to explore the correlation between differential taxonomic levels and the neurovascular coupling index as well as cognitive function and to elucidate the imaging and biomarkers of mild cognitive impairment (MCI) in CSVD. METHODS We enrolled 104 patients with CSVD and 40 healthy controls (HC). Based on cognitive test scores, CSVD patients were categorized into a cognitively normal group (CSVD-NCI, n=61) and a mild cognitive impairment group (CSVD-MCI, n=43). Performing magnetic resonance imaging (MRI) scans, gut microbiota analysis, as well as clinical and neuropsychological assessments for all participants. Based on arterial spin labeling (ASL) and blood oxygen level-dependent (BOLD) imaging data, cerebral blood flow (CBF) and neural activity indices are computed. The coupling indices of CBF/mReHo, CBF/mfALFF, CBF/mALFF, and CBF/mDC are calculated to assess the whole-brain neurovascular coupling changes in patients with CSVD. RESULTS Species annotation revealed differences in the composition at the phylum and genus levels among the HC, CSVD-NCI, and CSVD-MCI groups. Additionally, differential analysis using the Kruskal-Wallis test demonstrated specific dominant microbial communities in all three groups. The relative abundance of certain dominant microbial communities in CSVD patients exhibited correlations with neurovascular coupling and cognitive function. The combined assessment of Bacteroides genus and CBF/mDC proved effective in distinguishing between CSVD-NCI and CSVD-MCI, providing a novel non-invasive approach for the diagnosis of MCI in CSVD.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Xianfeng Yu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China; Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Min Wei
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Jie Zhou
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Yajun Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China
| | - Xia Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China.
| | - Kai Zhao
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China.
| | - Xiaoqun Zhu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, China.
| |
Collapse
|
4
|
Lu J, Zuo X, Cai A, Xiao F, Xu Z, Wang R, Miao C, Yang C, Zheng X, Wang J, Ding X, Xiong W. Cerebral small vessel injury in mice with damage to ACE2-expressing cerebral vascular endothelial cells and post COVID-19 patients. Alzheimers Dement 2024; 20:7971-7988. [PMID: 39352003 PMCID: PMC11567838 DOI: 10.1002/alz.14279] [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: 02/26/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 10/03/2024]
Abstract
INTRODUCTION The angiotensin-converting enzyme 2 (ACE2), which is expressed in cerebral vascular endothelial cells (CVECs), has been currently identified as a functional receptor for SARS-CoV-2. METHODS We specifically induced injury to ACE2-expressing CVECs in mice and evaluated the effects of such targeted damage through magnetic resonance imaging (MRI) and cognitive behavioral tests. In parallel, we recruited a single-center cohort of COVID-19 survivors and further assessed their brain microvascular injury based on cognition and emotional scales, cranial MRI scans, and blood proteomic measurements. RESULTS Here, we show an array of pathological and behavioral alterations characteristic of cerebral small vessel disease (CSVD) in mice that targeted damage to ACE2-expressing CVECs, and COVID-19 survivors. These CSVD-like manifestations persist for at least 7 months post-recovery from COVID-19. DISCUSSION Our findings suggest that SARS-CoV-2 may induce cerebral small vessel damage with persistent sequelae, underscoring the imperative for heightened clinical vigilance in mitigating or treating SARS-CoV-2-mediated cerebral endothelial injury throughout infection and convalescence. HIGHLIGHTS Cerebral small vessel disease-associated changes were observed after targeted damage to angiotensin-converting enzyme 2-expressing cerebral vascular endothelial cells. SARS-CoV-2 may induce cerebral small vessel damage with persistent sequelae. Clinical vigilance is needed in preventing SARS-CoV-2-induced cerebral endothelial damage during infection and recovery.
Collapse
Affiliation(s)
- Jieping Lu
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Xin Zuo
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial IntelligenceHefei Comprehensive National Science CenterHefeiChina
| | - Aoling Cai
- Key Laboratory of Magnetic Resonance in Biological SystemsState Key Laboratory of Magnetic Resonance and Atomic and Molecular PhysicsNational Center for Magnetic Resonance in WuhanWuhan Institute of Physics and MathematicsInnovation Academy for Precision Measurement Science and TechnologyChinese Academy of Sciences‐Wuhan National Laboratory for OptoelectronicsWuhanChina
- The Affiliated Changzhou Second People's Hospital of Nanjing Medical UniversityChangzhou Second People's HospitalChangzhou Medical CenterNanjing Medical UniversityChangzhouChina
| | - Fang Xiao
- Department of RadiologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Zhenyu Xu
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Rui Wang
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Chenjian Miao
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Chen Yang
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Xingxing Zheng
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Jie Wang
- Key Laboratory of Magnetic Resonance in Biological SystemsState Key Laboratory of Magnetic Resonance and Atomic and Molecular PhysicsNational Center for Magnetic Resonance in WuhanWuhan Institute of Physics and MathematicsInnovation Academy for Precision Measurement Science and TechnologyChinese Academy of Sciences‐Wuhan National Laboratory for OptoelectronicsWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xiaoling Ding
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Wei Xiong
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial IntelligenceHefei Comprehensive National Science CenterHefeiChina
- Anhui Province Key Laboratory of Biomedical Aging ResearchHefeiChina
- CAS Key Laboratory of Brain Function and DiseaseHefeiChina
| |
Collapse
|
5
|
Lv Y. The effects of immunomodulatory drugs on cerebral small vessel disease: A mediation Mendelian randomization analysis. Int Immunopharmacol 2024; 140:112786. [PMID: 39121606 DOI: 10.1016/j.intimp.2024.112786] [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: 04/09/2024] [Revised: 06/05/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND There are only a few recognized drug targets for cerebral small vessel disease (CSVD). Though inflammation is increasingly implicated in the development of CSVD, it remains unclear whether immunomodulation could become a therapeutic target. Accordingly, the Mendelian randomization (MR) method was used to assess the genetically proxied impacts of IL6 receptor (IL6R) inhibitor, IL1β inhibitor, Tumor necrosis factor (TNF) inhibitor and β-tubulin inhibitor on CSVD through. METHODS Single nucleotide polymorphisms (SNPs) near the IL6R, IL1β, TNFRSF1A and β-tubulin genes were identified as genetic proxies for immunomodulatory drugs. These SNPs exhibited significant associations with serum C-reactive protein (CRP) levels in a large European genome-wide association study. The causal effects of immunomodulatory drugs on CSVD manifestations and the mediation influence of 731 peripheral blood immune phenotypes linking these drugs to CSVD manifestations were examined using a two-sample two-step MR approach. RESULTS A total of 9, 18, 4 and 1 SNP were identified to proxy the effects of IL1β inhibitor, IL6R inhibitor, TNF inhibitor and β-tubulin inhibitor, respectively. MR analysis showed a significant causal relationship between IL1β inhibition and reduced volume of periventricular white matter hyperintensity (PWMH). IL6R inhibition was associated with a reduced risk of small vessel stroke, decreased axial diffusivity and mean diffusivity. Genetically proxied TNF inhibition may decrease the occurrence of cerebral microbleeds (CMBs) and severe enlarged perivascular spaces located at white matter (WM-EPVS). It could also protect WM integrity, as evidenced by the reduced volumes of PWMH and deep white matter hyperintensity (DWMH). Various peripheral blood immune phenotypes exhibited significant associations with immunomodulatory drugs. Notably, the median fluorescence intensity (MFI) of CD45 on CD8br cells partially mediated the effects of IL1β inhibitor on PWMH volume. Indirect effects of TNF inhibition on PWMH and DWMH volume through the MFI of CD127 on CD28- CD8br cells were observed. The effects of TNF inhibition on the occurrence of any CMBs were partially mediated by the MFI of CD45 on natural killer T cells, and the effects of TNF inhibition on the occurrence of lobar CMBs were partially mediated by the MFI of HLA DR on CD33- HLA DR+ cells. Furthermore, the MFI of HLA DR on CD33- HLA DR+ cells partially mediated the effects of TNF inhibition on WM-EPVS. CONCLUSIONS IL1β inhibitor, IL6R inhibitor and TNF inhibitor were associated with lower burden of CSVD while the activation of certain immune cells such as Tregs and myeloid cells partially mediated their protective effects.
Collapse
Affiliation(s)
- Yanchen Lv
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
Wang J, Du K, Liu C, Chen X, Ban W, Zhu G, Yang J. Exploring the effects of moxibustion on cognitive function in rats with multiple cerebral infarctions from the perspective of glial vascular unit repairing. Front Pharmacol 2024; 15:1428907. [PMID: 39508044 PMCID: PMC11539022 DOI: 10.3389/fphar.2024.1428907] [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: 06/03/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024] Open
Abstract
Objective This study aimed to explore the effect of moxibustion at Governor Vessel (GV) acupoints, including Baihui (GV 20), Shenting (GV 24) and Dazhui (GV 14) for 14 days on glial vascular unit (GVU) in rats with multiple microinfarctions (MMI), and to explore its action mechanism. Methods The effect and mechanism of moxibustion on vascular dementia (VD) were studied in MMI rats by means of behavioral and molecular biology experiments. Results Rats receiving MMI showed impairment of memory function, reduction of cerebral blood flow, damage of blood-brain barrier (BBB) integrity and increased brain mass. MMI also increased neuronal degeneration in the hippocampus. Notably, levels of glial fibrillary acidic protein (GFAP) and complement component 3 significantly increased, but those of Connexin43 (CX43) and platelet derived growth factor receptor β (PDGFRβ) significantly decreased in the hippocampus of the rats receiving MMI. Moxibustion, as well as oxiracetam (ORC) treatment improved memory function and neuronal degeneration, ameliorated BBB integrity, increased cerebral blood flow and decreased brain mass. In addition, moxibustion as well as oxiracetam (ORC) treatment reduced the decrease of CX43 protein and increased PDGFRβ protein level in the hippocampus of MMI rats. Moreover, moxibustion treatment reversed MMI-induced increase of the GFAP/CX43 ratio in vascular structural units. Importantly, after PDGFRβ inhibition, VD rats treated with moxibustion had impaired learning and memory, decreased cerebral blood flow, and BBB disruption. Conclusion Moxibustion treatment at various GV acupoints improved cerebral blood flow and repaired BBB function in rats with MMI, likely through protecting GVU.
