1
|
Si J, Yu K, Hao J, Wang J, Zhang L. The therapeutic effects and mechanisms of glucagon-like peptide-1 receptor agonists in neurocognitive disorders. Ther Adv Neurol Disord 2025; 18:17562864251332035. [PMID: 40291753 PMCID: PMC12033604 DOI: 10.1177/17562864251332035] [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: 08/09/2024] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
Chronic cerebral hypoperfusion (CCH) represents a key pathogenic contributor to neurocognitive disorders. It can lead to multifaceted pathological alterations including neuroinflammation, neuronal apoptosis, blood-brain barrier disruption, synaptic plasticity deficits, and mitochondrial dysfunction. The glucagon-like peptide-1 receptor (GLP-1R), ubiquitously expressed across multiple organ systems, exerts neuroprotective effects by maintaining intracellular homeostasis and mitigating neuronal damage triggered by oxidative stress, inflammatory cascades, apoptotic signaling, and ischemic insults. Furthermore, GLP-1R activity is modulated by gut microbiota composition and short-chain fatty acid abundance, implicating the gut-brain axis in its regulatory influence on neurological function. This review systematically examines the pathophysiological mechanisms underlying CCH and highlights the therapeutic potential of GLP-1R activation. Specifically, GLP-1R-targeted interventions attenuate hypoperfusion-induced damage through pleiotropic pathways and gut-brain crosstalk, thereby offering novel perspectives for advancing both fundamental research and clinical management of neurocognitive disorders.
Collapse
Affiliation(s)
- Junchen Si
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Kai Yu
- Department of Burn and Plastic Surgery, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Jiheng Hao
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Jiyue Wang
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng People’s Hospital, No. 45 Huashan Road, Liaocheng, Shandong 252000, China
| |
Collapse
|
2
|
Le S, Xu F, Luo Z, Shi W, Lu S, Zhang Z, Guo Z, Xu W, Yang M, Li T, Li X, Liang K, Zhu L. Integrated analysis of chromatin and transcriptomic profiling of the striatum after cerebral hypoperfusion in mice. BMC Genomics 2025; 26:71. [PMID: 39856551 PMCID: PMC11762485 DOI: 10.1186/s12864-025-11256-y] [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: 09/21/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Vascular cognitive impairment (VCI) is a significant contributor to dementia, yet the precise mechanisms underlying the cognitive decline associated with chronic cerebral hypoperfusion (CCH) remain unclear. This study investigated the molecular and epigenetic changes in the striatum, a brain region critical for motor function and cognition, following chronic hypoperfusion using a bilateral common carotid artery stenosis (BCAS) model in mice. METHODS RNA-seq was utilized to identify differentially expressed genes (DEGs) associated with hypoperfusion. In parallel, ATAC-seq was used to assess changes in chromatin accessibility within the striatum, providing insight into the epigenome and potential regulatory mechanisms. The integration of these datasets allowed us to correlate chromatin accessibility with transcriptional activity and to identify key transcription factors driving the observed gene expression changes. RESULTS Analysis of striatum-specific transcriptome revealed significant upregulation of immune response genes, particularly type II interferon signaling, and downregulation of neural activation pathways. Analysis of striatum-specific epigenome showed increased chromatin accessibility at promoters of immune-related genes. Integrated analysis highlighted PU.1 as a key transcription factor in upregulated pathways, while neural pathways lacked epigenetic regulation, revealing distinct molecular responses in the striatum following chronic hypoperfusion. CONCLUSIONS Our findings indicate that upregulated pathways in the striatum following BCAS-induced CCH are driven by epigenetic changes, while downregulated pathways occur independently of these modifications. Additionally, PU.1 plays a critical role in mediating immune responses, offering a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Shijia Le
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Fengyiyang Xu
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhi Luo
- Department of Surgery, Shanghai Deji Hospital, Qingdao University, Shanghai, 200331, China
| | - Weihao Shi
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shuangshuang Lu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Zengyu Zhang
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Zimin Guo
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Wenshi Xu
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Mingqi Yang
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Tianyi Li
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xu Li
- Department of Vascular Surgery, Zhongshan Hospital, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Fudan University, Shanghai, 200032, China.
| | - Kun Liang
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Lei Zhu
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
3
|
Zhang Y, Wu L, He K, Cheng Y, Li L, Han D. Electroacupuncture alleviated chronic cerebral hypoperfusion damages via targeting aquaporin 4 to prevent amyloid-beta accumulation. Neuroreport 2025; 36:11-21. [PMID: 39526677 DOI: 10.1097/wnr.0000000000002114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The present study aimed to investigate the therapeutic effects of electroacupuncture (EA) on chronic cerebral hypoperfusion (CCH). We first applied the Morris water maze approach to determine the effects of EA and TGN-020 [an inhibitor of aquaporin 4 (AQP4)] on the learning and memory ability of CCH rats. The hematoxylin and eosin, and Nissl staining were further used to investigate the effects of EA and TGN-020 on the neuropathological changes of the dentate gyrus. Next, the ELISA kits were adopted to determine the effects of EA and TGN-020 on the content of amyloid-beta (Aβ) in the cerebrospinal fluid of CCH rats. Finally, we respectively employed technologies of immunohistochemical staining, quantitative real-time PCR, and Western blot to further explore the effects of EA and TGN-020 on the mRNA expression level of amyloid precursor protein (APP) and AQP4 as well as the protein expression level of Aβ1-42 and AQP4 in the dentate gyrus of CCH rats. Our results indicated that EA not only enhanced the learning and memory abilities of CCH rats but also improved the neuropathological damages of CCH rats by upregulating the mRNA and protein expression level of AQP4 to reduce the accumulation of Aβ, especially for the reduction of the mRNA expression level of APP and the protein expression level of Aβ1-42, but TGN-020 effectively reversed the therapeutic effects mentioned above of EA. In summary, we proved that EA, as the activator of AQP4, prevents the accumulation of Aβ during the treatment of CCH.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Acupuncture and Moxibustion, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | | | | | | | | | | |
Collapse
|
4
|
He Z, Sun J. The role of the neurovascular unit in vascular cognitive impairment: Current evidence and future perspectives. Neurobiol Dis 2025; 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] [MESH Headings] [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
|
5
|
AlRawili N, Al‐Kuraishy HM, Al‐Gareeb AI, Abdel‐Fattah MM, Al‐Harchan NA, Alruwaili M, Papadakis M, Alexiou A, Batiha GE. Trajectory of Cardiogenic Dementia: A New Perspective. J Cell Mol Med 2025; 29:e70345. [PMID: 39828641 PMCID: PMC11742966 DOI: 10.1111/jcmm.70345] [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: 07/16/2024] [Revised: 09/27/2024] [Accepted: 12/25/2024] [Indexed: 01/22/2025] Open
Abstract
The functions of the heart and brain are closely linked and essential to support human life by the heart-brain axis, which is a complex interconnection between the heart and brain. Also, cardiac function and cerebral blood flow regulate the brain's metabolism and function. Therefore, deterioration of cardiac function may affect cognitive function and may increase the risk of dementia. Cardiogenic dementia is defined as a cognitive deterioration due to heart diseases such as heart failure, myocardial infarction, and atrial fibrillation. The prevalence of cognitive impairment in patients with heart failure was 29%. In addition, coronary artery disease (CAD) is also associated with the development of cognitive impairment. CAD and reduction of myocardial contractility reduced cerebral blood flow and increased the risk of dementia in CAD patients. Furthermore, myocardial infarction and subsequent systemic haemodynamic instability promote the development and progression of cardiogenic dementia. These findings indicated that many cardiac diseases are implicated in the development and progression of cognitive impairment. Nevertheless, the underlying mechanism for the development of cardiogenic dementia was not fully elucidated. Consequently, this review aims to discuss the potential mechanisms involved in the pathogenesis of cardiogenic dementia.
Collapse
Affiliation(s)
- Nawaf AlRawili
- Department of Internal Medicine, College of MedicineNorthern Border UniversityArarSaudi Arabia
| | - Hayder M. Al‐Kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical PharmacologyJabir ibn Hayyan Medical UniversityKufaIraq
| | - Maha M. Abdel‐Fattah
- Department of Pharmacology and Toxicology, Faculty of PharmacyBeni‐Suef UniversityBeni‐SuefEgypt
| | - Nasser A. Al‐Harchan
- Department of Clinical Pharmacology, College of DentistryAl‐Rasheed UniversityBaghdadIraq
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of MedicineJouf UniversitySaudi Arabia
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐Herdecke, University of Witten‐ HerdeckeWuppertalGermany
| | - Athanasios Alexiou
- University Centre for Research & DevelopmentChandigarh UniversityMohaliIndia
- Department of Science and EngineeringNovel Global Community Educational FoundationNew South WalesAustralia
- Department of Research & DevelopmentAthensGreece
| | - Gaber El‐Saber Batiha
- Department of Research & DevelopmentAFNP MedWienAustria
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourAlBeheiraEgypt
| |
Collapse
|
6
|
Li L, Yuan R, Wu M, Yin X, Zhang M, Chen Z. Progress in the regulatory mechanism of mitophagy in chronic cerebral ischemic neuronal injury. Exp Neurol 2025; 383:115003. [PMID: 39419436 DOI: 10.1016/j.expneurol.2024.115003] [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: 08/17/2024] [Revised: 10/09/2024] [Accepted: 10/13/2024] [Indexed: 10/19/2024]
Abstract
Chronic cerebral ischemia (CCI) is a common clinical syndrome that can impact various cerebrovascular diseases. Its pathological mechanism of injury involves energy imbalance, oxidative stress, inflammatory response, and many other processes. Neuronal damage occurs in a complex and multifaceted manner. This article provides a detailed discussion of the activation and inhibition mechanisms of mitophagy under cerebral ischemia and considers the advantages and disadvantages of mitophagy in the recovery process of ischemic brain injury. Finally, we address the future direction of research on neuronal injury and the regulatory mechanisms of mitophagy in chronic cerebral ischemia. Future studies should focus on drug intervention at specific regulatory points and the cross-regulation of related signaling pathways to comprehensively deepen understanding of the mechanisms of neuronal injury in chronic cerebral ischemia. Promising interventions could potentially improve the treatment and outcomes of chronic cerebral ischemia.