Collapse
Affiliation(s)
- Jingji Wang
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
- Acupuncture and Moxibustion Clinical Medical Research Center of Anhui Province, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Kunrui Du
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Chang Liu
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Xiaoyu Chen
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Wenming Ban
- Department of Neurology, Taihe County Hospital of Traditional Chinese Medicine, Fuyang, China
| | - Guoqi Zhu
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Jun Yang
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
- The First Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
7
|
Fan F, Song H, Jiang J, He H, Sun D, Xu Z, Peng S, Zhang R, Li T, Cao J, Xu J, Peng X, Lei M, He C, Zhang J. Development and validation of a multimodal deep learning framework for vascular cognitive impairment diagnosis. iScience 2024; 27:110945. [PMID: 39391736 PMCID: PMC11465129 DOI: 10.1016/j.isci.2024.110945] [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: 04/03/2024] [Revised: 06/15/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
Cerebrovascular disease (CVD) is the second leading cause of dementia worldwide. The accurate detection of vascular cognitive impairment (VCI) in CVD patients remains an unresolved challenge. We collected the clinical non-imaging data and neuroimaging data from 307 subjects with CVD. Using these data, we developed a multimodal deep learning framework that combined the vision transformer and extreme gradient boosting algorithms. The final hybrid model within the framework included only two neuroimaging features and six clinical features, demonstrating robust performance across both internal and external datasets. Furthermore, the diagnostic performance of our model on a specific dataset was demonstrated to be comparable to that of expert clinicians. Notably, our model can identify the brain regions and clinical features that significantly contribute to the VCI diagnosis, thereby enhancing transparency and interpretability. We developed an accurate and explainable clinical decision support tool to identify the presence of VCI in patients with CVD.
Collapse
Affiliation(s)
- Fan Fan
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, China
| | - Hao Song
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, China
| | - Jiu Jiang
- Electronic Information School, Wuhan University, Wuhan, Hubei province, China
| | - Haoying He
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, China
| | - Dong Sun
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, China
| | - Zhipeng Xu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, China
| | - Sisi Peng
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, China
| | - Ran Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, China
| | - Tian Li
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, China
| | - Jing Cao
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, China
| | - Juan Xu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, China
| | - Xiaoxiang Peng
- Department of Neurology, Third People’s Hospital of Hubei Province, Wuhan, Hubei province, China
| | - Ming Lei
- Department of Neurology, General Hospital of the Yangtze River Shipping, Wuhan, Hubei province, China
| | - Chu He
- Electronic Information School, Wuhan University, Wuhan, Hubei province, China
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, China
| |
Collapse
|
8
|
Xu C, Xia J, Qiang Y, Wu Y. Relationship between burden of cerebral small vessel disease on imaging and cognitive impairment of COPD patients. J Neurosci Methods 2024; 410:110218. [PMID: 38996845 DOI: 10.1016/j.jneumeth.2024.110218] [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: 04/23/2024] [Revised: 06/18/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
OBJECTIVE This study aims to explore the relationship between the burden of cerebral small vessel disease (CSVD) on imaging and cognitive impairment (CI) in patients with chronic obstructive pulmonary disease (COPD). METHODS The study included 118 COPD patients admitted to Changxing People's Hospital between July 2020 and July 2023. All patients received a 1.5 T MRI of the brain and pulmonary function tests. A cognitive function assessment was conducted via the Montreal Cognitive Assessment (MoCA) scale, and patients were divided into two groups. The relationship between the MoCA and CSVD burden score was analyzed by Pearson correlation, and to identify risk factors, multiple logistic regression analysis was performed. RESULTS The study showed a negative correlation between the MoCA and CSVD burden score in COPD patients (r=-0.479, P<0.001). Multiple logistic regression analysis found that age (OR=2.264, 95 % CI: 1.426-3.596, P<0.001), COPD grade (OR=3.139, 95 % CI: 2.012-4.898, P<0.001), as well as CSVD burden score (OR=5.336, 95 % CI: 1.191-23.900, P<0.001) were the independent risk factors for CI in COPD patients (P<0.05). CONCLUSION When screening for cognitive impairment in COPD patients, the CSVD burden score can be used in conjunction with cognitive assessment scales to make judgments.
Collapse
Affiliation(s)
- Chentao Xu
- Department of Radiology, Changxing People's Hospital, Huzhou, Zhejiang 313100, China.
| | - Jinjin Xia
- Department of Neurology, Changxing People's Hospital, Huzhou, Zhejiang 313100, China.
| | - Yanfei Qiang
- Department of Respiration, Changxing People's Hospital, Huzhou, Zhejiang 313100, China.
| | - Yingzhe Wu
- Department of Radiology, Changxing People's Hospital, Huzhou, Zhejiang 313100, China.
| |
Collapse
|
9
|
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
|
10
|
Jiang Y, Du W, Li Y, Gao B, Liu N, Song Q, Wang N, Wu J, Miao Y. Disturbed Dynamic Brain Activity and Neurovascular Coupling in End-Stage Renal Disease Assessed With MRI. J Magn Reson Imaging 2024. [PMID: 39229904 DOI: 10.1002/jmri.29597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/29/2024] [Accepted: 08/09/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Pathophysiological mechanisms underlying cognitive impairment in end-stage renal disease (ESRD) remain unclear, with limited studies on the temporal variability of neural activity and its coupling with regional perfusion. PURPOSE To assess neural activity and neurovascular coupling (NVC) in ESRD patients, evaluate the classification performance of these abnormalities, and explore their relationships with cognitive function. STUDY TYPE Prospective. POPULATION Exactly 33 ESRD patients and 35 age, sex, and education matched healthy controls (HCs). FIELD STRENGTH/SEQUENCE The 3.0T/3D pseudo-continuous arterial spin labeling, resting-state functional MRI, and 3D-T1 weighted structural imaging. ASSESSMENT Dynamic (dfALFF) and static (sfALFF) fractional amplitude of low-frequency fluctuations and cerebral blood flow (CBF) were assessed. CBF-fALFF correlation coefficients and CBF/fALFF ratio were determined for ESRD patients and HCs. Their ability to distinguish ESRD patients from HCs was evaluated, alongside assessment of cerebral small vessel disease (CSVD) MRI features. All participants underwent blood biochemical and neuropsychological tests to evaluate cognitive decline. STATISTICAL TESTS Chi-squared test, two-sample t-test, Mann-Whitney U tests, covariance analysis, partial correlation analysis, family-wise error, false discovery rate, Bonferroni correction, area under the receiver operating characteristic curve (AUC) and multivariate pattern analysis. P < 0.05 denoted statistical significance. RESULTS ESRD patients exhibited higher dfALFF in triangular part of left inferior frontal gyrus (IFGtriang) and left middle temporal gyrus, lower CBF/dfALFF ratio in multiple brain regions, and decreased CBF/sfALFF ratio in bilateral superior temporal gyrus (STG). Compared with CBF/sfALFF ratio, dfALFF, and sfALFF, CBF/dfALFF ratio (AUC = 0.916) achieved the most powerful classification performance in distinguishing ESRD patients from HCs. In ESRD patients, decreased CBF/fALFF ratio correlated with more severe renal impairment, increased CSVD burden, and cognitive decline (0.4 < |r| < 0.6). DATA CONCLUSION ESRD patients exhibited abnormal dynamic brain activity and impaired NVC, with dynamic features demonstrating superior discriminative capacity and CBF/dfALFF ratio showing powerful classification performance. LEVEL OF EVIDENCE 1 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Yuhan Jiang
- Department of Radiology, Zhongshan Hospital Affiliated to Dalian University, Dalian, China
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wei Du
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuan Li
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bingbing Gao
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Na Liu
- Department of Radiology, Zhongshan Hospital Affiliated to Dalian University, Dalian, China
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qingwei Song
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Nan Wang
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jianlin Wu
- Department of Radiology, Zhongshan Hospital Affiliated to Dalian University, Dalian, China
| | - Yanwei Miao
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
11
|
Li H, Li Y, Zhong Q, Chen F, Wang H, Li X, Xie Y, Wang X. Dysfunction of neurovascular coupling in patients with cerebral small vessel disease: A combined resting-state fMRI and arterial spin labeling study. Exp Gerontol 2024; 194:112478. [PMID: 38866193 DOI: 10.1016/j.exger.2024.112478] [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: 04/15/2024] [Revised: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Cerebral small vessel disease (CSVD) closely correlates to cognitive impairment, but its pathophysiology and the neurovascular mechanisms of cognitive deficits were unclear. We aimed to explore the dysfunctional patterns of neurovascular coupling (NVC) in patients with CSVD and further investigate the neurovascular mechanisms of CSVD-related cognitive impairment. METHODS Forty-three patients with CSVD and twenty-four healthy controls were recruited. We adopted resting-state functional magnetic resonance imaging combined with arterial spin labeling to investigate the NVC dysfunctional patterns in patients with CSVD. The Human Brain Atlas with 246 brain regions was applied to extract the NVC coefficients for each brain region. Partial correlation analysis and mediation analysis were used to explore the relationship between CSVD pathological features, NVC dysfunctional patterns, and cognitive decline. RESULTS 8 brain regions with NVC dysfunction were found in patients with CSVD (p < 0.025, Bonferroni correction). The NVC dysfunctional patterns in regions of the default mode network and subcortical nuclei were negatively associated with lacunes, white matter hyperintensities burden, and the severity of CSVD (FDR correction, q < 0.05). The NVC decoupling in regions located in the default mode network positively correlated with delayed recall deficits (FDR correction, q < 0.05). Mediation analysis suggested that the decreased NVC pattern of the left superior frontal gyrus partially mediated the impact of white matter hyperintensities on delayed recall (Mediation effect: -0.119; 95%CI: -11.604,-0.458; p < 0.05). CONCLUSION The findings of this study reveal the NVC dysfunctional pattern in patients with CSVD and illustrate the neurovascular mechanism of CSVD-related cognitive impairment. The NVC function in the left superior frontal gyrus may serve as a promising biomarker and therapeutic target for memory deficits in patients with CSVD.
Collapse
Affiliation(s)
- Hui Li
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China
| | - You Li
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China
| | - Qin Zhong
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China
| | - Faxiang Chen
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China
| | - Hui Wang
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China
| | - Xiang Li
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China
| | - Yuanliang Xie
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China.
| | - Xiang Wang
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430014, China.
| |
Collapse
|
12
|
Zeng S, Ma L, Mao H, Shi Y, Xu M, Gao Q, Kaidong C, Li M, Ding Y, Ji Y, Hu X, Feng W, Fang X. Dynamic functional network connectivity in patients with a mismatch between white matter hyperintensity and cognitive function. Front Aging Neurosci 2024; 16:1418173. [PMID: 39086757 PMCID: PMC11288916 DOI: 10.3389/fnagi.2024.1418173] [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: 04/16/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024] Open
Abstract
Objective White matter hyperintensity (WMH) in patients with cerebral small vessel disease (CSVD) is strongly associated with cognitive impairment. However, the severity of WMH does not coincide fully with cognitive impairment. This study aims to explore the differences in the dynamic functional network connectivity (dFNC) of WMH with cognitively matched and mismatched patients, to better understand the underlying mechanisms from a quantitative perspective. Methods The resting-state functional magnetic resonance imaging (rs-fMRI) and cognitive function scale assessment of the patients were acquired. Preprocessing of the rs-fMRI data was performed, and this was followed by dFNC analysis to obtain the dFNC metrics. Compared the dFNC and dFNC metrics within different states between mismatch and match group, we analyzed the correlation between dFNC metrics and cognitive function. Finally, to analyze the reasons for the differences between the mismatch and match groups, the CSVD imaging features of each patient were quantified with the assistance of the uAI Discover system. Results The 149 CSVD patients included 20 cases of "Type I mismatch," 51 cases of Type I match, 38 cases of "Type II mismatch," and 40 cases of "Type II match." Using dFNC analysis, we found that the fraction time (FT) and mean dwell time (MDT) of State 2 differed significantly between "Type I match" and "Type I mismatch"; the FT of States 1 and 4 differed significantly between "Type II match" and "Type II mismatch." Correlation analysis revealed that dFNC metrics in CSVD patients correlated with executive function and information processing speed among the various cognitive functions. Through quantitative analysis, we found that the number of perivascular spaces and bilateral medial temporal lobe atrophy (MTA) scores differed significantly between "Type I match" and "Type I mismatch," while the left MTA score differed between "Type II match" and "Type II mismatch." Conclusion Different mechanisms were implicated in these two types of mismatch: Type I affected higher-order networks, and may be related to the number of perivascular spaces and brain atrophy, whereas Type II affected the primary networks, and may be related to brain atrophy and the years of education.