Collapse
Affiliation(s)
- Lihong Li
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Rui Yuan
- Department of Otolaryngology, Head and Neck Surgery, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Moxin Wu
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Xiaoping Yin
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Manqing Zhang
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang 332005, Jiangxi, China.
| | - Zhiying Chen
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang 332005, Jiangxi, China.
| |
Collapse
|
7
|
Cheng YW, Yang LY, Chen YT, Chou SC, Chen KW, Chen YH, Deng CR, Chen IC, Chou WJ, Chang CC, Chen YR, Hwa HL, Wang KC, Kuo MF. Endothelial progenitor cell-derived conditioned medium mitigates chronic cerebral ischemic injury through macrophage migration inhibitory factor-activated AKT pathway. Stem Cell Res Ther 2024; 15:428. [PMID: 39543689 PMCID: PMC11566597 DOI: 10.1186/s13287-024-04015-5] [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: 05/25/2024] [Accepted: 10/26/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Chronic cerebral ischemia (CCI) is a significant health issue characterized by hypoperfusion due to damage or occlusion of the cerebral or carotid arteries. CCI may lead to progressive cognitive impairment that is considered as a prelude to neurodegenerative diseases, including dementia and Alzheimer's disease (AD). Endothelial progenitor cells (EPCs) have been implicated in vascular repair in ischemic cerebrovascular diseases, primarily by differentiating into endothelial cells (ECs) or through paracrine effects. However, the clinical transplantation of stem cell therapies remains limited. In this study, we investigated the effects of EPC-derived conditioned medium (EPC-CM) on the impaired vasculature and neurological function in a rodent model of CCI and the mechanism involved. METHODS EPC-CM was analyzed by cytokine array to identify key factors involved in angiogenesis and cellular senescence. The effects and mechanism of the candidate factors in the EPC-CM were validated in vitro using oxygen-glucose deprivation (OGD)-injured ECs and EPCs. The therapeutic effects of EPC-CM and the identified key factor were further examined in a rat model of CCI, which was induced by bilateral internal carotid artery ligation (BICAL). EPC-CM was administered via intracisternal injection one week post BICAL. The cerebral microvasculature and neurobehavior of the rats were examined three weeks after BICAL. RESULTS Macrophage migration inhibitory factor (MIF) was identified as a key factor in the EPC-CM. Recombinant MIF protein promoted angiogenesis and prevented senescence in the injured EPCs and ECs. The effect was similar to that of the EPC-CM. These therapeutic effects were diminished when the EPC-CM was co-treated with MIF-specific antibody (Ab). Additionally, the vascular, motor, and cognitive improvements observed in the BICAL rats treated with EPC-CM were abolished by co-treated with MIF Ab. Furthermore, we found MIF promoted angiogenesis and anti-senescence via activating the AKT pathway. Inhibition of the AKT pathway diminished the protective effects of MIF in the in vitro study. CONCLUSIONS We demonstrated that EPC-CM protected the brain from chronic ischemic injury and promoted functional recovery through MIF-mediated AKT pathway. These findings suggest EPC-CM holds potential as a novel cell-free therapeutic approach for treating CCI through the actions of MIF.
Collapse
Affiliation(s)
- Ya-Wen Cheng
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
| | - Ling-Yu Yang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
| | - Yi-Tzu Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sheng-Che Chou
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kuo-Wei Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yi-Hsing Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chuan-Rou Deng
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
| | - I-Chin Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
| | - Wan-Ju Chou
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chen-Chih Chang
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yong-Ren Chen
- Non-Invasive Cancer Therapy Research Institute, Taipei, Taiwan
- Adjunct Visiting Staff, Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsiao-Lin Hwa
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuo-Chuan Wang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan.
| | - Meng-Fai Kuo
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, No.7, Chung-Shan South Road, Taipei, 100, Taiwan.
| |
Collapse
|
8
|
Qin L, Tong F, Li S, Ren C. Beyond Pharmacology: The Biological Mechanisms of Remote Ischemic Conditioning in Cerebrovascular Disease. Biomolecules 2024; 14:1408. [PMID: 39595584 PMCID: PMC11592304 DOI: 10.3390/biom14111408] [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: 09/24/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Cerebrovascular diseases (CVDs), comprising predominantly ischemic stroke and chronic cerebral hypoperfusion (CCH), are a significant threat to global health, often leading to disability and mortality. Remote ischemic conditioning (RIC) has emerged as a promising, non-pharmacological strategy to combat CVDs by leveraging the body's innate defense mechanisms. This review delves into the neuroprotective mechanisms of RIC, categorizing its effects during the acute and chronic phases of stroke recovery. It also explores the synergistic potential of RIC when combined with other therapeutic strategies, such as pharmacological treatments and physical exercise. Additionally, this review discusses the pathways through which peripheral transmission can confer central neuroprotection. This review concludes by addressing the challenges regarding and future directions for RIC, emphasizing the need for standardized protocols, biomarker identification, and expanded clinical trials to fully realize its therapeutic potential.
Collapse
Affiliation(s)
| | | | | | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; (L.Q.); (F.T.); (S.L.)
| |
Collapse
|
9
|
Yang M, Yang L, Zhang Q, Xu L, Yang B, Li Y, Cheng X, Zhang F, Liu M, Yu N. Deep learning-based magnetic resonance imaging analysis for chronic cerebral hypoperfusion risk. Med Phys 2024; 51:5270-5282. [PMID: 38820428 DOI: 10.1002/mp.17237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/06/2024] [Accepted: 05/19/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) is a frequently encountered clinical condition that poses a diagnostic challenge due to its nonspecific symptoms. PURPOSE To enhance the diagnosis of CCH and non-CCH through Magnetic Resonance Imaging (MRI), offering support in clinical decision-making and recommendations to ultimately elevate diagnostic accuracy and optimize patient treatment outcomes. METHODS In the retrospective research, we collected 204 routine brain magnetic resonance imaging (MRI) from March 1 to September 10 2022, as training and testing cohorts. And a validation cohort with 108 samples was collected from November 14 2022 to August 4 2023. MRI sequences were processed to obtain T1-weighted (T1WI) and T2-weighted (T2WI) sequence images for each patient. We propose CCH-Network (CCHNet), an end-to-end deep learning model, integrating convolution and Transformer modules to capture local and global structural information. Our novel adversarial training method improves feature knowledge capture, enhancing both generalization ability and efficiency in predicting CCH risk. We assessed the classification performance of the proposed model CCHNet by comparing it with existing state-of-the-art deep learning algorithms, including ResNet34, DenseNet121, VGG16, Convnext, ViT, Coat, and TransFG. To better validate model performance, we compared the results of the proposed model with eight neurologists to evaluate their consistency. RESULTS CCHNet achieved an AUC of 91.6% (95% CI: 86.8-99.1), with an accuracy (ACC) of 85.0% (95% CI: 75.6-95.2). It demonstrated a sensitivity (SE) of 80.0% (95% CI: 71.6-95.6) and a specificity (SP) of 90.0% (95% CI: 82.3-97.8) in the testing cohort. In the validation cohort, the model demonstrated an AUC of 86.0% (95% CI: 80.3-93.0), an ACC of 84.2% (95% CI: 70.2-93.6), a SE of 83.3% (95% CI: 68.3-95.5), and a SP of 84.7% (95% CI: 70.3-96.8). CONCLUSIONS The model improved the diagnostic performance of MRI with high SE and SP, providing a promising method for the diagnosis of CCH.
Collapse
Affiliation(s)
- Meiyi Yang
- Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, China
- College of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Lili Yang
- Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Qi Zhang
- Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Lifeng Xu
- Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, China
| | - Bo Yang
- Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yingjie Li
- Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xudong Cheng
- Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Feng Zhang
- Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ming Liu
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang, China
| | - Nengwei Yu
- Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Whitehead BJ, Corbin D, Alexander ML, Bumgarner J, Zhang N, Karelina K, Weil ZM. Cerebral hypoperfusion exacerbates traumatic brain injury in male but not female mice. Eur J Neurosci 2024; 60:4346-4361. [PMID: 38858126 PMCID: PMC11533132 DOI: 10.1111/ejn.16439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/07/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
Mild-moderate traumatic brain injuries (TBIs) are prevalent, and while many individuals recover, there is evidence that a significant number experience long-term health impacts, including increased vulnerability to neurodegenerative diseases. These effects are influenced by other risk factors, such as cardiovascular disease. Our study tested the hypothesis that a pre-injury reduction in cerebral blood flow (CBF), mimicking cardiovascular disease, worsens TBI recovery. We induced bilateral carotid artery stenosis (BCAS) and a mild-moderate closed-head TBI in male and female mice, either alone or in combination, and analyzed CBF, spatial learning, memory, axonal damage, and gene expression. Findings showed that BCAS and TBI independently caused a ~10% decrease in CBF. Mice subjected to both BCAS and TBI experienced more significant CBF reductions, notably affecting spatial learning and memory, particularly in males. Additionally, male mice showed increased axonal damage with both BCAS and TBI compared to either condition alone. Females exhibited spatial memory deficits due to BCAS, but these were not worsened by subsequent TBI. Gene expression analysis in male mice highlighted that TBI and BCAS individually altered neuronal and glial profiles. However, the combination of BCAS and TBI resulted in markedly different transcriptional patterns. Our results suggest that mild cerebrovascular impairments, serving as a stand-in for preexisting cardiovascular conditions, can significantly worsen TBI outcomes in males. This highlights the potential for mild comorbidities to modify TBI outcomes and increase the risk of secondary diseases.
Collapse
Affiliation(s)
- Bailey J. Whitehead
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Deborah Corbin
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Megan L. Alexander
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Jacob Bumgarner
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Ning Zhang
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Kate Karelina
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Zachary M. Weil
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| |
Collapse
|
11
|
Zhang Q, Zhang X, Zhang J, Yu N. Peripheral blood diagnostic markers of chronic cerebral hypoperfusion. Am J Transl Res 2024; 16:2973-2981. [PMID: 39114695 PMCID: PMC11301487 DOI: 10.62347/orsn1228] [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: 02/20/2024] [Accepted: 06/03/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVE To investigate the efficacy of ischemia-modified albumin (IMA), lipoprotein-associated phospholipase A2 (Lp-PLA2), brain-derived neurotrophic factor (BDNF), and visinin-like protein-1 (VILIP-1) in diagnosing chronic cerebral hypoperfusion (CCH). METHODS This retrospective study included 84 patients with suspected chronic cerebral ischemia admitted to Sichuan Provincial People's Hospital between February 2021 and April 2022. Arterial spin labeling (ASL) imaging and biological examinations were performed. According to the ASL perfusion imaging patterns, the patients were divided into a CCH group (n = 55) and a non-CCH group (n = 29). Serum markers of the two groups were compared, and correlation analysis was conducted between ischemic marker levels and cerebral blood flow (CBF) in the ischemic region, as measured by ASL. Receiver operating characteristic (ROC) curve analysis was used to evaluate the efficacy of each marker for diagnosing chronic cerebral ischemia. The Delong test was used to compare AUC size between groups. RESULTS Compared to the non-CCH group, the CCH group exhibited higher IMA levels and lower BDNF concentrations (P < 0.05). However, VILIP-1 and Lp-PLA2 concentrations were not significantly different between the two groups (P > 0.05). Moreover, IMA and BDNF levels were not correlated with CBF in the hypoperfused area. ROC curve analysis demonstrated that the cut-off values of 24.2915 U/mL and 6.714 ng/L for IMA and BDNF achieved a sensitivity of 83.6% and 41.8% and a specificity of 62.1% and 93.1%, respectively. Lastly, the areas under the curve for IMA and BDNF were 0.738 (95% confidence interval [CI], 0.627-0.848) and 0.631 (95% CI, 0.512-0.751), respectively. CONCLUSION IMA and BDNF may have clinical value in the diagnosis of CCH.