Collapse
Affiliation(s)
- Siyuan Zeng
- Medical Imaging Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi, China
| | - Lin Ma
- Medical Imaging Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi, China
| | - Haixia Mao
- Medical Imaging Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi, China
| | - Yachen Shi
- Department of Neurology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi, China
| | - Min Xu
- Medical Imaging Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi, China
| | - Qianqian Gao
- Medical Imaging Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi, China
| | - Chen Kaidong
- Medical Imaging Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi, China
| | - Mingyu Li
- Medical Imaging Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi, China
| | - Yuxiao Ding
- Medical Imaging Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi, China
| | - Yi Ji
- Medical Imaging Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi, China
| | - Xiaoyun Hu
- Medical Imaging Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi, China
| | - Wang Feng
- Department of Neurology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi, China
| | - Xiangming Fang
- Medical Imaging Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi, China
| |
Collapse
|
13
|
Mu R, Yang P, Qin X, Zheng W, Li X, Huang B, Zhu X. Aberrant baseline brain activity and disrupted functional connectivity in patients with vascular cognitive impairment due to cerebral small vessel disease. Front Neurol 2024; 15:1421283. [PMID: 39022734 PMCID: PMC11252026 DOI: 10.3389/fneur.2024.1421283] [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: 04/22/2024] [Accepted: 06/21/2024] [Indexed: 07/20/2024] Open
Abstract
Objective This study aims to examine the alterations in aberrant brain activity and network connectivity between individuals with mild and major vascular cognitive impairment (VCI). Materials and methods A total of 114 patients with cerebral small vessel disease (CSVD) were included in this study, comprising 61 individuals with mild VCI (mean age, 55.7 ± 6.9 years; male, 42.6%) and 53 cases with major VCI (mean age, 57.6 ± 5.5 years; male, 58.5%). Additionally, 53 age-, gender-, and education-matched healthy subjects were recruited as normal controls (NC) (mean age, 54.9 ± 7.9 years; male, 52.9%). All participants underwent neuropsychological assessments and magnetic resonance imaging scans. One-way analysis of variance was used to compare fractional amplitude of low-frequency fluctuation (fALFF) values among the three groups. Two-sample t-tests were conducted to assess functional connectivity matrices between different groups for each connection. Moreover, mediation analyses were performed to explore the mediating effect of aberrant brain activity on the relationship between cognitive impairment and CSVD total burden. Results VCI patients exhibited aberrant brain activity in regions such as the right thalamus (THA_R), right cuneus (CUN_R), left postcentral gyrus (PoCG_L), right postcentral gyrus (PoCG_R), right median cingulate, paracingulate gyri (PCG_R), and left precuneus (PCUN_L). Reduced positive functional connectivity was predominantly observed among nodes including PCUN_L, CUN_R, PoCG_L, PoCG_R, right posterior cingulate (PCG_R), and left occipital gyrus (IOG_L) in VCI patients. The aberrant baseline brain activity and disrupted brain network were more pronounced with worsening cognitive function. Increased fALFF values in THA_R, CUN_R, and PoCG_L mediated cognitive impairment in CSVD patients. Conclusion Abnormal brain activities in THA_R, CUN_R, and PoCG_L, along with their associated abnormal functional connections, play a significant role in VCI. The study revealed a progressive increase in aberrant brain activity and network connectivity with advancing stages of VCI.
Collapse
Affiliation(s)
- Ronghua Mu
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Peng Yang
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Xiaoyan Qin
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Wei Zheng
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Xin Li
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Bingqin Huang
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
- Graduate School, Guilin Medical University, Guilin, China
| | - Xiqi Zhu
- Department of Radiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Life Science and Clinical Medicine Research Center, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
14
|
Xu Y, Wang M, Li X, Lu T, Wang Y, Zhang X, Wang Z, Yan F. Glymphatic dysfunction mediates the influence of choroid plexus enlargement on information processing speed in patients with white matter hyperintensities. Cereb Cortex 2024; 34:bhae265. [PMID: 38912605 DOI: 10.1093/cercor/bhae265] [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: 05/03/2024] [Revised: 06/04/2024] [Accepted: 06/13/2024] [Indexed: 06/25/2024] Open
Abstract
Glymphatic dysfunction has been correlated with cognitive decline, with a higher choroid plexus volume (CPV) being linked to a slower glymphatic clearance rate. Nevertheless, the interplay between CPV, glymphatic function, and cognitive impairment in white matter hyperintensities (WMHs) has not yet been investigated. In this study, we performed neuropsychological assessment, T1-weighted three-dimensional (3D-T1) images, and diffusion tensor imaging (DTI) in a cohort of 206 WMHs subjects and 43 healthy controls (HCs) to further explore the relationship. The DTI analysis along the perivascular space (DTI-ALPS) index, as a measure of glymphatic function, was calculated based on DTI. Severe WMHs performed significantly worse in information processing speed (IPS) than other three groups, as well as in executive function than HCs and mild WMHs. Additionally, severe WMHs demonstrated lower DTI-ALPS index and higher CPV than HCs and mild WMHs. Moderate WMHs displayed higher CPV than HCs and mild WMHs. Mini-Mental State Examination, IPS, and executive function correlated negatively with CPV but positively with DTI-ALPS index in WMHs patients. Glymphatic function partially mediated the association between CPV and IPS, indicating a potential mechanism for WMHs-related cognitive impairment. CPV may act as a valuable prognostic marker and glymphatic system as a promising therapeutic target for WMHs-related cognitive impairment.
Collapse
Affiliation(s)
- Yanan Xu
- Nanjing Medical University, Nanjing, China
- Department of Neurology, ZhongDa Hospital Southeast University (JiangBei) (NanJing DaChang Hospital), Nanjing, China
| | - Mengxue Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiaoli Li
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Tong Lu
- Department of Radiology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yanjuan Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xuezi Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zan Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Fuling Yan
- Nanjing Medical University, Nanjing, China
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
15
|
Wu Y, Ke J, Ye S, Shan LL, Xu S, Guo SF, Li MT, Qiao TC, Peng ZY, Wang YL, Liu MY, Wang H, Feng JF, Han Y. 3D Visualization of Whole Brain Vessels and Quantification of Vascular Pathology in a Chronic Hypoperfusion Model Causing White Matter Damage. Transl Stroke Res 2024; 15:659-671. [PMID: 37222915 DOI: 10.1007/s12975-023-01157-1] [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: 02/20/2023] [Revised: 05/02/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023]
Abstract
Chronic cerebral hypoperfusion is an important pathological factor in many neurodegenerative diseases, such as cerebral small vessel disease (CSVD). One of the most used animal models for chronic cerebral hypoperfusion is the bilateral common carotid artery stenosis (BCAS) mouse. For the therapy of CSVD and other diseases, it will be beneficial to understand the pathological alterations of the BCAS mouse, particularly vascular pathological changes. A mouse model of BCAS was used, and 8 weeks later, cognitive function of the mice was examined by using novel object recognition test and eight-arm radial maze test. 11.7 T magnetic resonance imaging (MRI) and luxol fast blue staining were used to evaluate the injury of the corpus callosum (CC), anterior commissure (AC), internal capsule (IC), and optic tract (Opt) in the cerebral white matter of mice. Three-dimensional vascular images of the whole brain of mice were acquired using fluorescence micro-optical sectioning tomography (fMOST) with a high resolution of 0.32 × 0.32 × 1.00 μm3. Then, the damaged white matter regions were further extracted to analyze the vessel length density, volume fraction, tortuosity, and the number of vessels of different internal diameters. The mouse cerebral caudal rhinal vein was also extracted and analyzed for its branch number and divergent angle in this study. BCAS modeling for 8 weeks resulted in impaired spatial working memory, reduced brain white matter integrity, and myelin degradation in mice, and CC showed the most severe white matter damage. 3D revascularization of the whole mouse brain showed that the number of large vessels was reduced and the number of small vessels was increased in BCAS mice. Further analysis revealed that the vessel length density and volume fraction in the damaged white matter region of BCAS mice were significantly reduced, and the vascular lesions were most noticeable in the CC. At the same time, the number of small vessels in the above white matter regions was significantly reduced, while the number of microvessels was significantly increased in BCAS mice, and the vascular tortuosity was also significantly increased. In addition, the analysis of caudal rhinal vein extraction revealed that the number of branches and the average divergent angle in BCAS mice were significantly reduced. The BCAS modeling for 8 weeks will lead to vascular lesions in whole brain of mice, and the caudal nasal vein was also damaged, while BCAS mice mainly mitigated the damages by increasing microvessels. What is more, the vascular lesions in white matter of mouse brain can cause white matter damage and spatial working memory deficit. These results provide evidence for the vascular pathological alterations caused by chronic hypoperfusion.
Collapse
Affiliation(s)
- Yang Wu
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Jia Ke
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Song Ye
- Wuhan OE-Bio Co., Ltd., G2 zone, Future City 999, Gaoxin boulevard East Lake High-Tech Development zone, Wuhan, 430074, China
| | - Li-Li Shan
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Shuai Xu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 825 Zhangheng Road, Shanghai, 200127, China
| | - Shu-Fen Guo
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Meng-Ting Li
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Tian-Ci Qiao
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Zheng-Yu Peng
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Yi-Lin Wang
- Georgetown Preparatory School, Washington, DC, USA
| | - Ming-Yuan Liu
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - He Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 825 Zhangheng Road, Shanghai, 200127, China.
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 825 Zhangheng Road, Shanghai, 200127, China.
| | - Yan Han
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China.
| |
Collapse
|
16
|
Goyal A, Ekelmans A, Frishman W. Exploring the Intersection of Dementia and Myocardial Infarction: Vascular Perspectives. Cardiol Rev 2024:00045415-990000000-00272. [PMID: 38771949 DOI: 10.1097/crd.0000000000000718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Emerging evidence underscores the relationship between myocardial infarction and dementia, implicating a profound influence on patient health. The bidirectional relationship between myocardial infarction and dementia is highlighted by pathophysiological changes in vasculature function, lifestyle factors, and environmental influences. Our literature review aims to explore the complex relationship between these 2 pathologies and highlight the pathways by which they mutually influence each other.