Collapse
Affiliation(s)
- Qiong Zhang
- Clinical School of Medicine, Southwest Medical UniversityLuzhou 646000, Sichuan, China
- Jiangyou City People’s HospitalJiangyou 621700, Sichuan, China
| | - Xin Zhang
- Department of Internal Medicine-Neurology, Chengdu 363 Hospital Affiliated to Southwest Medical UniversityChengdu 610041, Sichuan, China
| | - Jing Zhang
- Chengdu Sixth People’s HospitalChengdu 610051, Sichuan, China
- Clinical Medical School, University of Electronic Science and Technology of ChinaChengdu 610054, Sichuan, China
| | - Nengwei Yu
- Clinical School of Medicine, Southwest Medical UniversityLuzhou 646000, Sichuan, China
- Clinical Medical School, University of Electronic Science and Technology of ChinaChengdu 610054, Sichuan, China
- Department of Neurology, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s HospitalChengdu 610072, Sichuan, China
| |
Collapse
|
12
|
Jearjaroen P, Thangwong P, Tocharus C, Lungkaphin A, Chaichompoo W, Srijun J, Suksamrarn A, Tocharus J. Hexahydrocurcumin Attenuates Neuronal Injury and Modulates Synaptic Plasticity in Chronic Cerebral Hypoperfusion in Rats. Mol Neurobiol 2024; 61:4304-4317. [PMID: 38087168 DOI: 10.1007/s12035-023-03821-x] [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: 05/19/2023] [Accepted: 11/20/2023] [Indexed: 07/11/2024]
Abstract
Dementia is the most common age-related problem due predominantly to Alzheimer's disease (AD) and vascular dementia (VaD). It has been shown that these contributors are associated with a high amount of oxidative stress that leads to changes in neurological function and cognitive impairment. The aim of study was to explore the mechanism by which hexahydrocurcumin (HHC) attenuates oxidative stress, amyloidogenesis, phosphorylated Tau (pTau) expression, neuron synaptic function, and cognitive impairment and also the potential mechanisms involved in induced permanent occlusion of bilateral common carotid arteries occlusion (BCCAO) or 2-vessel occlusion (2VO) in rats. After surgery, rats were treated with HHC (40 mg/kg) or piracetam (600 mg/kg) by oral gavage daily for 4 weeks. The results showed that HHC or piracetam attenuated oxidative stress by promoting nuclear factor erythroid 2-related factor 2 (Nrf2) activity, and alleviated expression of synaptic proteins (pre- and post-synaptic proteins) mediated by the Wingless/Integrated (Wnt)/β-catenin signaling pathway. Moreover, HHC or piracetam also improved synaptic plasticity via the brain-derived neurotrophic factor (BDNF)/Tyrosine receptor kinase B (TrkB)/cAMP responsive element binding protein (CREB) signaling pathway. In addition, HHC reduced amyloid beta (Aβ) production and pTau expression and improved memory impairment as evidenced by the Morris water maze. In conclusion, HHC exerted remarkable improvement in cognitive function in the 2VO rats possibly via the attenuation of oxidative stress, improvement in synaptic function, attenuation of amyloidogenesis, pTau, and neuronal injury, thereby improving cognitive performance.
Collapse
Affiliation(s)
- Pranglada Jearjaroen
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Phakkawat Thangwong
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Anusorn Lungkaphin
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Waraluck Chaichompoo
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Jaranwit Srijun
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Apichart Suksamrarn
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Functional Food Research Center for Well-being, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
13
|
Zhu W, Shen Y, Zhao H, Tang Y, Wang X, Li S. Predicting postoperative delirium after percutaneous transluminal angioplasty and stenting in patients with intracranial atherosclerotic stenosis. Neurol Res 2024:1-9. [PMID: 38909321 DOI: 10.1080/01616412.2024.2370730] [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: 09/26/2023] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
OBJECTIVE Known as a major surgical complication, postoperative delirium (POD) has not been well studied in patients with intracranial atherosclerotic stenosis (ICAS). This study aimed to investigate the correlation between perioperative clinical characteristics and the occurrence of POD. METHODS Patients' demographic characteristics and perioperative testing data were collected. Binary logistic regression was conducted for assessing related risk factors. A nomogram was developed to predict the occurrence of POD after percutaneous transluminal angioplasty and stenting (PTAS) in patients with ICAS. RESULTS The occurrence of POD in this study was 30.67%. Among all the clinical and laboratory characteristics in patients, age (OR = 1.234, 95%CI = 1.004-1.517, p = 0.046), gender (OR = 5.676, 95%CI = 1.028-31.334, p = 0.046), preoperative MMSE scores (OR = 2.298, 95%CI = 1.005-5.259, p = 0.049), the degree of stenosis (OR = 6.294, 95%CI = 1.043-37.974, p = 0.045), operating time (OR = 1.088, 95%CI = 1.023-1.157, p = 0.006), and HbA1c levels (OR = 2.226, 95%CI = 1.199-4.130, p = 0.011) were the independent risk factors. CONCLUSION Male patients with advanced-age, lower preoperative MMSE scores, severe stenosis, longer operating time, and higher HbA1c levels are closely related to POD after PTAS. Fully perioperative assessments may play an important role in predicting the occurrence of POD.
Collapse
Affiliation(s)
- Wanchun Zhu
- Department of Neurosurgery, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yiman Shen
- Department of Neurosurgery, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hua Zhao
- Department of Neurosurgery, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yinda Tang
- Department of Neurosurgery, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuhui Wang
- Department of Neurosurgery, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shiting Li
- Department of Neurosurgery, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
He Y, He T, Li H, Chen W, Zhong B, Wu Y, Chen R, Hu Y, Ma H, Wu B, Hu W, Han Z. Deciphering mitochondrial dysfunction: Pathophysiological mechanisms in vascular cognitive impairment. Biomed Pharmacother 2024; 174:116428. [PMID: 38599056 DOI: 10.1016/j.biopha.2024.116428] [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: 12/20/2023] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 04/12/2024] Open
Abstract
Vascular cognitive impairment (VCI) encompasses a range of cognitive deficits arising from vascular pathology. The pathophysiological mechanisms underlying VCI remain incompletely understood; however, chronic cerebral hypoperfusion (CCH) is widely acknowledged as a principal pathological contributor. Mitochondria, crucial for cellular energy production and intracellular signaling, can lead to numerous neurological impairments when dysfunctional. Recent evidence indicates that mitochondrial dysfunction-marked by oxidative stress, disturbed calcium homeostasis, compromised mitophagy, and anomalies in mitochondrial dynamics-plays a pivotal role in VCI pathogenesis. This review offers a detailed examination of the latest insights into mitochondrial dysfunction within the VCI context, focusing on both the origins and consequences of compromised mitochondrial health. It aims to lay a robust scientific groundwork for guiding the development and refinement of mitochondrial-targeted interventions for VCI.
Collapse
Affiliation(s)
- Yuyao He
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Tiantian He
- Sichuan Academy of Chinese Medicine Sciences, China
| | - Hongpei Li
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Wei Chen
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Biying Zhong
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yue Wu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Runming Chen
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yuli Hu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Huaping Ma
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Bin Wu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Wenyue Hu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China.
| | - Zhenyun Han
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China.
| |
Collapse
|
15
|
Wang DP, Kang K, Hai J, Lv QL, Wu ZB. Alleviating CB2-Dependent ER Stress and Mitochondrial Dysfunction Improves Chronic Cerebral Hypoperfusion-Induced Cognitive Impairment. J Neuroimmune Pharmacol 2024; 19:1. [PMID: 38214766 PMCID: PMC10786746 DOI: 10.1007/s11481-024-10098-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 12/06/2023] [Indexed: 01/13/2024]
Abstract
Augmentation of endoplasmic reticulum (ER) stress may trigger excessive oxidative stress, which induces mitochondrial dysfunction. The fatty acid amide hydrolase inhibitor, URB597, shows anti-oxidation characteristics in multiple neurological disorders. The present study aimed to determine whether inhibition of ER stress was involved in the protective effects of URB597 against chronic cerebral hypoperfusion (CCH)-induced cognitive impairment. Hippocampal HT-22 cells were exposed to oxygen-glucose deprivation. The cell viability, apoptosis, ER stress, mitochondrial ATP, and oxidative stress levels were assessed following treatment with URB597, benzenebutyric acid (4-PBA), and thapsigargin (TG). Furthermore, the effects of URB597 on ER stress and related pathways were investigated in the CCH animal model, including Morris water maze testing of cognition, western blotting analysis of ER stress signaling, and transmission electron microscopy of mitochondrial and ER ultrastructure changes. The results suggested that cerebral ischemia caused ER stress with upregulation of ER stress signaling-related proteins, mitochondrial dysfunction, neuronal apoptosis, ultrastructural injuries of mitochondria-associated ER membranes, and cognitive decline. Co-immunoprecipitation experiments confirmed the interaction between CB2 and β-Arrestin1. Inhibiting ER stress by URB597 improved these changes by activating CB2/β-Arrestin1 signaling, which was reversed by the CB2 antagonist, AM630. Together, the results identified a novel mechanism of URB597, involving CCH-induced cognitive impairment alleviation of CB2-dependent ER stress and mitochondrial dysfunction. Furthermore, this study identified CB2 as a potential target for therapy of ischemic cerebrovascular diseases.
Collapse
Affiliation(s)
- Da Peng Wang
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Huangpu District, Shanghai, 200025, China
- Department of Neurosurgery, Tong Ji Hospital, School of Medicine, Tong Ji University, Shanghai, 200065, China
| | - Kai Kang
- School of Public Health, Fudan University, Shanghai, 200032, China
- Department of Research and Surveillance Evaluation, Shanghai Municipal Center for Health Promotion, Shanghai, 200040, China
| | - Jian Hai
- Department of Neurosurgery, Tong Ji Hospital, School of Medicine, Tong Ji University, Shanghai, 200065, China
| | - Qiao Li Lv
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Jiangxi, 330029, China.
| | - Zhe Bao Wu
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Huangpu District, Shanghai, 200025, China.
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
16
|
Niu Z, Yu M, Xu P, Liu R, Li S, Wu C, Huang B, Ye X, Hu J, Xu Y, Lu S. Effect of 40 Hz light flicker on cognitive impairment and transcriptome of hippocampus in right unilateral common carotid artery occlusion mice. Sci Rep 2023; 13:21361. [PMID: 38049571 PMCID: PMC10695931 DOI: 10.1038/s41598-023-48897-6] [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: 10/14/2023] [Revised: 11/18/2023] [Accepted: 11/30/2023] [Indexed: 12/06/2023] Open
Abstract
Vascular cognitive impairment caused by chronic cerebral hypoperfusion (CCH) seriously affects the quality of life of elderly patients. However, there is no effective treatment to control this disease. This study investigated the potential neuroprotective effect of the 40 Hz light flicker in a mouse model of CCH. CCH was induced in male C57 mice by right unilateral common carotid artery occlusion (rUCCAO), leading to chronic brain injury. The mice underwent 40 Hz light flicker stimulation for 30 days after surgery. The results showed that 40 Hz light flicker treatment ameliorated memory deficits after rUCCAO and alleviated the damage to neurons in the frontal lobe and hippocampus. Light flicker administration at 40 Hz decreased IL-1β and TNF-α levels in the frontal lobe and hippocampus, but immunohistochemistry showed that it did not induce angiogenesis in mice with rUCCAO. Gene expression profiling revealed that the induction of genes was mainly enriched in inflammatory-related pathways. Our findings demonstrate that 40 Hz light flicker can suppress cognitive impairment caused by rUCCAO and that this effect may be involved in the attenuation of neuroinflammation.
Collapse
Affiliation(s)
- Zhaorui Niu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Minjie Yu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Peixia Xu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Renchuan Liu
- Zhejiang Xinyue Health Consulting Service Medical Institution, Hangzhou, 310003, China
| | - Shangda Li
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Congchong Wu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Bochao Huang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
- Zhejiang Xinyue Health Consulting Service Medical Institution, Hangzhou, 310003, China
| | - Xinyi Ye
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Jianbo Hu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China
- Brain Research Institute of Zhejiang University, Hangzhou, 310003, China
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, 310003, China
| | - Yi Xu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China.
- Brain Research Institute of Zhejiang University, Hangzhou, 310003, China.
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, 310003, China.
- Zhejiang Xinyue Health Consulting Service Medical Institution, Hangzhou, 310003, China.
| | - Shaojia Lu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China.
- Brain Research Institute of Zhejiang University, Hangzhou, 310003, China.