Collapse
Affiliation(s)
- Anjali Goyal
- From the School of Medicine, New York Medical College, Valhalla, NY
| | | | - William Frishman
- From the School of Medicine, New York Medical College, Valhalla, NY
- Department of Medicine, New York Medical College, Valhalla, NY
| |
Collapse
|
17
|
Lv Y, Cheng X, Dong Q. SGLT1 and SGLT2 inhibition, circulating metabolites, and cerebral small vessel disease: a mediation Mendelian Randomization study. Cardiovasc Diabetol 2024; 23:157. [PMID: 38715111 PMCID: PMC11077823 DOI: 10.1186/s12933-024-02255-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Sodium-glucose cotransporter 2 (SGLT2) and SGLT1 inhibitors may have additional beneficial metabolic effects on circulating metabolites beyond glucose regulation, which could contribute to a reduction in the burden of cerebral small vessel disease (CSVD). Accordingly, we used Mendelian Randomization (MR) to examine the role of circulating metabolites in mediating SGLT2 and SGLT1 inhibition in CSVD. METHODS Genetic instruments for SGLT1/2 inhibition were identified as genetic variants, which were both associated with the expression of encoding genes of SGLT1/2 inhibitors and glycated hemoglobin A1c (HbA1c) level. A two-sample two-step MR was used to determine the causal effects of SGLT1/2 inhibition on CSVD manifestations and the mediating effects of 1400 circulating metabolites linking SGLT1/2 inhibition with CSVD manifestations. RESULTS A lower risk of deep cerebral microbleeds (CMBs) and small vessel stroke (SVS) was linked to genetically predicted SGLT2 inhibition. Better white matter structure integrity was also achieved, as evidenced by decreased mean diffusivity (MD), axial diffusivity (AD), and radial diffusivity (RD), as well as lower deep (DWMH) and periventrivular white matter hyperintensity (PWMH) volume. Inhibiting SGLT2 could also lessen the incidence of severe enlarged perivascular spaces (EPVS) located at white matter, basal ganglia (BG) and hippocampus (HIP). SGLT1 inhibition could preserve white matter integrity, shown as decreased MD of white matter and DWMH volume. The effect of SGLT2 inhibition on SVS and MD of white matter through the concentration of 4-acetamidobutanoate and the cholesterol to oleoyl-linoleoyl-glycerol (18:1 to 18:2) ratio, with a mediated proportion of 30.3% and 35.5% of the total effect, respectively. CONCLUSIONS SGLT2 and SGLT1 inhibition play protective roles in CSVD development. The SGLT2 inhibition could lower the risk of SVS and improve the integrity of white matter microstructure via modulating the level of 4-acetamidobutanoate and cholesterol metabolism. Further mechanistic and clinical studies research are needed to validate our findings.
Collapse
Affiliation(s)
- Yanchen Lv
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
- , 12 Wulumuqi Zhong Road, 200040, Shanghai, P. R. China.
| | - Xin Cheng
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Zhu X, Chu X, Wang H, Liao Z, Xiang H, Zhao W, Yang L, Wu P, Liu X, Chen D, Xie J, Dai W, Li L, Wang J, Zhao H. Investigating neuropathological changes and underlying neurobiological mechanisms in the early stages of primary blast-induced traumatic brain injury: Insights from a rat model. Exp Neurol 2024; 375:114731. [PMID: 38373483 DOI: 10.1016/j.expneurol.2024.114731] [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: 10/29/2023] [Revised: 02/06/2024] [Accepted: 02/14/2024] [Indexed: 02/21/2024]
Abstract
The utilization of explosives and chemicals has resulted in a rise in blast-induced traumatic brain injury (bTBI) in recent times. However, there is a dearth of diagnostic biomarkers and therapeutic targets for bTBI due to a limited understanding of biological mechanisms, particularly in the early stages. The objective of this study was to examine the early neuropathological characteristics and underlying biological mechanisms of primary bTBI. A total of 83 Sprague Dawley rats were employed, with their heads subjected to a blast shockwave of peak overpressure ranging from 172 to 421 kPa in the GI, GII, and GIII groups within a closed shock tube, while the body was shielded. Neuromotor dysfunctions, morphological changes, and neuropathological alterations were detected through modified neurologic severity scores, brain water content analysis, MRI scans, histological, TUNEL, and caspase-3 immunohistochemical staining. In addition, label-free quantitative (LFQ)-proteomics was utilized to investigate the biological mechanisms associated with the observed neuropathology. Notably, no evident damage was discernible in the GII and GI groups, whereas mild brain injury was observed in the GIII group. Neuropathological features of bTBI were characterized by morphologic changes, including neuronal injury and apoptosis, cerebral edema, and cerebrovascular injury in the shockwave's path. Subsequently, 3153 proteins were identified and quantified in the GIII group, with subsequent enriched neurological responses consistent with pathological findings. Further analysis revealed that signaling pathways such as relaxin signaling, hippo signaling, gap junction, chemokine signaling, and sphingolipid signaling, as well as hub proteins including Prkacb, Adcy5, and various G-protein subunits (Gnai2, Gnai3, Gnao1, Gnb1, Gnb2, Gnb4, and Gnb5), were closely associated with the observed neuropathology. The expression of hub proteins was confirmed via Western blotting. Accordingly, this study proposes signaling pathways and key proteins that exhibit sensitivity to brain injury and are correlated with the early pathologies of bTBI. Furthermore, it highlights the significance of G-protein subunits in bTBI pathophysiology, thereby establishing a theoretical foundation for early diagnosis and treatment strategies for primary bTBI.
Collapse
Affiliation(s)
- Xiyan Zhu
- Department of Military Traffic Injury Prevention and Control, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiang Chu
- Cognitive Development and Learning and Memory Disorders Translational Medicine Laboratory, Children's Hospital, Chongqing Medical University, Chongqing, China; Emergency department, Daping Hospital, Army Medical University, Chongqing, China
| | - Hao Wang
- Neurosurgery department, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhikang Liao
- Department of Military Traffic Injury Prevention and Control, Daping Hospital, Army Medical University, Chongqing, China
| | - Hongyi Xiang
- Department of Military Traffic Injury Prevention and Control, Daping Hospital, Army Medical University, Chongqing, China
| | - Wenbing Zhao
- Department of Military Traffic Injury Prevention and Control, Daping Hospital, Army Medical University, Chongqing, China
| | - Li Yang
- Department of Military Traffic Injury Prevention and Control, Daping Hospital, Army Medical University, Chongqing, China
| | - Pengfei Wu
- Department of Military Traffic Injury Prevention and Control, Daping Hospital, Army Medical University, Chongqing, China
| | - Xing Liu
- Department of Military Traffic Injury Prevention and Control, Daping Hospital, Army Medical University, Chongqing, China
| | - Diyou Chen
- Department of Military Traffic Injury Prevention and Control, Daping Hospital, Army Medical University, Chongqing, China
| | - Jingru Xie
- Department of Military Traffic Injury Prevention and Control, Daping Hospital, Army Medical University, Chongqing, China
| | - Wei Dai
- Department of Military Traffic Injury Prevention and Control, Daping Hospital, Army Medical University, Chongqing, China
| | - Lei Li
- Trauma Medical Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Jianmin Wang
- Department of Weapon Bioeffect Assessment, Daping Hospital, Army Medical University, Chongqing, China.
| | - Hui Zhao
- Department of Military Traffic Injury Prevention and Control, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
19
|
Sun W, Huang S, Yang X, Luo Y, Liu L, Wu D. Association of ADAMTS13 activity with cerebral deep medullary vein: A community-based cross-sectional study. Heliyon 2024; 10:e29534. [PMID: 38665574 PMCID: PMC11043937 DOI: 10.1016/j.heliyon.2024.e29534] [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: 05/09/2023] [Revised: 03/27/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Objectives This study aims to investigate whether circulating ADAMTS13 activity can offer insights into the mechanism of pathophysiological changes in deep medullary veins (DMVs). Methods This study was conducted on a community cohort of elderly individuals in Shanghai. Plasma von Willebrand factor (VWF) levels and ADAMTS13 activity were measured. A validated DMV score described the overall burden of DMV on the brain. Through ordinal regression models, we investigated the correlation between VWF levels, ADAMTS13 activity, and increasing severity of DMV score while adjusting for demographics and cardiovascular risk factors. Results The study enrolled 262 subjects according to the inclusion criteria. The mean VWF level (1.35 ± 0.25) was higher in the DMV group than in the group without DMV (1.25 ± 0.30) (p = 0.025), and ADAMTS13 activity (83.76 ± 7.96) was relatively lower. After adjusting for age, sex, alcohol consumption, smoking, hypertension, and diabetes, reduced ADAMTS13 activity [β = -7.78; 95 % CI (-10.21, -5.35) p < 0.01] was associated with DMV. Moreover, correlation analysis indicated that ADAMTS13 activity was negatively correlated with the DMV score (Kendall's tau-b = -0.53, p < 0.001). Discussion In summary, there was an inverse correlation observed between ADAMTS13 activity and the DMV score, which may provide some clinical clues for exploring the potential pathogenesis of DMV.
Collapse
Affiliation(s)
- Wenbo Sun
- Department of Neurology, Fifth People's Hospital of Shanghai Fudan University, Fudan University, Shanghai, China
| | - Shengwen Huang
- Department of Neurology, Fifth People's Hospital of Shanghai Fudan University, Fudan University, Shanghai, China
| | - Xiaoli Yang
- Department of Neurology, Fifth People's Hospital of Shanghai Fudan University, Fudan University, Shanghai, China
| | - Yufan Luo
- Department of Neurology, Fifth People's Hospital of Shanghai Fudan University, Fudan University, Shanghai, China
| | - Luqiong Liu
- Department of General Medicine, Shaoxing People's Hospital, Zhejiang University, Shaoxing, China
| | - Danhong Wu
- Department of Neurology, Fifth People's Hospital of Shanghai Fudan University, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Li Z, Wu M, Yin C, Wang Z, Wang J, Chen L, Zhao W. Machine learning based on the EEG and structural MRI can predict different stages of vascular cognitive impairment. Front Aging Neurosci 2024; 16:1364808. [PMID: 38646447 PMCID: PMC11026635 DOI: 10.3389/fnagi.2024.1364808] [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: 01/03/2024] [Accepted: 03/22/2024] [Indexed: 04/23/2024] Open
Abstract
Background Vascular cognitive impairment (VCI) is a major cause of cognitive impairment in the elderly and a co-factor in the development and progression of most neurodegenerative diseases. With the continuing development of neuroimaging, multiple markers can be combined to provide richer biological information, but little is known about their diagnostic value in VCI. Methods A total of 83 subjects participated in our study, including 32 patients with vascular cognitive impairment with no dementia (VCIND), 21 patients with vascular dementia (VD), and 30 normal controls (NC). We utilized resting-state quantitative electroencephalography (qEEG) power spectra, structural magnetic resonance imaging (sMRI) for feature screening, and combined them with support vector machines to predict VCI patients at different disease stages. Results The classification performance of sMRI outperformed qEEG when distinguishing VD from NC (AUC of 0.90 vs. 0,82), and sMRI also outperformed qEEG when distinguishing VD from VCIND (AUC of 0.8 vs. 0,0.64), but both underperformed when distinguishing VCIND from NC (AUC of 0.58 vs. 0.56). In contrast, the joint model based on qEEG and sMRI features showed relatively good classification accuracy (AUC of 0.72) to discriminate VCIND from NC, higher than that of either qEEG or sMRI alone. Conclusion Patients at varying stages of VCI exhibit diverse levels of brain structure and neurophysiological abnormalities. EEG serves as an affordable and convenient diagnostic means to differentiate between different VCI stages. A machine learning model that utilizes EEG and sMRI as composite markers is highly valuable in distinguishing diverse VCI stages and in individually tailoring the diagnosis.