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, 310003, China.
| |
Collapse
|
17
|
Ann P, Chen M, Naidich T, Belani P, Nael K. Arterial spin labeling perfusion in acute Wernicke encephalopathy: a case series discussion. BJR Case Rep 2023; 9:20220137. [PMID: 37928701 PMCID: PMC10621587 DOI: 10.1259/bjrcr.20220137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/19/2023] Open
Abstract
Wernicke's encephalopathy (WE) is a life-threatening neurologic disorder resulting from thiamine (vitamin B1) deficiency that can be secondary to chronic alcohol abuse, gastrointestinal surgery, systemic infectious and non-infectious diseases, and chemotherapy. WE is classically characterized on MRI by reduced diffusion and T2 prolongation along the mammillothalamic tracts, periaqueductal gray and tectal plate. We present two patients with acute WE who had baseline arterial spin labeling (ASL) perfusion at the time of presentation, demonstrating increase in cerebral blood flow (CBF) within the classically involved brain regions and concurrent global cerebral cortical hypoperfusion. Both patients were successfully treated with intravenous thiamine infusion. Post-treatment MRI demonstrated improvement of reduced diffusion and normalization of CBF within the involved structures. Prior histopathological studies have documented prominent undulation and luminal dilatation of arteries and arterioles in acute WE lesions, likely explaining the increased perfusion shown by imaging. The root of this pathophysiologic process may trace back to thiamine's biochemical role in maintaining osmotic gradients and glucose metabolism, that if failed can lead to arterial hyper-perfusion. Our findings show that ASL-CBF can highlight the underlying pathophysiology in patients with acute WE by demonstrating increased CBF in involved central structures. This luxury perfusion may be a compensatory or protective mechanism by which increased metabolic demand is met in the acute setting and which, if treated timely, will show normalization of CBF on ASL imaging.
Collapse
Affiliation(s)
- Phoebe Ann
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, 757 Westwood Plaza, Los Angeles, California
| | - Mark Chen
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, 1176 5th Ave, New York, NY, United States
| | - Thomas Naidich
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, 1176 5th Ave, New York, NY, United States
| | - Puneet Belani
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, 1176 5th Ave, New York, NY, United States
| | - Kambiz Nael
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, 757 Westwood Plaza, Los Angeles, California
| |
Collapse
|
18
|
Kang K, Wang DP, Lv QL, Chen F. VEGF-A ameliorates ischemia hippocampal neural injury via regulating autophagy and Akt/CREB signaling in a rat model of chronic cerebral hypoperfusion. J Stroke Cerebrovasc Dis 2023; 32:107367. [PMID: 37734181 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023] Open
Abstract
OBJECTIVE Chronic cerebral hypoperfusion (CCH) can cause a series of pathophysiological processes, including neuronal autophagy and apoptosis. VEGF-A has been reported to affect angiogenesis and neurogenesis in many CNS diseases. However, its effects on neuronal autophagy and apoptosis, as well as the underlying mechanisms in CCH remain unclear. METHODS To address these issues, the CCH model was established by permanent bilateral common carotid artery occlusion (2VO). Rats were sacrificed at different stages of CCH. Hippocampal morphological and ultrastructural changes were detected using HE staining and electron microscopy. The immunoreactivities of microtubule-associated protein 1 light chain 3 (LC3) and phospho-cAMP response element binding protein (p-CREB) were examined by immunofluorescence staining. The neuronal apoptosis was detected via TUNEL staining. The levels of LC3-II, Beclin-1, Akt, p-Akt, CREB, p-CREB, Caspase-3, and Bad were accessed by Western blotting. Furthermore, mouse hippocampal HT22 neurons received the oxygen and glucose deprivation (OGD) treatment, VEGF-A treatment, and GSK690693 (an Akt inhibitor) treatment, respectively. RESULTS LC3-II protein started to increase at 3 days of CCH, peaked at 4 weeks of CCH, then decreased. CCH increased the levels of LC3-II, Caspase-3, and Bad, and decreased the levels of p-Akt, CREB, and p-CREB, which were reversed by VEGF-A treatment. VEGF-A also improved CCH-induced neuronal ultrastructural injuries and apoptosis in the hippocampus in vitro. In HT22, the anti-apoptosis and pro-phosphorylation of VEGF-A were reversed by GSK690693. CONCLUSION Present results provide a novel neuroprotective effect of VEGF-A in CCH that is related to the inhibition of neuronal autophagy and activation of the Akt/CREB signaling, suggesting a potential therapeutic strategy for ischemic brain damage.
Collapse
Affiliation(s)
- Kai Kang
- School of Public Health, Fudan University, Shanghai 200032, China; Department of Research and Surveillance Evaluation, Shanghai Municipal Center for Health Promotion, Shanghai 200040, China
| | - Da-Peng Wang
- Department of Neurosurgery, Tong Ji Hospital, Tong Ji University School of Medicine, Shanghai, China
| | - Qiao-Li Lv
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Jiangxi 330029, China.
| | - Feng Chen
- Department of Neurosurgery, Tong Ji Hospital, Tong Ji University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Schneider SC, Kaczmarz S, Göttler J, Kufer J, Zott B, Priller J, Kallmayer M, Zimmer C, Sorg C, Preibisch C. Stronger influence of systemic than local hemodynamic-vascular factors on resting-state BOLD functional connectivity. Neuroimage 2023; 281:120380. [PMID: 37741595 DOI: 10.1016/j.neuroimage.2023.120380] [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: 04/06/2023] [Revised: 08/28/2023] [Accepted: 09/13/2023] [Indexed: 09/25/2023] Open
Abstract
Correlated fluctuations in the blood oxygenation level dependent (BOLD) signal of resting-state functional MRI (i.e., BOLD-functional connectivity, BOLD-FC) reflect a spectrum of neuronal and non-neuronal processes. In particular, there are multiple hemodynamic-vascular influences on BOLD-FC on both systemic (e.g., perfusion delay) and local levels (e.g., neurovascular coupling). While the influence of individual factors has been studied extensively, combined and comparative studies of systemic and local hemodynamic-vascular factors on BOLD-FC are scarce, notably in humans. We employed a multi-modal MRI approach to investigate and compare distinct hemodynamic-vascular processes and their impact on homotopic BOLD-FC in healthy controls and patients with unilateral asymptomatic internal carotid artery stenosis (ICAS). Asymptomatic ICAS is a cerebrovascular disorder, in which neuronal functioning is largely preserved but hemodynamic-vascular processes are impaired, mostly on the side of stenosis. Investigated indicators for local hemodynamic-vascular processes comprise capillary transit time heterogeneity (CTH) and cerebral blood volume (CBV) from dynamic susceptibility contrast (DSC) MRI, and cerebral blood flow (CBF) from pseudo-continuous arterial spin labeling (pCASL). Indicators for systemic processes are time-to-peak (TTP) from DSC MRI and BOLD lags from functional MRI. For each of these parameters, their influence on BOLD-FC was estimated by a comprehensive linear mixed model. Equally across groups, we found that individual mean BOLD-FC, local (CTH, CBV, and CBF) and systemic (TTP and BOLD lag) hemodynamic-vascular factors together explain 40.7% of BOLD-FC variance, with 20% of BOLD-FC variance explained by hemodynamic-vascular factors, with an about two-times larger contribution of systemic versus local factors. We conclude that regional differences in blood supply, i.e., systemic perfusion delays, exert a stronger influence on BOLD-FC than impairments in local neurovascular coupling.
Collapse
Affiliation(s)
- Sebastian C Schneider
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Clinic for Psychiatry, Ismaningerstr. 22, 81675 Munich, Germany; Technical University of Munich, School of Medicine, Klinikum rechts der Isar, TUM Neuroimaging Center, Ismaningerstr. 22, 81675 Munich, Germany; Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Diagnostic and Interventional Neuroradiology, Ismaningerstr. 22, 81675 Munich, Munich, Germany.
| | - Stephan Kaczmarz
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, TUM Neuroimaging Center, Ismaningerstr. 22, 81675 Munich, Germany; Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Diagnostic and Interventional Neuroradiology, Ismaningerstr. 22, 81675 Munich, Munich, Germany; Philips GmbH Market DACH, Hamburg, Germany
| | - Jens Göttler
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, TUM Neuroimaging Center, Ismaningerstr. 22, 81675 Munich, Germany; Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Diagnostic and Interventional Neuroradiology, Ismaningerstr. 22, 81675 Munich, Munich, Germany
| | - Jan Kufer
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, TUM Neuroimaging Center, Ismaningerstr. 22, 81675 Munich, Germany; Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Diagnostic and Interventional Neuroradiology, Ismaningerstr. 22, 81675 Munich, Munich, Germany; Department of Radiology & Biomedical Imaging, Yale University, New Haven, CT, United States of America
| | - Benedikt Zott
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, TUM Neuroimaging Center, Ismaningerstr. 22, 81675 Munich, Germany; Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Diagnostic and Interventional Neuroradiology, Ismaningerstr. 22, 81675 Munich, Munich, Germany
| | - Josef Priller
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Clinic for Psychiatry, Ismaningerstr. 22, 81675 Munich, Germany
| | - Michael Kallmayer
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Clinic for vascular surgery, Ismaningerstr. 22, 81675 Munich, Munich, Germany
| | - Claus Zimmer
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Diagnostic and Interventional Neuroradiology, Ismaningerstr. 22, 81675 Munich, Munich, Germany
| | - Christian Sorg
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Clinic for Psychiatry, Ismaningerstr. 22, 81675 Munich, Germany; Technical University of Munich, School of Medicine, Klinikum rechts der Isar, TUM Neuroimaging Center, Ismaningerstr. 22, 81675 Munich, Germany; Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Diagnostic and Interventional Neuroradiology, Ismaningerstr. 22, 81675 Munich, Munich, Germany
| | - Christine Preibisch
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, TUM Neuroimaging Center, Ismaningerstr. 22, 81675 Munich, Germany; Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Diagnostic and Interventional Neuroradiology, Ismaningerstr. 22, 81675 Munich, Munich, Germany; Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Clinic for Neurology, Ismaningerstr. 22, 81675 Munich, Munich, Germany
| |
Collapse
|
20
|
Yu M, Zhang M, Fu P, Wu M, Yin X, Chen Z. Research progress of mitophagy in chronic cerebral ischemia. Front Aging Neurosci 2023; 15:1224633. [PMID: 37600521 PMCID: PMC10434995 DOI: 10.3389/fnagi.2023.1224633] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Chronic cerebral ischemia (CCI), a condition that can result in headaches, dizziness, cognitive decline, and stroke, is caused by a sustained decrease in cerebral blood flow. Statistics show that 70% of patients with CCI are aged > 80 years and approximately 30% are 45-50 years. The incidence of CCI tends to be lower, and treatment for CCI is urgent. Studies have confirmed that CCI can activate the corresponding mechanisms that lead to mitochondrial dysfunction, which, in turn, can induce mitophagy to maintain mitochondrial homeostasis. Simultaneously, mitochondrial dysfunction can aggravate the insufficient energy supply to cells and various diseases caused by CCI. Regulation of mitophagy has become a promising therapeutic target for the treatment of CCI. This article reviews the latest progress in the important role of mitophagy in CCI and discusses the induction pathways of mitophagy in CCI, including ATP synthesis disorder, oxidative stress injury, induction of reactive oxygen species, and Ca2+ homeostasis disorder, as well as the role of drugs in CCI by regulating mitophagy.
Collapse
Affiliation(s)
- Mayue Yu
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Manqing Zhang
- School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi, China
| | - Peijie Fu
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Moxin Wu
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Xiaoping Yin
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Zhiying Chen
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| |
Collapse
|
21
|
Day H, Yellman B, Hammer S, Rond C, Bell J, Abbaszadeh S, Stoddard G, Unutmaz D, Bateman L, Vernon SD. Cognitive impairment in post-acute sequelae of COVID-19 and short duration myalgic encephalomyelitis patients is mediated by orthostatic hemodynamic changes. Front Neurosci 2023; 17:1203514. [PMID: 37434760 PMCID: PMC10330752 DOI: 10.3389/fnins.2023.1203514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/19/2023] [Indexed: 07/13/2023] Open
Abstract
Introduction Cognitive impairment is experienced by people with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) and post-acute sequelae of COVID-19 (PASC). Patients report difficulty remembering, concentrating, and making decisions. Our objective was to determine whether orthostatic hemodynamic changes were causally linked to cognitive impairment in these diseases. Methods This prospective, observational cohort study enrolled PASC, ME/CFS, and healthy controls. All participants underwent clinical evaluation and assessment that included brief cognitive testing before and after an orthostatic challenge. Cognitive testing measured cognitive efficiency which is defined as the speed and accuracy of subject's total correct responses per minute. General linear mixed models were used to analyze hemodynamics and cognitive efficiency during the orthostatic challenge. Additionally, mediation analysis was used to determine if hemodynamic instability induced during the orthostatic challenge mediated the relationship between disease status and cognitive impairment. Results Of the 276 participants enrolled, 256 were included in this study (34 PASC, 71 < 4 year duration ME/CFS, 69 > 10 year ME/CFS duration, and 82 healthy controls). Compared to healthy controls, the disease cohorts had significantly lower cognitive efficiency scores immediately following the orthostatic challenge. Cognitive efficiency remained low for the >10 year ME/CFS 2 and 7 days after orthostatic challenge. Narrow pulse pressure less than 25% of systolic pressure occurred at 4 and 5 min into the orthostatic challenge for the PASC and ME/CFS cohorts, respectively. Abnormally narrow pulse pressure was associated with slowed information processing in PASC patients compared to healthy controls (-1.5, p = 0.04). Furthermore, increased heart rate during the orthostatic challenge was associated with a decreased procedural reaction time in PASC and < 4 year ME/CFS patients who were 40 to 65 years of age. Discussion For PASC patients, both their disease state and hemodynamic changes during orthostatic challenge were associated with slower reaction time and decreased response accuracy during cognitive testing. Reduced cognitive efficiency in <4 year ME/CFS patients was associated with higher heart rate in response to orthostatic stress. Hemodynamic changes did not correlate with cognitive impairment for >10 year ME/CFS patients, but cognitive impairment remained. These findings underscore the need for early diagnosis to mitigate direct hemodynamic and other physiological effects on symptoms of cognitive impairment.
Collapse
Affiliation(s)
- Heather Day
- School of Medicine, The University of Utah, Salt Lake City, UT, United States
| | | | - Sarah Hammer
- Bateman Horne Center, Salt Lake City, UT, United States
| | - Candace Rond
- Bateman Horne Center, Salt Lake City, UT, United States
| | - Jennifer Bell
- Bateman Horne Center, Salt Lake City, UT, United States
| | | | - Greg Stoddard
- School of Medicine, The University of Utah, Salt Lake City, UT, United States
| | - Derya Unutmaz
- Jackson Laboratory for Genomic Medicine, School of Medicine, University of Connecticut, Farmington, CT, United States
| | | | | |
Collapse
|
22
|
Rajeev V, Chai YL, Poh L, Selvaraji S, Fann DY, Jo DG, De Silva TM, Drummond GR, Sobey CG, Arumugam TV, Chen CP, Lai MKP. Chronic cerebral hypoperfusion: a critical feature in unravelling the etiology of vascular cognitive impairment. Acta Neuropathol Commun 2023; 11:93. [PMID: 37309012 PMCID: PMC10259064 DOI: 10.1186/s40478-023-01590-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/14/2023] Open
Abstract
Vascular cognitive impairment (VCI) describes a wide spectrum of cognitive deficits related to cerebrovascular diseases. Although the loss of blood flow to cortical regions critically involved in cognitive processes must feature as the main driver of VCI, the underlying mechanisms and interactions with related disease processes remain to be fully elucidated. Recent clinical studies of cerebral blood flow measurements have supported the role of chronic cerebral hypoperfusion (CCH) as a major driver of the vascular pathology and clinical manifestations of VCI. Here we review the pathophysiological mechanisms as well as neuropathological changes of CCH. Potential interventional strategies for VCI are also reviewed. A deeper understanding of how CCH can lead to accumulation of VCI-associated pathology could potentially pave the way for early detection and development of disease-modifying therapies, thus allowing preventive interventions instead of symptomatic treatments.
Collapse
Affiliation(s)
- Vismitha Rajeev
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Luting Poh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Sharmelee Selvaraji
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, Singapore
| | - David Y Fann
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - T Michael De Silva
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore.
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
23
|
Zhang Z, Guo Z, Tu Z, Yang H, Li C, Hu M, Zhang Y, Jin P, Hou S. Cortex-specific transcriptome profiling reveals upregulation of interferon-regulated genes after deeper cerebral hypoperfusion in mice. Front Physiol 2023; 14:1056354. [PMID: 36994418 PMCID: PMC10040763 DOI: 10.3389/fphys.2023.1056354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Background: Chronic cerebral hypoperfusion (CCH) is commonly accompanied by brain injury and glial activation. In addition to white matter lesions, the intensity of CCH greatly affects the degree of gray matter damage. However, little is understood about the underlying molecular mechanisms related to cortical lesions and glial activation following hypoperfusion. Efforts to investigate the relationship between neuropathological alternations and gene expression changes support a role for identifying novel molecular pathways by transcriptomic mechanisms.Methods: Chronic cerebral ischemic injury model was induced by the bilateral carotid artery stenosis (BCAS) using 0.16/0.18 mm microcoils. Cerebral blood flow (CBF) was evaluated using laser speckle contrast imaging (LSCI) system. Spatial learning and memory were assessed by Morris water maze test. Histological changes were evaluated by Hematoxylin staining. Microglial activation and neuronal loss were further examined by immunofluorescence staining. Cortex-specific gene expression profiling analysis was performed in sham and BCAS mice, and then validated by quantitative RT-PCR and immunohistochemistry (IHC).Results: In our study, compared with the sham group, the right hemisphere CBF of BCAS mice decreased to 69% and the cognitive function became impaired at 4 weeks postoperation. Besides, the BCAS mice displayed profound gray matter damage, including atrophy and thinning of the cortex, accompanied by neuronal loss and increased activated microglia. Gene set enrichment analysis (GSEA) revealed that hypoperfusion-induced upregulated genes were significantly enriched in the pathways of interferon (IFN)-regulated signaling along with neuroinflammation signaling. Ingenuity pathway analysis (IPA) predicted the importance of type I IFN signaling in regulating the CCH gene network. The obtained RNA-seq data were validated by qRT-PCR in cerebral cortex, showing consistency with the RNA-seq results. Also, IHC staining revealed elevated expression of IFN-inducible protein in cerebral cortex following BCAS-hypoperfusion.Conclusion: Overall, the activation of IFN-mediated signaling enhanced our understanding of the neuroimmune responses induced by CCH. The upregulation of IFN-regulated genes (IRGs) might exert a critical impact on the progression of cerebral hypoperfusion. Our improved understanding of cortex-specific transcriptional profiles will be helpful to explore potential targets for CCH.
Collapse
Affiliation(s)
- Zengyu Zhang
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Zimin Guo
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Zhilan Tu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Hualan Yang
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Chao Li
- School of Pharmacy, Hubei University of Science and Technology, Hubei, China
| | - Mengting Hu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Yuan Zhang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Pengpeng Jin
- Department of Chronic Disease Management, Shanghai Pudong Hospital, Fudan University, Shanghai, China
- *Correspondence: Shuangxing Hou, ; Pengpeng Jin,
| | - Shuangxing Hou
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
- *Correspondence: Shuangxing Hou, ; Pengpeng Jin,
| |
Collapse
|
24
|
Tang X, Wei C, Zhang R, You J, Chen X. CCL21/CCR7 axis regulates demyelination and vascular cognitive impairment in a mouse model for chronic cerebral hypoperfusion. Neurol Res 2023; 45:248-259. [PMID: 36215431 DOI: 10.1080/01616412.2022.2132456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVES White matter lesions (WML) are usually accompanied by cognitive decline, which consist of axonal loss and demyelination. CC chemokine ligand 21 (CCL21) and its receptor C-C chemokine receptor 7 (CCR7) belong to the chemokine family, which are involved in many diseases. However, their function in the central nervous system (CNS) is still unexplored. This study aimed to explore the role of CCL21/CCR7 axis in the pathological process of chronic ischemia-induced WML. METHODS Bilateral common carotid artery stenosis (BCAS) was employed in C57BL/6 mice as the in vivo WML model. Microarray analysis was performed to detect the overall molecular changes induced in the endothelial cells by BCAS. Q-PCR, Western blotting, and immunofluorescence staining were performed to evaluate expression levels of the related molecules. The mice were injected with LV-CCL21-GFP virus in the corpus callosum to overexpress CCL21. WML degree was determined via MRI, and cognitive ability was assessed by Y-maze and novel object recognition tests. Myelin sheath integrity was evaluated via immunofluorescence staining. RESULTS CCL21 was significantly downregulated in endothelial cells after BCAS and CCL21 overexpression alleviated BCAS-induced cognitive deficits and demyelination. Furthermore, CCR7 was found to be mainly expressed in oligodendrocytes (OLs) after exposed to hypoxia and CCR7 silencing blocked the protective effects induced by CCL21 overexpression. Conclusions CCL21/CCR7 axis may play a key role in demyelination induced by BCAS. This might provide a novel therapeutic target for WML.
Collapse
Affiliation(s)
- Xuelian Tang
- These authors have contributed equally to this work and share the first authorship
| | - Cunsheng Wei
- These authors have contributed equally to this work and share the first authorship
| | - Rui Zhang
- Department of Neurology, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| | - Jie You
- Department of Neurology, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| | - Xuemei Chen
- Department of Neurology, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
25
|
Sun R, Shang J, Yan X, Zhao J, Wang W, Wang W, Li W, Gao C, Wang F, Zhang H, Wang Y, Cao H, Zhang J. VCAM1 Drives Vascular Inflammation Leading to Continuous Cortical Neuronal Loss Following Chronic Cerebral Hypoperfusion. J Alzheimers Dis 2023; 91:1541-1555. [PMID: 36641679 DOI: 10.3233/jad-221059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) is associated with neuronal loss and blood-brain barrier (BBB) impairment in vascular dementia (VaD). However, the relationship and the molecular mechanisms between BBB dysfunction and neuronal loss remain elusive. OBJECTIVE We explored the reasons for neuron loss following CCH. METHODS Using permanent bilateral common carotid artery occlusion (2VO) rat model, we observed the pathological changes of cortical neurons and BBB in the sham group as well as rats 3d, 7d, 14d and 28d post 2VO. In order to further explore the factors influencing neuron loss following CCH with regard to cortical blood vessels, we extracted cortical brain microvessels at five time points for transcriptome sequencing. Finally, integrin receptor a4β1 (VLA-4) inhibitor was injected into the tail vein, and cortical neuron loss was detected again. RESULTS We found that cortical neuron loss following CCH is a continuous process, but damage to the BBB is acute and transient. Results of cortical microvessel transcriptome analysis showed that biological processes related to vascular inflammation mainly occurred in the chronic phase. Meanwhile, cell adhesion molecules, cytokine-cytokine receptor interaction were significantly changed at this phase. Among them, the adhesion molecule VCAM1 plays an important role. Using VLA-4 inhibitor to block VCAM1-VLA-4 interaction, cortical neuron damage was ameliorated at 14d post 2VO. CONCLUSION Injury of the BBB may not be the main reason for persistent loss of cortical neurons following CCH. The continuous inflammatory response within blood vessels maybe an important factor in the continuous loss of cortical neurons following CCH.