Collapse
Affiliation(s)
- Zihao Li
- Department of Neurology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
- Department of Neurology, Taizhou Second People’s Hospital, Taizhou, Zhejiang, China
| | - Meini Wu
- Department of Neurology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
- Department of Neurology, Taizhou Second People’s Hospital, Taizhou, Zhejiang, China
| | - Changhao Yin
- Department of Neurology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Zhenqi Wang
- Department of Neurology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Jianhang Wang
- Department of Neurology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
- Mudanjiang Medical College, Mudanjiang, China
| | - Lingyu Chen
- Department of Neurology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
- Mudanjiang Medical College, Mudanjiang, China
| | - Weina Zhao
- Department of Neurology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
- Center for Mudanjiang North Medicine Resource Development and Application Collaborative Innovation, Mudanjiang, China
| |
Collapse
|
21
|
Wallace C, Smirl JD, Adhikari SP, Jones KE, Rieger M, Rothlander K, van Donkelaar P. Neurovascular coupling is altered in women who have a history of brain injury from intimate partner violence: a preliminary study. Front Glob Womens Health 2024; 5:1344880. [PMID: 38495125 PMCID: PMC10940333 DOI: 10.3389/fgwh.2024.1344880] [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: 11/26/2023] [Accepted: 02/22/2024] [Indexed: 03/19/2024] Open
Abstract
Introduction Intimate partner violence (IPV) is a global health crisis with 30% of women over the age of 15 experiencing at least one event in their lifetime. Brain injury (BI) due to head impacts and/or strangulation is a common but understudied part of this experience. Previous research has shown BI from other injury mechanisms can disrupt neurovascular coupling (NVC). To gain further insight into whether similar changes occur in this population, we assessed NVC responses in women with a history of IPV-BI. Methods NVC responses were measured for the middle and posterior cerebral arteries (MCA, PCA) using transcranial Doppler ultrasound while participants performed a complex visual search task. The lifetime history of previous exposure to IPV-BI was captured using the Brain Injury Severity Assessment (BISA) along with measures of post-traumatic stress disorder (PTSD), anxiety, depression, substance use, and demographic information. Initial analyses of NVC metrics were completed comparing participants who scored low vs. high on the BISA or did or did not experience non-fatal strangulation followed by a stepwise multiple regression to examine the impact of PTSD, anxiety, and depression on the relationship between the NVC metrics and IPV-BI. Results Baseline and peak cerebral blood velocity were higher and the percentage increase was lower in the PCA in the low compared to the high BISA group whereas no differences between the groups were apparent in the MCA. In addition, those participants who had been strangled had a lower initial slope and area under the curve in the PCA than those who had not experienced strangulation. Finally, the stepwise multiple regression demonstrated the percentage increase in the PCA was significantly related to the BISA score and both depression and anxiety significantly contributed to different components of the NVC response. Conclusions This preliminary study demonstrated that a lifetime history of IPV-BI leads to subtle but significant disruptions to NVC responses which are modulated by comorbid depression and anxiety. Future studies should examine cerebrovascular function at the acute and subacute stages after IPV episodes to shed additional light on this experience and its outcomes.
Collapse
Affiliation(s)
- Colin Wallace
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
- Department of Kinesiology, Okanagan College, Penticton, BC, Canada
| | - Jonathan D. Smirl
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Cerebrovascular Concussion Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Integrated Concussion Research Program, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - Shambhu P. Adhikari
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| | - K. Elisabeth Jones
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| | - Matt Rieger
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
- Faculty of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Krystal Rothlander
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| | - Paul van Donkelaar
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| |
Collapse
|
22
|
Chen DH, Huang JR, Su SL, Chen Q, Wu BY. Therapeutic potential of mesenchymal stem cells for cerebral small vessel disease. Regen Ther 2024; 25:377-386. [PMID: 38414558 PMCID: PMC10899004 DOI: 10.1016/j.reth.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/12/2023] [Accepted: 11/05/2023] [Indexed: 02/29/2024] Open
Abstract
Cerebral small vessel disease (CSVD), as the most common, chronic and progressive vascular disease on the brain, is a serious neurological disease, whose pathogenesis remains unclear. The disease is a leading cause of stroke and vascular cognitive impairment and dementia, and contributes to about 20% of strokes, including 25% of ischemic strokes and 45% of dementias. Undoubtedly, the high incidence and poor prognosis of CSVD have brought a heavy economic and medical burden to society. The present treatment of CSVD focuses on the management of vascular risk factors. Although vascular risk factors may be important causes or accelerators of CSVD and should always be treated in accordance with best clinical practice, controlling risk factors alone could not curb the progression of CSVD brain injury. Therefore, developing safer and more effective treatment strategies for CSVD is urgently needed. Recently, mesenchymal stem cells (MSCs) therapy has become an emerging therapeutic modality for the treatment of central nervous system disease, given their paracrine properties and immunoregulatory. Herein, we discussed the therapeutic potential of MSCs for CSVD, aiming to enable clinicians and researchers to understand of recent progress and future directions in the field.
Collapse
Affiliation(s)
- Dong-Hua Chen
- Neurology Department, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Baiyun District, Guangzhou, 510515, China
| | - Jia-Rong Huang
- Neurology Department, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Baiyun District, Guangzhou, 510515, China
| | - Shuo-Lei Su
- Shaoguan University, No.288 University Road, Xinshaozhen Zhenjiang District, Shaoguan, 512005, China
| | - Qiong Chen
- Medical Research center of Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Baiyun District, Guangzhou, 510515, China
- Precision Medicine Center of Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Baiyun District, Guangzhou, 510515, China
| | - Bing-Yi Wu
- Medical Research center of Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Baiyun District, Guangzhou, 510515, China
- Precision Medicine Center of Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Baiyun District, Guangzhou, 510515, China
| |
Collapse
|
23
|
Voorter PHM, van Dinther M, Jansen WJ, Postma AA, Staals J, Jansen JFA, van Oostenbrugge RJ, van der Thiel MM, Backes WH. Blood-Brain Barrier Disruption and Perivascular Spaces in Small Vessel Disease and Neurodegenerative Diseases: A Review on MRI Methods and Insights. J Magn Reson Imaging 2024; 59:397-411. [PMID: 37658640 DOI: 10.1002/jmri.28989] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/17/2023] [Accepted: 08/17/2023] [Indexed: 09/03/2023] Open
Abstract
Perivascular spaces (PVS) and blood-brain barrier (BBB) disruption are two key features of cerebral small vessel disease (cSVD) and neurodegenerative diseases that have been linked to cognitive impairment and are involved in the cerebral waste clearance system. Magnetic resonance imaging (MRI) offers the possibility to study these pathophysiological processes noninvasively in vivo. This educational review provides an overview of the MRI techniques used to assess PVS functionality and BBB disruption. MRI-visible PVS can be scored on structural images by either (subjectively) counting or (automatically) delineating the PVS. We highlight emerging (diffusion) techniques to measure proxies of perivascular fluid and its movement, which may provide a more comprehensive understanding of the role of PVS in diseases. For the measurement of BBB disruption, we explain the most commonly used MRI technique, dynamic contrast-enhanced (DCE) MRI, as well as a more recently developed technique based on arterial spin labeling (ASL). DCE MRI and ASL are thought to measure complementary characteristics of the BBB. Furthermore, we describe clinical studies that have utilized these MRI techniques in cSVD and neurodegenerative diseases, particularly Alzheimer's disease (AD). These studies demonstrate the role of PVS and BBB dysfunction in these diseases and provide insight into the large overlap, but also into the differences between cSVD and AD. Overall, MRI techniques may provide valuable insights into the pathophysiological mechanisms underlying these diseases and have the potential to be used as markers for disease progression and treatment response. LEVEL OF EVIDENCE: 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Paulien H M Voorter
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Maud van Dinther
- School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
- Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Willemijn J Jansen
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, Maastricht University, Maastricht, the Netherlands
| | - Alida A Postma
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Julie Staals
- School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
- Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Robert J van Oostenbrugge
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
- Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Merel M van der Thiel
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, Maastricht University, Maastricht, the Netherlands
| | - Walter H Backes
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
24
|
Liao L, Wang H, Wei D, Yi M, Gu Y, Zhang M, Wang L. Exosomal microRNAs: implications in the pathogenesis and clinical applications of subarachnoid hemorrhage. Front Mol Neurosci 2023; 16:1300864. [PMID: 38143562 PMCID: PMC10748509 DOI: 10.3389/fnmol.2023.1300864] [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: 09/23/2023] [Accepted: 11/22/2023] [Indexed: 12/26/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) is a severe acute neurological disorder with a high fatality rate. Early brain injury (EBI) and cerebral vasospasm are two critical complications of SAH that significantly contribute to poor prognosis. Currently, surgical intervention and interventional therapy are the main treatment options for SAH, but their effectiveness is limited. Exosomes, which are a type of extracellular vesicles, play a crucial role in intercellular communication and have been extensively studied in the past decade due to their potential influence on disease progression, diagnosis, and treatment. As one of the most important components of exosomes, miRNA plays both direct and indirect roles in affecting disease progression. Previous research has found that exosomal miRNA is involved in the development of various diseases, such as tumors, chronic hepatitis, atherosclerosis, diabetes, and SAH. This review focuses on exploring the impact of exosomal miRNA on SAH, including its influence on neuronal apoptosis, inflammatory response, and immune activation following SAH. Furthermore, this review highlights the potential clinical applications of exosomal miRNA in the treatment of SAH. Although current research on this topic is limited and the clinical application of exosomal miRNA has inherent limitations, we aim to provide a concise summary of existing research progress and offer new insights for future research directions and trends in this field.