Collapse
Affiliation(s)
- Ruihua Sun
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China.,Department of Neurology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Junkui Shang
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Xi Yan
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Jingran Zhao
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China.,Department of Neurology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Wan Wang
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China.,Department of Neurology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Wenjing Wang
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Wei Li
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Chenhao Gao
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Fengyu Wang
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Haohan Zhang
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yanliang Wang
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China.,Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, Zhengzhou, Henan, China
| | - Huixia Cao
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China.,Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, Zhengzhou, Henan, China
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China.,Department of Neurology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
26
|
The impact of cerebral vasomotor reactivity on cerebrovascular diseases and cognitive impairment. J Neural Transm (Vienna) 2022; 129:1321-1330. [PMID: 36205784 PMCID: PMC9550758 DOI: 10.1007/s00702-022-02546-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022]
Abstract
The regulation of cerebral blood flow (CBF) is a complex and tightly controlled function ensuring delivery of oxygen and nutrients and removal of metabolic wastes from brain tissue. Cerebral vasoreactivity (CVR) refers to the ability of the nervous system to regulate CBF according to metabolic demands or changes in the microenvironment. This can be assessed through a variety of nuclear medicine and imaging techniques and protocols. Several studies have investigated the association of CVR with physiological and pathological conditions, with particular reference to the relationship with cognitive impairment and cerebrovascular disorders (CVD). A better understanding of the interaction between CVR and cognitive dysfunction in chronic and particularly acute CVD could help improving treatment and rehabilitation strategies in these patients. In this paper, we reviewed current knowledge on CVR alterations in the context of acute and chronic CVD and cognitive dysfunction. Alterations in CVR and hemodynamics have been described in patients with both neurodegenerative and vascular cognitive impairment, and the severity of these alterations seems to correlate with CVR derailment. Furthermore, an increased risk of cognitive impairment progression has been associated with alterations in CVR parameters and hemodynamics. Few studies have investigated these associations in acute cerebrovascular disorders and the results are inconsistent; thus, further research on this topic is encouraged.
Collapse
|
27
|
Bhat JA, Kumar M. Neuroprotective Effects of Theobromine in permanent bilateral common carotid artery occlusion rat model of cerebral hypoperfusion. Metab Brain Dis 2022; 37:1787-1801. [PMID: 35587851 DOI: 10.1007/s11011-022-00995-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 04/27/2022] [Indexed: 10/18/2022]
Abstract
Cerebral hypoperfusion (CH) is a common underlying mechanism of dementia disorders linked to aberrations in the neurovascular unit. Hemodynamic disturbances adversely affect cellular energy homeostasis that triggers a sequence of events leading to irrevocable damage to the brain and neurobehavioral discrepancies. Theobromine is a common ingredient of many natural foods consumed by a large population worldwide. Theobromine has shown health benefits in several studies, attributed to regulation of calcium homeostasis, phosphodiesterase, neurotransmission, and neurotrophins. The current study evaluated the neuroprotective potential of theobromine against CH in the permanent bilateral common carotid artery occlusion (BCCAO) prototype. Wistar rats were distributed in Sham-operated (S), S + T100, CH, CH + T50, and CH + T100 groups. Animals received permanent BCCAO or Sham treatment on day 1. Theobromine (50, 100 mg/kg) was given orally in animals subjected to BCCAO for 14 days daily. CH caused neurological deficits (12-point scale), motor dysfunction, and memory impairment in rats. Treatment with theobromine significantly attenuated neurological deficits and improved sensorimotor functions and memory in rats with CH. In biochemistry investigation of the entire brain, findings disclosed reduction in brain oxidative stress, inflammatory intermediaries (tumor necrosis factor-α, interleukin-1β and - 6, nuclear factor-κB), markers of cell demise (lactate dehydrogenase, caspase-3), acetylcholinesterase activity, and improvement in γ-aminobutyric acid quantity in rats that were given theobromine for 14 days daily after CH. Histopathological analysis substantiated attenuation of neurodegenerative changes by theobromine. The findings of this study indicated that theobromine could improve neurological scores, sensorimotor abilities, and memory in CH prototype.
Collapse
Affiliation(s)
- Javeed Ahmad Bhat
- Department of Pharmacology, Swift School of Pharmacy, Ghaggar Sarai, Rajpura, Punjab, India
| | - Manish Kumar
- Department of Pharmacology, Swift School of Pharmacy, Ghaggar Sarai, Rajpura, Punjab, India.
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| |
Collapse
|
28
|
Xu W, Bai Q, Dong Q, Guo M, Cui M. Blood–Brain Barrier Dysfunction and the Potential Mechanisms in Chronic Cerebral Hypoperfusion Induced Cognitive Impairment. Front Cell Neurosci 2022; 16:870674. [PMID: 35783093 PMCID: PMC9243657 DOI: 10.3389/fncel.2022.870674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic cerebral hypoperfusion (CCH) is a major cause of vascular cognitive impairment and dementia (VCID). Although the underlying mechanisms have not been fully elucidated, the emerging data suggest that blood–brain barrier (BBB) dysfunction is one of the pivotal pathological changes in CCH. BBB dysfunction appears early in CCH, contributing to the deterioration of white matter and the development of cognitive impairment. In this review, we summarize the latest experimental and clinical evidence implicating BBB disruption as a major cause of VCID. We discuss the mechanisms of BBB dysfunction in CCH, focusing on the cell interactions within the BBB, as well as the potential role of APOE genotype. In summary, we provide novel insights into the pathophysiological mechanisms underlying BBB dysfunction and the potential clinical benefits of therapeutic interventions targeting BBB in CCH.
Collapse
Affiliation(s)
- WenQing Xu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qingke Bai
- Department of Neurology, Pudong People’s Hospital, Shanghai, China
| | - Qiang Dong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Min Guo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Min Guo,
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- *Correspondence: Mei Cui,
| |
Collapse
|
29
|
Traub J, Otto M, Sell R, Homola GA, Steinacker P, Oeckl P, Morbach C, Frantz S, Pham M, Störk S, Stoll G, Frey A. Serum glial fibrillary acidic protein indicates memory impairment in patients with chronic heart failure. ESC Heart Fail 2022; 9:2626-2634. [PMID: 35611842 PMCID: PMC9288738 DOI: 10.1002/ehf2.13986] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/28/2022] [Accepted: 05/06/2022] [Indexed: 12/12/2022] Open
Abstract
Aims Cognitive dysfunction occurs frequently in patients with heart failure (HF), but early detection remains challenging. Serum glial fibrillary acidic protein (GFAP) is an emerging biomarker of cognitive decline in disorders of primary neurodegeneration such as Alzheimer's disease. We evaluated the utility of serum GFAP as a biomarker for cognitive dysfunction and structural brain damage in patients with stable chronic HF. Methods and results Using bead‐based single molecule immunoassays, we quantified serum levels of GFAP in patients with HF participating in the prospective Cognition.Matters‐HF study. Participants were extensively phenotyped, including cognitive testing of five separate domains and magnetic resonance imaging (MRI) of the brain. Univariable and multivariable models, also accounting for multiple testing, were run. One hundred and forty‐six chronic HF patients with a mean age of 63.8 ± 10.8 years were included (15.1% women). Serum GFAP levels (median 246 pg/mL, quartiles 165, 384 pg/mL; range 66 to 1512 pg/mL) did not differ between sexes. In the multivariable adjusted model, independent predictors of GFAP levels were age (T = 5.5; P < 0.001), smoking (T = 3.2; P = 0.002), estimated glomerular filtration rate (T = −4.7; P < 0.001), alanine aminotransferase (T = −2.1; P = 0.036), and the left atrial end‐systolic volume index (T = 3.4; P = 0.004). NT‐proBNP but not serum GFAP explained global cerebral atrophy beyond ageing. However, serum GFAP levels were associated with the cognitive domain visual/verbal memory (T = −3.0; P = 0.003) along with focal hippocampal atrophy (T = 2.3; P = 0.025). Conclusions Serum GFAP levels are affected by age, smoking, and surrogates of the severity of HF. The association of GFAP with memory dysfunction suggests that astroglial pathologies, which evade detection by conventional MRI, may contribute to memory loss beyond ageing in patients with chronic HF.
Collapse
Affiliation(s)
- Jan Traub
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University and University Hospital Würzburg, Würzburg, Germany.,Interdisciplinary Center for Clinical Research, University Würzburg, Würzburg, Germany
| | - Markus Otto
- Department of Neurology, University Hospital Ulm, Ulm, Germany.,Department of Neurology, University Hospital Halle-Wittenberg, Halle, Germany
| | - Roxane Sell
- Comprehensive Heart Failure Center, University and University Hospital Würzburg, Würzburg, Germany.,Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
| | - György A Homola
- Comprehensive Heart Failure Center, University and University Hospital Würzburg, Würzburg, Germany.,Department of Neuroradiology, University Hospital Würzburg, Würzburg, Germany
| | - Petra Steinacker
- Department of Neurology, University Hospital Ulm, Ulm, Germany.,Department of Neurology, University Hospital Halle-Wittenberg, Halle, Germany
| | - Patrick Oeckl
- Department of Neurology, University Hospital Ulm, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE e.V.), Ulm, Germany
| | - Caroline Morbach
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University and University Hospital Würzburg, Würzburg, Germany
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University and University Hospital Würzburg, Würzburg, Germany
| | - Mirko Pham
- Comprehensive Heart Failure Center, University and University Hospital Würzburg, Würzburg, Germany.,Department of Neuroradiology, University Hospital Würzburg, Würzburg, Germany
| | - Stefan Störk
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University and University Hospital Würzburg, Würzburg, Germany
| | - Guido Stoll
- Comprehensive Heart Failure Center, University and University Hospital Würzburg, Würzburg, Germany.,Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Anna Frey
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University and University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
30
|
Ren Q, An P, Jin K, Xia X, Huang Z, Xu J, Huang C, Jiang Q, Meng X. A Pilot Study of Radiomic Based on Routine CT Reflecting Difference of Cerebral Hemispheric Perfusion. Front Neurosci 2022; 16:851720. [PMID: 35431785 PMCID: PMC9009332 DOI: 10.3389/fnins.2022.851720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/03/2022] [Indexed: 11/30/2022] Open
Abstract
Background To explore the effectiveness of radiomics features based on routine CT to reflect the difference of cerebral hemispheric perfusion. Methods We retrospectively recruited 52 patients with severe stenosis or occlusion in the unilateral middle cerebral artery (MCA), and brain CT perfusion showed an MCA area with deficit perfusion. Radiomics features were extracted from the stenosis side and contralateral of the MCA area based on precontrast CT. Two different region of interest drawing methods were applied. Then the patients were randomly grouped into training and testing sets by the ratio of 8:2. In the training set, ANOVA and the Elastic Net Regression with fivefold cross-validation were conducted to filter and choose the optimized features. Moreover, different machine learning models were built. In the testing set, the area under the receiver operating characteristic (AUC) curve, calibration, and clinical utility were applied to evaluate the predictive performance of the models. Results The logistic regression (LR) for the triangle-contour method and artificial neural network (ANN) for the semiautomatic-contour method were chosen as radiomics models for their good prediction efficacy in the training phase (AUC = 0.869, 0.873) and the validation phase (AUC = 0.793, 0.799). The radiomics algorithms of the triangle-contour and semiautomatic-contour method were implemented in the whole training set (AUC = 0.870, 0.867) and were evaluated in the testing set (AUC = 0.760, 0.802). According to the optimal cutoff value, these two methods can classify the vascular stenosis side class and normal side class. Conclusion Radiomic predictive feature based on precontrast CT image could reflect the difference of cerebral hemispheric perfusion to some extent.