Collapse
Affiliation(s)
- Lishang Liao
- Department of Neurosurgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Haoran Wang
- Department of Neurosurgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Deli Wei
- Department of Neurosurgery, The People’s Hospital of Fushun County, Zigong, China
| | - Mingliang Yi
- Department of Neurosurgery, The People’s Hospital of Fushun County, Zigong, China
| | - Yingjiang Gu
- Department of Neurosurgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Department of Neurosurgery, The People’s Hospital of Fushun County, Zigong, China
| | - Mingwei Zhang
- Department of Neurosurgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Li Wang
- Department of Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
25
|
Su C, Miao J, Guo J. The relationship between TGF-β1 and cognitive function in the brain. Brain Res Bull 2023; 205:110820. [PMID: 37979810 DOI: 10.1016/j.brainresbull.2023.110820] [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: 09/13/2023] [Revised: 11/05/2023] [Accepted: 11/15/2023] [Indexed: 11/20/2023]
Abstract
Transforming growth factor-β1 (TGF-β1), a multifunctional cytokine, plays a pivotal role in synaptic formation, plasticity, and neurovascular unit regulation. This review highlights TGF-β1's potential impact on cognitive function, particularly in the context of neurodegenerative disorders. However, despite the growing body of evidence, a comprehensive understanding of TGF-β1's precise role remains elusive. Further research is essential to unravel the complex mechanisms through which TGF-β1 influences cognitive function and to explore therapeutic avenues for targeting TGF-β1 in neurodegenerative conditions. This investigation sheds light on TGF-β1's contribution to cognitive function and offers prospects for innovative treatments and interventions. This review delves into the intricate relationship between TGF-β1 and cognitive function.
Collapse
Affiliation(s)
- Chen Su
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province 030000, China
| | - Jie Miao
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province 030000, China
| | - Junhong Guo
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province 030000, China.
| |
Collapse
|
26
|
Li Y, Chen J, Tu H, Ma Q, Wang M, Chen J, Chen M. Protective effects of GuanXinNing tablet (GXNT) on diabetic encephalopathy in zucker diabetic obesity (ZDF) rats. BMC Complement Med Ther 2023; 23:385. [PMID: 37891536 PMCID: PMC10605859 DOI: 10.1186/s12906-023-04195-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Diabetic encephalopathy (DE) is a complication of diabetes that leads to cognitive and behavioral decline. Utilizing safe and effective complementary and alternative medications for its management is a wise choice. Previous studies have shown that GuanXinNing Tablet (GXNT), an oral preparation primarily derived from two Chinese herbs, Salvia miltiorrhiza Bge. and Ligusticum chuanxiong Hort., exerts a beneficial neuroprotective effect. In this study, we explored the protective effects of GXNT on DE in male Zucker diabetic fatty (ZDF) rats induced by a high-fat diet, aiming to ascertain its significance and potential mechanisms. METHODS ZDF rats were induced to develop type 2 diabetes (T2DM) with DE by a high-fat diet and treated with GXNT for 8 weeks until they were 20 weeks old. Throughout the experiment, the animals' vital parameters, such as body weight, were continuously monitored. Cognitive function was evaluated using the Y maze test. Biochemical kits were employed to analyze blood glucose, lipids, and vascular endothelial-related factors. Cerebrovascular lesions were assessed using magnetic resonance angiography (MRA) imaging. Brain lesions were evaluated using hematoxylin and eosin (H&E) staining and ultrastructure observation. IgG and albumin (ALB) leakage were detected using immunofluorescence. RESULTS GXNT demonstrated an enhancement in the overall well-being of the animals. It notably improved cognitive and behavioral abilities, as demonstrated by extended retention time in the novel heterogeneous arm during the Y-maze test. GXNT effectively regulated glucose and lipid metabolism, reducing fasting and postprandial blood glucose, glycated hemoglobin (HbA1c), and total cholesterol (TC) levels. Additionally, it exhibited a protective effect on the vascular endothelium by reducing the serum TXB2/PGI2 ratio while elevating NO and PGI2 levels. Moreover, GXNT ameliorated stenosis and occlusion in cerebral vessel branches, increased the number of microvessels and neurons around the hippocampus, and improved microvascular occlusion in the cerebral cortex, along with addressing perivascular cell abnormalities. Immunofluorescence staining showed a decrease in the fluorescence intensity of IgG and ALB in the cerebral cortex. CONCLUSIONS GXNT demonstrated a highly satisfactory protective effect on DE in ZDF rats. Its mechanism of action could be based on the regulation of glucolipid metabolism and its protective effect on the vascular endothelium.
Collapse
Affiliation(s)
- Yajing Li
- The Department of Biopharmaceutical Technology, Zhejiang Institute of Economics and Trade, Hangzhou, 310018, China
| | - Jiaojiao Chen
- Institute of Comparative Medicine, Experimental Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310012, China
| | - Haiye Tu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Quanxin Ma
- Institute of Comparative Medicine, Experimental Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mulan Wang
- Institute of Comparative Medicine, Experimental Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- The Department of Medicine, Chiatai Qingchunbao Pharmaceutical Co., Ltd, Hangzhou, 310053, China
| | - Jie Chen
- Department of Vasculocardiology, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310006, China.
| | - Minli Chen
- Institute of Comparative Medicine, Experimental Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
27
|
Gao Y, Wang S, Xin H, Feng M, Zhang Q, Sui C, Guo L, Liang C, Wen H. Disrupted Gray Matter Networks Associated with Cognitive Dysfunction in Cerebral Small Vessel Disease. Brain Sci 2023; 13:1359. [PMID: 37891728 PMCID: PMC10605932 DOI: 10.3390/brainsci13101359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
This study aims to investigate the disrupted topological organization of gray matter (GM) structural networks in cerebral small vessel disease (CSVD) patients with cerebral microbleeds (CMBs). Subject-wise structural networks were constructed from GM volumetric features of 49 CSVD patients with CMBs (CSVD-c), 121 CSVD patients without CMBs (CSVD-n), and 74 healthy controls. The study used graph theory to analyze the global and regional properties of the network and their correlation with cognitive performance. We found that both the control and CSVD groups exhibited efficient small-world organization in GM networks. However, compared to controls, CSVD-c and CSVD-n patients exhibited increased global and local efficiency (Eglob/Eloc) and decreased shortest path lengths (Lp), indicating increased global integration and local specialization in structural networks. Although there was no significant global topology change, partially reorganized hub distributions were found between CSVD-c and CSVD-n patients. Importantly, regional topology in nonhub regions was significantly altered between CSVD-c and CSVD-n patients, including the bilateral anterior cingulate gyrus, left superior parietal gyrus, dorsolateral superior frontal gyrus, and right MTG, which are involved in the default mode network (DMN) and sensorimotor functional modules. Intriguingly, the global metrics (Eglob, Eloc, and Lp) were significantly correlated with MoCA, AVLT, and SCWT scores in the control group but not in the CSVD-c and CSVD-n groups. In contrast, the global metrics were significantly correlated with the SDMT score in the CSVD-s and CSVD-n groups but not in the control group. Patients with CSVD show a disrupted balance between local specialization and global integration in their GM structural networks. The altered regional topology between CSVD-c and CSVD-n patients may be due to different etiological contributions, which may offer a novel understanding of the neurobiological processes involved in CSVD with CMBs.
Collapse
Affiliation(s)
- Yian Gao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China; (Y.G.); (C.S.)
| | - Shengpei Wang
- Research Center for Brain-Inspired Intelligence, Institute of Automation, Chinese Academy of Sciences, Beijing 100040, China;
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Haotian Xin
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, No. 45 Chang-Chun St., Xicheng District, Beijing 100054, China; (H.X.); (M.F.)
| | - Mengmeng Feng
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, No. 45 Chang-Chun St., Xicheng District, Beijing 100054, China; (H.X.); (M.F.)
| | - Qihao Zhang
- Department of Radiology, Weill Cornell Medical College, New York. 407 East 61st Street, New York, NY 10044, USA;
| | - Chaofan Sui
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China; (Y.G.); (C.S.)
| | - Lingfei Guo
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China; (Y.G.); (C.S.)
| | - Changhu Liang
- Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jing-Wu Road No. 324, Jinan 250021, China
| | - Hongwei Wen
- Key Laboratory of Cognition and Personality (Ministry of Education), Faculty of Psychology, Southwest University, Chongqing 400715, China
| |
Collapse
|
28
|
Bennett J, van Dinther M, Voorter P, Backes W, Barnes J, Barkhof F, Captur G, Hughes AD, Sudre C, Treibel TA. Assessment of Microvascular Disease in Heart and Brain by MRI: Application in Heart Failure with Preserved Ejection Fraction and Cerebral Small Vessel Disease. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1596. [PMID: 37763715 PMCID: PMC10534635 DOI: 10.3390/medicina59091596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023]
Abstract
The objective of this review is to investigate the commonalities of microvascular (small vessel) disease in heart failure with preserved ejection fraction (HFpEF) and cerebral small vessel disease (CSVD). Furthermore, the review aims to evaluate the current magnetic resonance imaging (MRI) diagnostic techniques for both conditions. By comparing the two conditions, this review seeks to identify potential opportunities to improve the understanding of both HFpEF and CSVD.
Collapse
Affiliation(s)
- Jonathan Bennett
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Department of Cardiology, Barts Heart Centre, London EC1A 7BE, UK
| | - Maud van Dinther
- Department of Neurology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LX Maastricht, The Netherlands
| | - Paulien Voorter
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- School for Mental Health & Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Walter Backes
- School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LX Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- School for Mental Health & Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Josephine Barnes
- Dementia Research Centre, UCL Queens Square Institute of Neurology, University College London, London WC1E 6BT, UK
| | - Frederick Barkhof
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije University, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands
- Queen Square Institute of Neurology, University College London, London WC1E 6BT, UK
- Centre for Medical Image Computing, University College London, London WC1E 6BT, UK
| | - Gabriella Captur
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Medical Research Council Unit for Lifelong Health and Ageing, Department of Population Science and Experimental Medicine, University College London, London WC1E 6BT, UK
- Centre for Inherited Heart Muscle Conditions, Cardiology Department, The Royal Free Hospital, London NW3 2QG, UK
| | - Alun D. Hughes
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Medical Research Council Unit for Lifelong Health and Ageing, Department of Population Science and Experimental Medicine, University College London, London WC1E 6BT, UK
| | - Carole Sudre
- Dementia Research Centre, UCL Queens Square Institute of Neurology, University College London, London WC1E 6BT, UK
- Centre for Medical Image Computing, University College London, London WC1E 6BT, UK
- Medical Research Council Unit for Lifelong Health and Ageing, Department of Population Science and Experimental Medicine, University College London, London WC1E 6BT, UK
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London WC2R 2LS, UK
| | - Thomas A. Treibel
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Department of Cardiology, Barts Heart Centre, London EC1A 7BE, UK
| |
Collapse
|
29
|
Inoue Y, Shue F, Bu G, Kanekiyo T. Pathophysiology and probable etiology of cerebral small vessel disease in vascular dementia and Alzheimer's disease. Mol Neurodegener 2023; 18:46. [PMID: 37434208 PMCID: PMC10334598 DOI: 10.1186/s13024-023-00640-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/28/2023] [Indexed: 07/13/2023] Open
Abstract
Vascular cognitive impairment and dementia (VCID) is commonly caused by vascular injuries in cerebral large and small vessels and is a key driver of age-related cognitive decline. Severe VCID includes post-stroke dementia, subcortical ischemic vascular dementia, multi-infarct dementia, and mixed dementia. While VCID is acknowledged as the second most common form of dementia after Alzheimer's disease (AD) accounting for 20% of dementia cases, VCID and AD frequently coexist. In VCID, cerebral small vessel disease (cSVD) often affects arterioles, capillaries, and venules, where arteriolosclerosis and cerebral amyloid angiopathy (CAA) are major pathologies. White matter hyperintensities, recent small subcortical infarcts, lacunes of presumed vascular origin, enlarged perivascular space, microbleeds, and brain atrophy are neuroimaging hallmarks of cSVD. The current primary approach to cSVD treatment is to control vascular risk factors such as hypertension, dyslipidemia, diabetes, and smoking. However, causal therapeutic strategies have not been established partly due to the heterogeneous pathogenesis of cSVD. In this review, we summarize the pathophysiology of cSVD and discuss the probable etiological pathways by focusing on hypoperfusion/hypoxia, blood-brain barriers (BBB) dysregulation, brain fluid drainage disturbances, and vascular inflammation to define potential diagnostic and therapeutic targets for cSVD.
Collapse
Affiliation(s)
- Yasuteru Inoue
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Guojun Bu
- SciNeuro Pharmaceuticals, Rockville, MD 20850 USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| |
Collapse
|
30
|
Wang Y, Du W, Sun Y, Zhang J, Ma C, Jin X. CRTC1 is a potential target to delay aging-induced cognitive deficit by protecting the integrity of the blood-brain barrier via inhibiting inflammation. J Cereb Blood Flow Metab 2023; 43:1042-1059. [PMID: 37086081 PMCID: PMC10291461 DOI: 10.1177/0271678x231169133] [Citation(s) in RCA: 5] [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: 09/30/2022] [Revised: 03/09/2023] [Accepted: 03/24/2023] [Indexed: 04/23/2023]
Abstract
Aging can cause attenuation in the functioning of multiple organs, and blood-brain barrier (BBB) breakdown could promote the occurrence of disorders of the central nervous system during aging. Since inflammation is considered to be an important factor underlying BBB injury during aging, vascular endothelial cell senescence serves as a critical pathological basis for the destruction of BBB integrity. In the current review, we have first introduced the concepts related to aging-induced cognitive deficit and BBB integrity damage. Thereafter, we reviewed the potential relationship between disruption of BBB integrity and cognition deficit and the role of inflammation, vascular endothelial cell senescence, and BBB injury. We have also briefly introduced the function of CREB-regulated transcription co-activator 1 (CRTC1) in cognition and aging-induced CRTC1 changes as well as the critical roles of CRTC1/cyclooxygenase-2 (COX-2) in regulating inflammation, endothelial cell senescence, and BBB injury. Finally, the underlying mechanisms have been summarized and we propose that CRTC1 could be a promising target to delay aging-induced cognitive deficit by protecting the integrity of BBB through promoting inhibition of inflammation-mediated endothelial cell senescence.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Neurology, the Second Hospital of Jiaxing City, Jiaxing, China
| | - Weihong Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yanyun Sun
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junfang Zhang
- Department of Physiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Chaolin Ma
- School of Life Science and Institute of Life Science, Nanchang University, Nanchang, China
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
31
|
Du H, Xia J, Huang L, Zheng L, Gu W, Yi F. Relationship between insulin-like growth factor-1 and cerebral small vessel disease and its mechanisms: advances in the field. Front Aging Neurosci 2023; 15:1190869. [PMID: 37358957 PMCID: PMC10285072 DOI: 10.3389/fnagi.2023.1190869] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Insulin-like growth factor-1 (IGF-1) is an active polypeptide protein that closely resembles the structural sequence of insulin and is involved in a variety of metabolic processes in the body. Decreased IGF-1 circulation levels are associated with an increased risk of stroke and a poorer prognosis, but the relationship with cerebral small vessel disease (cSVD) is unclear. Some studies found that the level of IGF-1 in patients with cSVD was significantly reduced, but the clinical significance and underlying mechanisms are unknown. This article reviews the correlation between IGF-1 and cerebrovascular disease and explores the potential relationship and mechanism between IGF-1 and cSVD.
Collapse
Affiliation(s)
- Hao Du
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Xia
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Huang
- Department of Rehabilitation, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Lan Zheng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenping Gu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fang Yi
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
32
|
Yuan H, Zhu B, Li C, Zhao Z. Ceramide in cerebrovascular diseases. Front Cell Neurosci 2023; 17:1191609. [PMID: 37333888 PMCID: PMC10272456 DOI: 10.3389/fncel.2023.1191609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/18/2023] [Indexed: 06/20/2023] Open
Abstract
Ceramide, a bioactive sphingolipid, serves as an important second messenger in cell signal transduction. Under stressful conditions, it can be generated from de novo synthesis, sphingomyelin hydrolysis, and/or the salvage pathway. The brain is rich in lipids, and abnormal lipid levels are associated with a variety of brain disorders. Cerebrovascular diseases, which are mainly caused by abnormal cerebral blood flow and secondary neurological injury, are the leading causes of death and disability worldwide. There is a growing body of evidence for a close connection between elevated ceramide levels and cerebrovascular diseases, especially stroke and cerebral small vessel disease (CSVD). The increased ceramide has broad effects on different types of brain cells, including endothelial cells, microglia, and neurons. Therefore, strategies that reduce ceramide synthesis, such as modifying sphingomyelinase activity or the rate-limiting enzyme of the de novo synthesis pathway, serine palmitoyltransferase, may represent novel and promising therapeutic approaches to prevent or treat cerebrovascular injury-related diseases.
Collapse
|
33
|
Granata A. Functional genomics in stroke: current and future applications of iPSCs and gene editing to dissect the function of risk variants. BMC Cardiovasc Disord 2023; 23:223. [PMID: 37120540 PMCID: PMC10148993 DOI: 10.1186/s12872-023-03227-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 04/04/2023] [Indexed: 05/01/2023] Open
Abstract
Stroke is an important disease with unmet clinical need. To uncover novel paths for treatment, it is of critical importance to develop relevant laboratory models that may help to shed light on the pathophysiological mechanisms of stroke. Induced pluripotent stem cells (iPSCs) technology has enormous potential to advance our knowledge into stroke by creating novel human models for research and therapeutic testing. iPSCs models generated from patients with specific stroke types and specific genetic predisposition in combination with other state of art technologies including genome editing, multi-omics, 3D system, libraries screening, offer the opportunity to investigate disease-related pathways and identify potential novel therapeutic targets that can then be tested in these models. Thus, iPSCs offer an unprecedented opportunity to make rapid progress in the field of stroke and vascular dementia research leading to clinical translation. This review paper summarizes some of the key areas in which patient-derived iPSCs technology has been applied to disease modelling and discusses the ongoing challenges and the future directions for the application of this technology in the field of stroke research.
Collapse
Affiliation(s)
- Alessandra Granata
- Department of Clinical Neurosciences, Victor Phillip Dahdaleh Heart & Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0BB, UK.
| |
Collapse
|
34
|
Brisson RT, Fernandes RDCL, Arruda JFDL, Rocha TCCDSM, Santos NDGD, Silva LD, de Lima MASD, de Rosso ALZ. Altered Cerebral Vasoreactivity on Transcranial Color-Coded Sonography Related to Akinetic-Rigid Phenotype of Parkinson's Disease: Interim Analysis of a Cross-Sectional Study. Brain Sci 2023; 13:brainsci13050709. [PMID: 37239181 DOI: 10.3390/brainsci13050709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND A correlation between worse functional outcomes in Parkinson's disease (PD) patients with cerebrovascular disease (CVD) or the Akinetic-rigid phenotype has been argued in recent studies. We aimed to evaluate the association of cerebral hemodynamics impairments, assessed by Transcranial Color-coded Doppler sonography (TCCS), on PD patients with different phenotypes of the disease and with risk factors for CVD. METHODOLOGY Idiopathic PD patients (n = 51) were divided into motor subtypes: Akinetic-rigid (AR) (n = 27) and Tremor-dominant (TD) (n = 24) and into two groups regarding vascular risk factors: when ≥2 were present (PDvasc) (n = 18) and <2 (PDnvasc) (n = 33). In a parallel analysis, the Fazekas scale on brain magnetic resonance imaging (MRI) was applied to a sample to assess the degree of leukoaraiosis. TCCS examinations were prospectively performed obtaining middle cerebral artery Mean Flow Velocities (Vm), Resistance Index (RI), and Pulsatility Index (PI). The Breath-Holding Index (BHI) was calculated to assess cerebrovascular reactivity (cVR). Standardized functional scales were administered (UPDRS III and Hoehn&Yahr). RESULTS The phenotype groups were similar in age, disease duration and demographic parameters, but there were significantly higher H&Y scores than TD group. cVR was impaired in 66.7% of AR vs. 37.5% of TD. AR group exhibited lower BHI (0.53 ± 0.31 vs. 0.91 ± 0.62; p = 0.000), lower Vm after apnea (44.3 ± 9.0 cm/s vs. 53.4 ± 11.4 cm/s; p = 0.003), higher PI (0.91 ± 0.26 vs. 0.76 ± 0.12; p = 0.000) and RI (0.58 ± 0.11 vs. 0.52 ± 0.06; p = 0.021). PDvasc group showed higher PI (0.98 vs. 0.76; p = 0.001) and higher frequency of altered cVR (72.2% vs. 42.2%; p = 0.004). There was a significant predominance of higher values on Fazekas scale in the PDvasc group. We found no difference between the Fazekas scale when comparing motor subtypes groups but there was a trend toward higher scores in the AR phenotype. CONCLUSIONS TCCS, a cost-effective method, displayed impaired cVR in Parkinsonian patients with risk factors for CVD with higher degree of MRI leukoaraiosis. PD patients with the AR disease phenotype also presented impaired cVR on TCCS and greater functional impairment, although with just a trend to higher scores on MRI Fazekas.