Collapse
Affiliation(s)
- Qingguo Ren
- Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Panpan An
- Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Ke Jin
- Deepwise AI Lab, Beijing Deepwise and League of PHD Technology Co., Ltd., Beijing, China
| | - Xiaona Xia
- Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Zhaodi Huang
- Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Jingxu Xu
- Deepwise AI Lab, Beijing Deepwise and League of PHD Technology Co., Ltd., Beijing, China
| | - Chencui Huang
- Deepwise AI Lab, Beijing Deepwise and League of PHD Technology Co., Ltd., Beijing, China
| | - Qingjun Jiang
- Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Xiangshui Meng
- Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Qingdao, China
- *Correspondence: Xiangshui Meng,
| |
Collapse
|
31
|
Poh L, Sim WL, Jo DG, Dinh QN, Drummond GR, Sobey CG, Chen CLH, Lai MKP, Fann DY, Arumugam TV. The role of inflammasomes in vascular cognitive impairment. Mol Neurodegener 2022; 17:4. [PMID: 35000611 PMCID: PMC8744307 DOI: 10.1186/s13024-021-00506-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 12/02/2021] [Indexed: 12/11/2022] Open
Abstract
There is an increasing prevalence of Vascular Cognitive Impairment (VCI) worldwide, and several studies have suggested that Chronic Cerebral Hypoperfusion (CCH) plays a critical role in disease onset and progression. However, there is a limited understanding of the underlying pathophysiology of VCI, especially in relation to CCH. Neuroinflammation is a significant contributor in the progression of VCI as increased systemic levels of the proinflammatory cytokine interleukin-1β (IL-1β) has been extensively reported in VCI patients. Recently it has been established that CCH can activate the inflammasome signaling pathways, involving NLRP3 and AIM2 inflammasomes that critically regulate IL-1β production. Given that neuroinflammation is an early event in VCI, it is important that we understand its molecular and cellular mechanisms to enable development of disease-modifying treatments to reduce the structural brain damage and cognitive deficits that are observed clinically in the elderly. Hence, this review aims to provide a comprehensive insight into the molecular and cellular mechanisms involved in the pathogenesis of CCH-induced inflammasome signaling in VCI.
Collapse
Affiliation(s)
- Luting Poh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Liang Sim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Quynh Nhu Dinh
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Grant R. Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Christopher G. Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Christopher Li-Hsian Chen
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K. P. Lai
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Y. Fann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Healthy Longevity, National University Health System (NUHS), Singapore, Singapore
| | - Thiruma V. Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| |
Collapse
|
32
|
Vernon SD, Funk S, Bateman L, Stoddard GJ, Hammer S, Sullivan K, Bell J, Abbaszadeh S, Lipkin WI, Komaroff AL. Orthostatic Challenge Causes Distinctive Symptomatic, Hemodynamic and Cognitive Responses in Long COVID and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Front Med (Lausanne) 2022; 9:917019. [PMID: 35847821 PMCID: PMC9285104 DOI: 10.3389/fmed.2022.917019] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 06/01/2022] [Indexed: 12/02/2022] Open
Abstract
Background Some patients with acute COVID-19 are left with persistent, debilitating fatigue, cognitive impairment ("brain fog"), orthostatic intolerance (OI) and other symptoms ("Long COVID"). Many of the symptoms are like those of other post-infectious fatigue syndromes and may meet criteria for myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Common diagnostic laboratory tests are often unrevealing. Methods We evaluated whether a simple, standardized, office-based test of OI, the 10-min NASA Lean Test (NLT), would aggravate symptoms and produce objective hemodynamic and cognitive abnormalities, the latter being evaluated by a simple smart phone-based app. Participants People with Long COVID (N = 42), ME/CFS (N = 26) and healthy control subjects (N = 20) were studied just before, during, immediately after, 2 and 7 days following completion of the NLT. Results The NLT provoked a worsening of symptoms in the two patient groups but not in healthy control subjects, and the severity of all symptoms was similar and significantly worse in the two patient groups than in the control subjects (p < 0.001). In the two patient groups, particularly those with Long COVID, the NLT provoked a marked and progressive narrowing in the pulse pressure. All three cognitive measures of reaction time worsened in the two patient groups immediately following the NLT, compared to the healthy control subjects, particularly in the Procedural Reaction Time (p < 0.01). Conclusions A test of orthostatic stress easily performed in an office setting reveals different symptomatic, hemodynamic and cognitive abnormalities in people with Long COVID and ME/CFS, compared to healthy control subjects. Thus, an orthostatic challenge easily performed in an office setting, and the use of a smart phone app to assess cognition, can provide objective confirmation of the orthostatic intolerance and brain fog reported by patients with Long COVID and ME/CFS.
Collapse
Affiliation(s)
- Suzanne D. Vernon
- The Bateman Horne Center of Excellence, Salt Lake City, UT, United States
| | - Sherlyn Funk
- Department of Family & Preventive Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Lucinda Bateman
- The Bateman Horne Center of Excellence, Salt Lake City, UT, United States
| | - Gregory J. Stoddard
- Department of Family & Preventive Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Sarah Hammer
- The Bateman Horne Center of Excellence, Salt Lake City, UT, United States
| | - Karen Sullivan
- The Bateman Horne Center of Excellence, Salt Lake City, UT, United States
| | - Jennifer Bell
- The Bateman Horne Center of Excellence, Salt Lake City, UT, United States
| | - Saeed Abbaszadeh
- The Bateman Horne Center of Excellence, Salt Lake City, UT, United States
| | - W. Ian Lipkin
- Center for Solutions for ME/CFS, Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Anthony L. Komaroff
- Division of General Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- *Correspondence: Anthony L. Komaroff
| |
Collapse
|
33
|
Li J, Xiang H, Huang C, Lu J. Pharmacological Actions of Myricetin in the Nervous System: A Comprehensive Review of Preclinical Studies in Animals and Cell Models. Front Pharmacol 2021; 12:797298. [PMID: 34975495 PMCID: PMC8716845 DOI: 10.3389/fphar.2021.797298] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/29/2021] [Indexed: 12/01/2022] Open
Abstract
Myricetin is a natural flavonoid extracted from a variety of plants, such as medicinal herbs, vegetables, berries, and tea leaves. A growing body of evidence has reported that myricetin supplementation display therapeutic activities in a lot of nervous system disorders, such as cerebral ischemia, Alzheimer’s disease, Parkinson’s disease, epilepsy, and glioblastoma. Myricetin supplementation can also protect against pathological changes and behavioral impairment induced by multiple sclerosis and chronic stress. On the basis of these pharmacological actions, myricetin could be developed as a potential drug for the prevention and/or treatment of nervous system disorders. Mechanistic studies have shown that inhibition of oxidative stress, cellular apoptosis, and neuroinflammatory response are common mechanisms for the neuroprotective actions of myricetin. Other mechanisms, including the activation of the nuclear factor E2-related factor 2 (Nrf2), extracellular signal-regulated kinase 1/2 (ERK1/2), protein kinase B (Akt), cyclic adenosine monophosphate-response element binding protein (CREB), and brain-derived neurotrophic factor (BDNF) signaling, inhibition of intracellular Ca2+ increase, inhibition of c-Jun N-terminal kinase (JNK)-p38 activation, and suppression of mutant protein aggregation, may also mediate the neuroprotective effects of myricetin. Furthermore, myricetin treatment has been shown to promote the activation of the inhibitory neurons in the hypothalamic paraventricular nucleus, which subsequently produces anti-epilepsy effects. In this review, we make a comprehensive understanding about the pharmacological effects of myricetin in the nervous system, aiming to push the development of myricetin as a novel drug for the treatment of nervous system disorders.
Collapse
Affiliation(s)
- Jie Li
- Department of Gastroenterology, The People’s Hospital of Taizhou, The Fifth Affiliated Hospital of Nantong University, Taizhou, China
| | - Haitao Xiang
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Jiashu Lu
- Department of Pharmacy, The People’s Hospital of Taizhou, The Fifth Affiliated Hospital of Nantong University, Taizho, China
- *Correspondence: Jiashu Lu,
| |
Collapse
|
34
|
Neuroprotective Potential of Carnosine in Cerebrovascular Diseases. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10342-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
35
|
Yan F, Tian Y, Huang Y, Wang Q, Liu P, Wang N, Zhao F, Zhong L, Hui W, Luo Y. Xi-Xian-Tong-Shuan capsule alleviates vascular cognitive impairment in chronic cerebral hypoperfusion rats by promoting white matter repair, reducing neuronal loss, and inhibiting the expression of pro-inflammatory factors. Biomed Pharmacother 2021; 145:112453. [PMID: 34808554 DOI: 10.1016/j.biopha.2021.112453] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/06/2021] [Accepted: 11/16/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND While the number of cases of vascular cognitive impairment caused by chronic cerebral hypoperfusion (CCH) has been increasing every year, there are currently no clinically effective treatment methods. At present, Xi-Xian-Tong-Shuan capsule is predominantly used in patients with acute cerebral ischemia; however, its protective effect on CCH has rarely been reported. OBJECTIVE To explore the underlying mechanisms by which Xi-Xian-Tong-Shuan capsule alleviates cognitive impairment caused by CCH. METHODS A model of CCH was established in specific-pathogen-free (SPF)-grade male Sprague-Dawley (SD) rats using bilateral common carotid artery occlusion (BCCAO). Xi-Xian-Tong-Shuan capsules were intragastrically administered for 42 days after the BCCAO surgery. We then assessed for changes in cognitive function, expression levels of pro-inflammatory factors, and coagulation function as well as for the presence of white matter lesions and neuronal loss. One-way ANOVA and Tukey's test were used to analyze the experimental data. RESULTS The rats showed significant cognitive dysfunction after the BCCAO surgery along with white matter lesions, a loss of neurons, and elevated levels of inflammatory factors, all of which were significantly reversed after intervention with Xi-Xian-Tong-Shuan capsules. CONCLUSION Xi-Xian-Tong-Shuan capsules can ameliorate vascular cognitive impairment in CCH rats by preventing damage of white matter, reducing neuronal loss, and inhibiting the expression of pro-inflammatory factors. Our study provides a new reference for the clinical treatment of chronic cerebral ischemia with Xi-Xian-Tong-Shuan capsules.
Collapse
Affiliation(s)
- Feng Yan
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yue Tian
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yuyou Huang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ping Liu
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ningqun Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Fangfang Zhao
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Liyuan Zhong
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wuhan Hui
- Department of Hematology, Xuanwu Hospital of Capital Medical University, Beijing, China.
| | - Yumin Luo
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
36
|
Delirium and Clusters of Older Patients Affected by Multimorbidity in Acute Hospitals. J Am Med Dir Assoc 2021; 23:885-888. [PMID: 34798007 DOI: 10.1016/j.jamda.2021.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/28/2021] [Accepted: 10/09/2021] [Indexed: 01/18/2023]
Abstract
OBJECTIVES Delirium is commonly seen in older adults with multimorbidity, during a hospitalization, resulting from the interplay between predisposing factors such as advanced age, frailty, and dementia, and a series of precipitating factors. The association between delirium and specific multimorbidity is largely unexplored so far although of potential key relevance for targeted interventions. The aim of the study was to check for a potential association of multimorbidity with delirium in a large cohort of older patients hospitalized for an acute medical or surgical condition. DESIGN This is a cross-sectional study nested in the 2017 Delirium Day project. SETTING AND PARTICIPANTS The study includes 1829 hospitalized patients (age: 81.8, SD: 5.5). Of them, 419 (22.9%) had delirium. METHODS Sociodemographic and medical history were collected. The 4AT was used to assess the presence of delirium. The Charlson Comorbidity index was used to assess multimorbidity. RESULTS The results identified neurosensorial multimorbidity as the most prevalent, including patients with dementia, cerebrovascular diseases, and sensory impairments. In light of the highest co-occurrence of 3 neurosensorial chronic conditions, we could hypothesize that a baseline altered brain functional and neural connectivity might determine the vulnerability signature for incipient overall system disruption in presence of acute insults. CONCLUSIONS AND IMPLICATIONS Eventually, our findings moved a step forward in supporting the key importance of routine screening for sensory impairments and cognitive status of older patients for the highest risk of in-hospital delirium. In fact, preventive interventions could be particularly relevant and effective in preventing delirium in such vulnerable populations and might help refining this early diagnosis.
Collapse
|
37
|
Little PV, Arnberg F, Jussing E, Lu L, Ingemann Jensen A, Mitsios N, Mulder J, Tran TA, Holmin S. The cellular basis of increased PET hypoxia tracer uptake in focal cerebral ischemia with comparison between [ 18F]FMISO and [ 64Cu]CuATSM. J Cereb Blood Flow Metab 2021; 41:617-629. [PMID: 32423333 PMCID: PMC7922752 DOI: 10.1177/0271678x20923857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/31/2020] [Accepted: 04/12/2020] [Indexed: 11/16/2022]
Abstract
PET hypoxia imaging can assess tissue viability in acute ischemic stroke (AIS). [18F]FMISO is an established tracer but requires substantial accumulation time, limiting its use in hyperacute AIS. [64Cu]CuATSM requires less accumulation time and has shown promise as a hypoxia tracer. We compared these tracers in a M2-occlusion model (M2CAO) with preserved collateral blood flow. Rats underwent M2CAO and [18F]FMISO (n = 12) or [64Cu]CuATSM (n = 6) examinations. [64Cu]CuATSM animals were also examined with MRI. Pimonidazole was used as a surrogate for [18F]FMISO in an immunofluorescence analysis employed to profile levels of hypoxia in neurons (NeuN) and astrocytes (GFAP). There was increased [18F]FMISO uptake in the M2CAO cortex. No increase in [64Cu]CuATSM activity was found. The pimonidazole intensity of neurons and astrocytes was increased in hypoxic regions. The pimonidazole intensity ratio was higher in neurons than in astrocytes. In the majority of animals, immunofluorescence revealed a loss of astrocytes within the core of regions with increased pimonidazole uptake. We conclude that [18F]FMISO is superior to [64Cu]CuATSM in detecting hypoxia in AIS, consistent with an earlier study. [18F]FMISO may provide efficient diagnostic imaging beyond the hyperacute phase. Results do not provide encouragement for the use of [64Cu]CuATSM in experimental AIS.
Collapse
Affiliation(s)
- Philip V Little
- The Department of Clinical Neuroscience, Karolinska Institutet,
Stockholm Sweden
- The Department of Neuroradiology, BioClinicum, Karolinska
University Hospital, Stockholm, Sweden
| | - Fabian Arnberg
- The Department of Clinical Neuroscience, Karolinska Institutet,
Stockholm Sweden
- The Department of Neuroradiology, BioClinicum, Karolinska
University Hospital, Stockholm, Sweden
| | - Emma Jussing
- The Department of Clinical Neuroscience, Karolinska Institutet,
Stockholm Sweden
- The Department of Radiopharmacy, Karolinska University Hospital,
Stockholm, Sweden
- The Department of Oncology and Pathology, Karolinska Institutet,
Stockholm Sweden
| | - Li Lu
- The Department of Clinical Neuroscience, Karolinska Institutet,
Stockholm Sweden
- The Department of Radiopharmacy, Karolinska University Hospital,
Stockholm, Sweden
- The Department of Oncology and Pathology, Karolinska Institutet,
Stockholm Sweden
| | | | - Nicholas Mitsios
- The Department of Neuroscience, Karolinska Institutet,
Stockholm, Sweden
| | - Jan Mulder
- The Department of Neuroscience, Karolinska Institutet,
Stockholm, Sweden
| | - Thuy A Tran
- The Department of Clinical Neuroscience, Karolinska Institutet,
Stockholm Sweden
- The Department of Radiopharmacy, Karolinska University Hospital,
Stockholm, Sweden
- The Department of Oncology and Pathology, Karolinska Institutet,
Stockholm Sweden
| | - Staffan Holmin
- The Department of Clinical Neuroscience, Karolinska Institutet,
Stockholm Sweden
- The Department of Neuroradiology, BioClinicum, Karolinska
University Hospital, Stockholm, Sweden
| |
Collapse
|
38
|
Ek Olofsson H, Haglund M, Englund E. Are cortical microvascular raspberries caused by cerebral hypoperfusion? An exploratory pathological study. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100026. [PMID: 36324730 PMCID: PMC9616238 DOI: 10.1016/j.cccb.2021.100026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/03/2021] [Accepted: 08/15/2021] [Indexed: 12/03/2022]
Abstract
An exploratory study on the ‘raspberry’, a type of cortical microvascular formation. Hypothesis: raspberries form by angiogenesis induced by cerebral hypoperfusion. Are raspberries associated with clinical or pathological markers of hypoperfusion? Data: histopathological raspberry quantification, medical records, autopsy reports. Raspberries were associated with atherosclerosis of the basal cerebral arteries.
Introduction This retrospective study investigated a cortical microvascular formation, termed a ‘raspberry’ due to its appearance under a bright-field microscope. We examined whether there is support for the hypothesis that raspberry formation is an angiogenic process induced by cerebral hypoperfusion. Materials and Methods Raspberries were manually quantified in haematoxylin and eosin-stained cortical sections from the anterior frontal lobe of deceased individuals who had undergone a diagnostic neuropathological examination at the Department of Pathology, Lund, Sweden, during April 2019–January 2021. Subjects represented consecutively received cases during this 22-month period. The raspberry density was compared between subjects according to variables collected from medical records and autopsy reports: age, sex, hypertension, diabetes mellitus, atrial fibrillation, orthostatic hypotension, chronic heart failure, acute circulatory failure, aortic atherosclerosis, atherosclerosis of the basal cerebral arteries (referred to as ‘cerebral atherosclerosis’), cerebral small vessel disease, cerebral amyloid angiopathy, cerebral infarction, and ischaemic white matter disease. Results 62 subjects were included. The mean age was 71.9 years (range 46–97 years). 21 subjects (33.9%) were female. Independent-samples t-test showed a higher raspberry density in subjects with cerebral atherosclerosis (p = 0.029; 95% CI 0.7, 11.6 raspberries/cm²). The higher raspberry density in subjects with cerebral atherosclerosis remained in multiple linear regression (p = 0.003; 95% CI 2.3, 11.1 raspberries/cm²). Conclusion This exploratory study indicates that cortical raspberries could be associated with cerebral atherosclerosis. The remaining results were inconclusive but motivate further examination of variables such as acute circulatory failure.
Collapse
|
39
|
Lana D, Ugolini F, Giovannini MG. An Overview on the Differential Interplay Among Neurons-Astrocytes-Microglia in CA1 and CA3 Hippocampus in Hypoxia/Ischemia. Front Cell Neurosci 2020; 14:585833. [PMID: 33262692 PMCID: PMC7686560 DOI: 10.3389/fncel.2020.585833] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022] Open
Abstract
Neurons have been long regarded as the basic functional cells of the brain, whereas astrocytes and microglia have been regarded only as elements of support. However, proper intercommunication among neurons-astrocytes-microglia is of fundamental importance for the functional organization of the brain. Perturbation in the regulation of brain energy metabolism not only in neurons but also in astrocytes and microglia may be one of the pathophysiological mechanisms of neurodegeneration, especially in hypoxia/ischemia. Glial activation has long been considered detrimental for survival of neurons, but recently it appears that glial responses to an insult are not equal but vary in different brain areas. In this review, we first take into consideration the modifications of the vascular unit of the glymphatic system and glial metabolism in hypoxic conditions. Using the method of triple-labeling fluorescent immunohistochemistry coupled with confocal microscopy (TIC), we recently studied the interplay among neurons, astrocytes, and microglia in chronic brain hypoperfusion. We evaluated the quantitative and morpho-functional alterations of the neuron-astrocyte-microglia triads comparing the hippocampal CA1 area, more vulnerable to ischemia, to the CA3 area, less vulnerable. In these contiguous and interconnected areas, in the same experimental hypoxic conditions, astrocytes and microglia show differential, finely regulated, region-specific reactivities. In both areas, astrocytes and microglia form triad clusters with apoptotic, degenerating neurons. In the neuron-astrocyte-microglia triads, the cell body of a damaged neuron is infiltrated and bisected by branches of astrocyte that create a microscar around it while a microglial cell phagocytoses the damaged neuron. These coordinated actions are consistent with the scavenging and protective activities of microglia. In hypoxia, the neuron-astrocyte-microglia triads are more numerous in CA3 than in CA1, further indicating their protective effects. These data, taken from contiguous and interconnected hippocampal areas, demonstrate that glial response to the same hypoxic insult is not equal but varies significantly. Understanding the differences of glial reactivity is of great interest to explain the differential susceptibility of hippocampal areas to hypoxia/ischemia. Further studies may evidence the differential reactivity of glia in different brain areas, explaining the higher or lower sensitivity of these areas to different insults and whether glia may represent a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Section of Anatomopathology, University of Florence, Florence, Italy
| | - Maria G Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
40
|
Porcu M, Cocco L, Saloner D, Suri JS, Montisci R, Carriero A, Saba L. Extracranial Carotid Artery Stenosis: The Effects on Brain and Cognition with a Focus on Resting‐State Functional Connectivity. J Neuroimaging 2020; 30:736-745. [DOI: 10.1111/jon.12777] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/27/2022] Open
Affiliation(s)
- Michele Porcu
- Department of Radiology, AOU of Cagliari University of Cagliari Cagliari Italy
| | - Luigi Cocco
- Department of Neurology University of Genova Genova Italy
| | - David Saloner
- Department of Radiology and Biomedical Imaging University of California San Francisco CA
| | - Jasjit S. Suri
- Diagnostic and Monitoring Division AtheroPoint™ Roseville CA
| | - Roberto Montisci
- Department of Vascular Surgery AOU of Cagliari University of Cagliari Cagliari Italy
| | - Alessandro Carriero
- Radiology Department, University of Eastern Piedmont “Maggiore della Carità” Hospital Novara Italy
| | - Luca Saba
- Department of Radiology, AOU of Cagliari University of Cagliari Cagliari Italy
| |
Collapse
|
41
|
Heart Failure and Fitness to Drive. J Card Fail 2020; 26:564-565. [DOI: 10.1016/j.cardfail.2020.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 11/19/2022]
|
42
|
Moretti R, Caruso P. Small Vessel Disease-Related Dementia: An Invalid Neurovascular Coupling? Int J Mol Sci 2020; 21:1095. [PMID: 32046035 PMCID: PMC7036993 DOI: 10.3390/ijms21031095] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 12/18/2022] Open
Abstract
The arteriosclerosis-dependent alteration of brain perfusion is one of the major determinants in small vessel disease, since small vessels have a pivotal role in the brain's autoregulation. Nevertheless, as far as we know, endothelium distress can potentiate the flow dysregulation and lead to subcortical vascular dementia that is related to small vessel disease (SVD), also being defined as subcortical vascular dementia (sVAD), as well as microglia activation, chronic hypoxia and hypoperfusion, vessel-tone dysregulation, altered astrocytes, and pericytes functioning blood-brain barrier disruption. The molecular basis of this pathology remains controversial. The apparent consequence (or a first event, too) is the macroscopic alteration of the neurovascular coupling. Here, we examined the possible mechanisms that lead a healthy aging process towards subcortical dementia. We remarked that SVD and white matter abnormalities related to age could be accelerated and potentiated by different vascular risk factors. Vascular function changes can be heavily influenced by genetic and epigenetic factors, which are, to the best of our knowledge, mostly unknown. Metabolic demands, active neurovascular coupling, correct glymphatic process, and adequate oxidative and inflammatory responses could be bulwarks in defense of the correct aging process; their impairments lead to a potentially catastrophic and non-reversible condition.
Collapse
Affiliation(s)
- Rita Moretti
- Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy;
| | | |
Collapse
|