Collapse
Affiliation(s)
- Rodrigo Tavares Brisson
- Department of Neurology, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil
| | - Rita de Cássia Leite Fernandes
- Department of Neurology, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil
| | - Josevânia Fulgêncio de Lima Arruda
- Department of Neurology, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil
| | | | - Nathália de Góes Duarte Santos
- Department of Neurology, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil
| | - Liene Duarte Silva
- Department of Neurology, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil
| | - Marco Antônio Sales Dantas de Lima
- Department of Neurology, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil
| | - Ana Lucia Zuma de Rosso
- Department of Neurology, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil
| |
Collapse
|
35
|
Li X, Oh JS, Lee Y, Lee EC, Yang M, Kwon N, Ha TW, Hong DY, Song Y, Kim HK, Song BH, Choi S, Lee MR, Yoon J. Albumin-binding photosensitizer capable of targeting glioma via the SPARC pathway. Biomater Res 2023; 27:23. [PMID: 36945032 PMCID: PMC10031904 DOI: 10.1186/s40824-023-00360-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/05/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Malignant glioma is among the most lethal and frequently occurring brain tumors, and the average survival period is 15 months. Existing chemotherapy has low tolerance and low blood-brain barrier (BBB) permeability; therefore, the required drug dose cannot be accurately delivered to the tumor site, resulting in an insufficient drug effect. METHODS Herein, we demonstrate a precision photodynamic tumor therapy using a photosensitizer (ZnPcS) capable of binding to albumin in situ, which can increase the permeability of the BBB and accurately target glioma. Albumin-binding ZnPcS was designed to pass through the BBB and bind to secreted protein acidic and rich in cysteine (SPARC), which is abundant in the glioma plasma membrane. RESULTS When the upper part of a mouse brain was irradiated using a laser (0.2 W cm- 2) after transplantation of glioma and injection of ZnPcS, tumor growth was inhibited by approximately 83.6%, and the 50% survival rate of the treatment group increased by 14 days compared to the control group. In glioma with knockout SPARC, the amount of ZnPcS entering the glioma was reduced by 63.1%, indicating that it can target glioma through the SPARC pathway. CONCLUSION This study showed that the use of albumin-binding photosensitizers is promising for the treatment of malignant gliomas.
Collapse
Affiliation(s)
- Xingshu Li
- Fujian Provincial Key Laboratory for Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, China
| | - Jae Sang Oh
- Department of Neurosurgery, College of Medicine, Cheonan Hospital, Soonchunhyang University, Cheonan-si, Chungcheongnam-do, Republic of Korea
- Department of Neurosurgery, Uijeonbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoonji Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Eun Chae Lee
- Department of Neurosurgery, College of Medicine, Cheonan Hospital, Soonchunhyang University, Cheonan-si, Chungcheongnam-do, Republic of Korea
| | - Mengyao Yang
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Republic of Korea
| | - Nahyun Kwon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Republic of Korea
| | - Tae Won Ha
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungcheongnam-do, Republic of Korea
| | - Dong-Yong Hong
- Department of Neurosurgery, College of Medicine, Cheonan Hospital, Soonchunhyang University, Cheonan-si, Chungcheongnam-do, Republic of Korea
| | - Yena Song
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungcheongnam-do, Republic of Korea
| | - Hyun Kyu Kim
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungcheongnam-do, Republic of Korea
| | - Byung Hoo Song
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungcheongnam-do, Republic of Korea
| | - Sun Choi
- Global AI Drug Discovery Center, College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea.
| | - Man Ryul Lee
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungcheongnam-do, Republic of Korea.
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Uchida Y, Kan H, Sakurai K, Oishi K, Matsukawa N. Contributions of blood-brain barrier imaging to neurovascular unit pathophysiology of Alzheimer's disease and related dementias. Front Aging Neurosci 2023; 15:1111448. [PMID: 36861122 PMCID: PMC9969807 DOI: 10.3389/fnagi.2023.1111448] [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: 11/29/2022] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
The blood-brain barrier (BBB) plays important roles in the maintenance of brain homeostasis. Its main role includes three kinds of functions: (1) to protect the central nervous system from blood-borne toxins and pathogens; (2) to regulate the exchange of substances between the brain parenchyma and capillaries; and (3) to clear metabolic waste and other neurotoxic compounds from the central nervous system into meningeal lymphatics and systemic circulation. Physiologically, the BBB belongs to the glymphatic system and the intramural periarterial drainage pathway, both of which are involved in clearing interstitial solutes such as β-amyloid proteins. Thus, the BBB is believed to contribute to preventing the onset and progression for Alzheimer's disease. Measurements of BBB function are essential toward a better understanding of Alzheimer's pathophysiology to establish novel imaging biomarkers and open new avenues of interventions for Alzheimer's disease and related dementias. The visualization techniques for capillary, cerebrospinal, and interstitial fluid dynamics around the neurovascular unit in living human brains have been enthusiastically developed. The purpose of this review is to summarize recent BBB imaging developments using advanced magnetic resonance imaging technologies in relation to Alzheimer's disease and related dementias. First, we give an overview of the relationship between Alzheimer's pathophysiology and BBB dysfunction. Second, we provide a brief description about the principles of non-contrast agent-based and contrast agent-based BBB imaging methodologies. Third, we summarize previous studies that have reported the findings of each BBB imaging method in individuals with the Alzheimer's disease continuum. Fourth, we introduce a wide range of Alzheimer's pathophysiology in relation to BBB imaging technologies to advance our understanding of the fluid dynamics around the BBB in both clinical and preclinical settings. Finally, we discuss the challenges of BBB imaging techniques and suggest future directions toward clinically useful imaging biomarkers for Alzheimer's disease and related dementias.
Collapse
Affiliation(s)
- Yuto Uchida
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States,*Correspondence: Yuto Uchida, ; Noriyuki Matsukawa,
| | - Hirohito Kan
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keita Sakurai
- Department of Radiology, National Center for Geriatrics and Gerontology, Ōbu, Aichi, Japan
| | - Kenichi Oishi
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Noriyuki Matsukawa
- Department of Neurology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan,*Correspondence: Yuto Uchida, ; Noriyuki Matsukawa,
| |
Collapse
|
37
|
Tan BBJW, Chua SKK, Soh QY, Chan LL, Tan EK. Chimeric antigen receptor (CAR) T therapy and cognitive functions. J Neurol Sci 2023; 444:120495. [PMID: 36446259 DOI: 10.1016/j.jns.2022.120495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 11/17/2022]
Affiliation(s)
| | - Shaun Kai Kiat Chua
- National Neuroscience Institute, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Qian Ying Soh
- National Neuroscience Institute, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Ling-Ling Chan
- Duke-NUS Medical School, Singapore, Singapore; Singapore General Hospital, Singapore
| | - Eng-King Tan
- National Neuroscience Institute, Singapore, Singapore; Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
38
|
Ma W, Yang YB, Xie TT, Xu Y, Liu N, Mo XN. Cerebral Small Vessel Disease: A Bibliometric Analysis. J Mol Neurosci 2022; 72:2345-2359. [PMID: 36203072 PMCID: PMC9726762 DOI: 10.1007/s12031-022-02070-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 09/18/2022] [Indexed: 12/14/2022]
Abstract
Cerebral small vessel disease is a common neurological disease, and its incidence is increasing year by year worldwide. In recent years, research on cerebral small vessel disease has gained more and more attention. Our research aims to visualize publications to identify the hotspots and frontiers of cerebral small vessel disease research, and to provide reference and guidance for further research. Publications related to cerebral small vessel disease were searched from the Web of Science Core Collection and screened according to inclusion criteria. CiteSpace 5.8.R3 was used to evaluate and visualize results, including generating web maps and analyzing annual publications, countries, institutions, bibliographic and co-cited references, and keywords; in this article, we use CiteSpace and VOSviewer for the 2012 Cerebral small vessel disease and bibliometric analysis from January 1, 2022 to April 30, 2022. A total of 3037 papers related to cerebral small vessel disease were retrieved, and the number of published papers showed a steady upward trend. Among them, Neuroimaging standards for research into small vessel disease and its contribution to ageing and neurodegeneration, the most symbolic references in the field of cerebral small vessel disease have been cited a total of 438 times. Stroke is the most active journal (227 articles) and USA publishes up to 800 articles. Harvard Med SchUniv Edinburgh (133 papers) and Charidimou (85 papers) are the institutions and authors who have made the most contributions in this field, respectively. Among the keywords, most of them are related to the pathogenesis of cerebral small vessel disease. After 2018, gut-brain axis and cortex are the keywords with the strongest number of cited outbreaks. There is increasing evidence that cerebral small vessel disease is a research frontier and may remain a research hotspot in the future.
Collapse
Affiliation(s)
- Wei Ma
- Guangxi University of Chinese Medicine, Nanning, China
| | - Yi-Bao Yang
- Ruikang Hospital Affiliated of Guangxi University of Chinese Medicine, Nanning, China
| | - Ting-Ting Xie
- The First Affiliated Hospital of Guangxi, University of Traditional Chinese Medicine, Nanning, China
| | - Yi Xu
- Maternal and Child Health Care Of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Na Liu
- Guangxi University of Chinese Medicine, Nanning, China
| | - Xue-Ni Mo
- Guangxi University of Chinese Medicine, Nanning, China.
| |
Collapse
|
39
|
Zhou L, Chen L, Ma L, Diao S, Qin Y, Fang Q, Li T. A new nomogram including total cerebral small vessel disease burden for individualized prediction of early-onset depression in patients with acute ischemic stroke. Front Aging Neurosci 2022; 14:922530. [PMID: 36238936 PMCID: PMC9552538 DOI: 10.3389/fnagi.2022.922530] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectivesThe present study was designed to evaluate the effects of total cerebral small vessel disease (CSVD) on early-onset depression after acute ischemic stroke (AIS), and to develop a new nomogram including total CSVD burden to predict early-onset post-stroke depression (PSD).MethodsWe continuously enrolled patients with AIS who were hospitalized at the First Affiliated Hospital of Soochow University between October 2017 and June 2019. All patients were assessed for depressive symptoms using the 17-item Hamilton Depression Scale (HAMD-17) at 14 ± 2 days after the onset of AIS. The diagnosis for depression was made according to the American Diagnostic and Statistical Manual of Mental Disorders Version 5 (DSM-5). The demographic and clinical data were collected including total CSVD burden. On the basis of a multivariate logistic model, the independent factors of early-onset PSD were identified and the predictive nomogram was generated. The performance of the nomogram was evaluated by Harrell's concordance index (C-index) and calibration plot.ResultsA total of 346 patients were enrolled. When contrasted to a 0 score of total CSVD burden, the score ≥2 (moderate to severe total CSVD burden) was an independent risk factor for early-onset PSD. Besides, gender, cognitive impairments, baseline Barthel Index (BI), and plasma fibrinogen were independently associated with early-onset PSD. The nomogram based on all these five independent risk factors was developed and validated with an Area Under Curve (AUC) of 0.780. In addition, the calibration plot revealed an adequate fit of the nomogram in predicting the risk of early-onset depression in patients with AIS.ConclusionsOur study found the total CSVD burden score of 2–4 points was an independent risk factor of early-onset PSD. The proposed nomogram based on total CSVD burden, gender, cognitive impairments, baseline BI, and plasma fibrinogen concentration gave rise to a more accurate and more comprehensive prediction for early-onset PSD.
Collapse
Affiliation(s)
- Lihua Zhou
- Department of Neurology, The People's Hospital of Suzhou New District, Suzhou, China
| | - Licong Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Linqing Ma
- Department of Neurology, The People's Hospital of Suzhou New District, Suzhou, China
| | - Shanshan Diao
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yiren Qin
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Qi Fang
| | - Tan Li
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Tan Li
| |
Collapse
|
40
|
|